U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Cytotechnology
  • v.65(4); 2013 Aug

Logo of cytotech

Cancer stem cell hypothesis: a brief summary and two proposals

Shuhua zheng.

Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006 People’s Republic of China

University of Miami Miller School of Medicine, Miami, FL 33136 USA

Longzuo Xin

School of Agriculture and Forest Science, Hebei North University, Zhangjiakou, 075000 People’s Republic of China

Aihua Liang

The investigation and development of the cancer stem cell (CSC) model has received much focus during these years. CSC is characterized as a small fraction of cancer cells that have an indefinite ability for self-renewal and pluripotency and are responsible for initiating and sustaining of the bulk of cancer. So, whether current treatment strategies, most of which target the rapid division of cancer cells, could interfere with the slow-cycling CSCs is broadly questioned. Meanwhile, however, the new understanding of tumorigenesis has led to the development of new drug screening strategies. Both stem cells and mesenchymal stem cells have been vigorously used in pre-clinical studies of their anti-tumor potential, mainly due to their inherent tumoritropic migratory properties and their ability to carry anti-tumor transgenes. Here, based on the tumorigenic and tumoritropic characteristics of CSCs, we proposed two hypotheses exploring possible usage of CSCs as novel anti-tumor agents and potential sources for tissue regeneration. Further experimental validation of these hypotheses may unravel some new research topics.

Evil enters like a needle and spreads like an oak tree. - by Proverb, Ethiopian

Introduction

Hierarchical organization of human acute myeloid leukemia (AML) was proposed first by John Dick and colleagues in 1994 and since then similar models were identified for many solid tumors (Lapidot et al. 1994 ; Visvader and Lindeman 2008 ). Central to the heterogeneity of human malignancies is the existence of cancer stem cells (CSCs), a rare fraction of slow-cycling cancer cells that have the ability of self-renewal, pluripotency and chemo-resistance. In the presumptive CSC model, only the CSCs are tumorigenic and can produce all of the cells necessary to repopulate a tumor, whereas the bulk of the tumor is comprised of cells that are differentiated and have lost tumorigenic potential. The conceptual similarity between normal tissue-specific stem cells and CSCs implicates that a tumor is an abnormal organ composed of multiple cell types and maintained by pluripotent stem cells (Shackleton 2010 ; Reya et al. 2001 ). The existence of CSCs has substantial therapeutic implications, since it well explains why conventional chemo- and radio-therapy, most of which target the rapid division of cancer cells, cannot eradicate tumors and are frequently accompanied by lethal recurrence as shown in Fig.  1 a (Jordan et al. 2006 ). So, there is a recent surge in developing ways to eliminate the CSCs. It was reported recently that dysfunctional proliferation and differentiation of CSCs required the dysregulation of evolutionally conserved mechanisms that were required for the self-renewal and pluripotency of embryonic stem cells (Takebe et al. 2011 ). This fact has significant therapeutic implications, since a targeted therapy against CSCs may also be hostile to normal tissue stem cells and interfere with homeostatic maintenance of organs and tissues (Bonnett and Dick 1997 ). Evil enters like a needle and spreads like an oak tree. This proverb well depicts the tumorigenic process as implicated by the CSC model.

An external file that holds a picture, illustration, etc.
Object name is 10616_2012_9517_Fig1_HTML.jpg

Schematic representation of possible ways to cure cancer. a The CSC model pointed out that tumors are heterogeneous and are regarded as perturbed versions of the normal tissue, containing a small fraction of drug-resistant CSCs that are responsible for high frequency of relapse after chemotherapy. b Stem cells and in particular MSCs have been experimentally used and clinically explored for efficient delivery of therapeutic transgenes. However, conflicting reports showed that in some cases these cells could promote the metastasis and growth of a tumor. The therapeutic uncertainty of this treatment strategy stalls its clinical application. Our hypothesis is illustrated in c . In this treatment strategy, the stem-like cells transplanted resemble the ‘cancer cells’ of the abnormal tissue–the tumor. In the presence of the drug one, which acts as an environmental pressure favoring the self-renewal of the transplanted cells over that of CSCs, transplanted cells gradually take over the CSC niche. Then, the drug two, which could induce the expression of the incorporated suicide genes in the stem-like cells, kills the progenies of those seeded cells. Since in our model CSCs are replaced by other stem-like cells rather than killed by environmentally hostile agents to kill cancer cells, this strategy may circumvent the shortcomings of treatment methods as illustrated in a and b

In the CSC model, the bulk of a tumor could be regarded as an abnormal organ that expanded constantly. Then, could we also destroy the abnormal ‘organ’ with a spreading ‘evil needle’ as CSCs do to normal organs? Recently, the strategy of stem cell based drug delivery vehicles was proven more efficacious and with lower cytotoxicity, compared with traditional systematic delivery of drug or gene therapy. Theoretically, stem cells migrate towards and engraft into the tumor sites that provide the micro-environment amenable for them to survive. However, controversies and concerns remain regarding the applicability of this treatment method. In this mini-review, we discussed the basic scenario of the CSC model and its implication for future stem cell based cancer treatment. Based on the initial understanding of CSCs’ character, we hypothesize/propose two novel yet promising strategies that might shed some light on future anti-tumor research.

The evolving concept of the cancer stem cell model

Despite the clonal origin of many cancers, primary tumors are heterogeneous comprised by multiple distinct cell types that arise from a sub-population of pluripotent cancer cells named CSCs or tumor-initiating cells. However, given some intrinsic unresolved issues of the CSC hypothesis, the validity of this model remains a topic of considerable controversy (Rosen and Jordan 2009 ). For one thing, the precise identification or definition of CSCs is not clear. According to a consensus definition, CSCs can only be defined experimentally by their ability to recapitulate the generation of a continuously growing tumor (Clarke et al. 2006 ). The concomitant problem of this definition is that the empirically tumorigenic potential of serial transplantation in animal models is also affected by factors such as the tissue site of xenotransplantation and the immune status of the tumor host mice (Cho and Clarke 2008 ). So, it is possible that a subpopulation of cancer cells is tumorigenic but cannot form xenografts due to inappropriate micro-environment, rendering the frequency of CSCs in tumor tissue highly variable depending on the specific protocols used and procedures undertaken. Of note, the proportion of CSCs in the tumor mass varied remarkably among patients and CSC-rich was believed to be associated with aggressive tumor progression and poor prognosis (Ailles and Weissman 2007 ; Quintana et al. 2008 ). A more operational definition of CSC and a more standardized way to identify them are pivotal for future tumor diagnosis and treatment. For this, CSC-specific surface biomarkers that allow antibody-directed cell sorting constitute a promising candidate. However, at present, CSC biomarkers for most cancer cell lines remain largely elusive and no universal biomarker for CSC has yet been identified (Visvader and Lindeman 2008 ). It was reported that AML cells with CD34+CD38− surface antigens were highly tumorigenic when transplanted into nonobese diabetic severe combined immunodeficiency disease (NOD/SCID) mice. In a breast cancer model, however, cells with CD44+CD24-lineage negative phenotype were believed as CSCs (Bonnett and Dick 1997 ; Al-Hajj et al. 2003 ). More recently, CD44+ and CD133+ were identified on the surface of gastric and lung CSCs, respectively (Eramo et al. 2008 ; Takaishi et al. 2009 ). It is difficult to find a universal CSC marker for tumors derived from different tissues or even from the same tissue, which implicates that different lineages of CSCs may have host-specific tumorigenic mechanisms. The variability in CSC properties introduces problems when developing new anti-tumor drugs (Hwang-Verslues et al. 2009 ). Another major issue of the CSC model is the origin of these stem-like cells. So, it was proposed that CSCs might have originated from normal stem cells, early stem cell progenitors or normal differentiated cells in a unidirectional manner (Bjerkvig et al. 2005 ). However, an unexpected new origin of CSCs was identified recently by Chaffer et al. ( 2011 ). They found that transformed human CD44 lo cancerous mammary epithelial cells could spontaneously dedifferentiate into CD44 hi -CSCs in vivo, indicating the existence of bidirectional inter-conversions between CSC and non-CSC cancer cells. This seminal finding implicates that tumors may regenerate CSCs under the treatment that exclusively target CSCs, compromising the therapeutic significance of CSC specific agents that have been screened (Gupta et al. 2009 ). The CSC model for tumor progression is illustrated in Fig.  2 . Mainly due to the issues outlined above, criticisms concerning the proper conceptualization and nomination of CSCs exist (Maenhaut et al. 2010 ). However, no one would doubt that the concept of CSCs is a giant leap towards a mechanistic understanding of human malignancies and may lead to a new era of anti-tumor therapy.

An external file that holds a picture, illustration, etc.
Object name is 10616_2012_9517_Fig2_HTML.jpg

Possible origins of cancer stem cells. The cancer stem cell might be derived from normal stem cells, early stem cell progenitors or differentiated cells (Bjerkvig et al. 2005 ). Most recent studies showed differentiated cancer cells could dedifferentiate spontaneously into cancer stem cells (Chaffer et al. 2011 )

Homeostatic tissue maintenance and tissue regeneration after injury depend on the delicate balance between the two cardinal characteristics of stem cells that are to differentiate, or to produce specialized cells that comprise the tissue, and to self-renew, or to make more stem cells. The existence of a physiological micro-environment or a niche that could maintain stem cell stemness and sequester them away from differentiation and apoptotic stimuli while protecting the host from excessive stem-cell proliferation was envisaged. Although the existence of stem cell niches has been identified in multiple mammalian tissues, we have little affirmative information about how signaling pathways/bio-factors mediate the interactions between stem cells and the niche (Voog and Jones 2010 ). However, given that the CSC is intimately associated with its micro-environment, targeting this niche may become a desirable therapeutic target. Anti-angiogenic drugs like thalidomide, bevacizumab and the protease inhibitor bortezomib were proposed to exert their anti-tumor effect by modulating the micro-environment (Burness and Sipkins 2010 ).

We hypothesize/propose here two potential therapeutic and/or experimental methodologies that might hold some therapeutic significance.

Proposal 1. Cure cancer by replacing CSCs with implanted stem-like cells?

At present, tissue derived stem cells have been largely used in pre-clinical studies of their anti-tumor potential, since these stem cells or mesenchymal stem cells (MSC) can act as vehicles for targeted delivery and releasing of anti-tumor factors, such as anti-angiogenic, cytotoxic and immunostimulatory factors (Shah 2012 ). However, the major hurdle that thwarts the clinical application of this strategy is the inherent uncertainty after the injection of these stem cells or MSCs. For example, the hostile anti-tumor compounds might also unpredictably harm normal organs, and interleukin-6 (IL-6) and chemokine (C–C motif) ligand 5 (CCL5) endogenously expressed by MSCs were reported to promote the proliferation and metastasis of breast cancer cells (Karnoub et al. 2007 ; Albarenque et al. 2011 ). Also, MCSs were reported to promote formation of colorectal tumors (Tsai et al. 2011 ). Moreover, given the fact that non-CSCs can spontaneously dedifferentiate into CSCs, it is even more difficult to eradicate CSCs by killing them (Gupta et al. 2009 ). The bidirectional inter-conversions between CSCs and non-CSCs may enable tumors to maintain a relatively constant number of CSCs under treatment (Fig.  2 ).

Potentially, stem cells derived from the patients’ own tissue would not provoke the immune system. However, most recent pre-clinical studies showed contradictory results. Zhao et al. ( 2011 ) found that induced pluripotent stem (iPS) cells could trigger severe immune reactions after injection into the recipient from whom the iPSCs were derived. This finding seriously limits cell types that could be engineered as vehicles for transgenes and we categorized possible limitations of this strategy in Fig.  1 b.

Considering all these limitations that seriously limit the applicability of the strategy of using stem cells as drug delivery vehicles, we proposed an alternative strategy as illustrated in Fig.  1 c. In this treatment strategy, the replacement of CSCs by the seeded stem-like cells is induced by the presence of environmental stress i.e. ‘drug one’. Technically, it is accomplishable to favor the growth of one cell line while killing other cell lines. For example, we can incorporate the tet R gene into the genome of the seeding cells and at the presence of tetracycline, which is used clinically in treating infections, the seeded stem-like cells may overgrow the CSCs (Speer et al. 1992 ; Matthess et al. 2005 ). Subsequently, after successfully replacement, the seeded stem-like cells could be eradicated by adding ‘drug two’ which would activate the suicide genes incorporated before seeding. One possible system fits this scenario was developed by Dong et al. ( 2004 ). They found that the solid tumor mass formed by the breast cancer cells, which were genetically modified to incorporate the Grp78 promoter driven HSV - tk suicide gene, could be completely eradicated with the treatment of drug ganciclovir (GCV), which is in clinical use for treating cytomegalovirus (CMV) infections. To make sure that all the progenies of the seed cells could be killed upon induction, multiple suicide genes, flanked by strong controllable promoters, should be recombined into the genome of these stem-like cells before transplantation. In all these procedures, it is not necessary for the seeded cells to secrete hostile anti-tumor substances and consequently, it may reduce therapeutic uncertainty. This strategy may be advantageous given that it incorporates the benefits of both traditional chemo-therapy and the most recent stem cell based drug delivery system.

We hypothesize that this strategy has higher tumor specificity and lower cytotoxicity. Empirically, the seeded cells could be regarded as the ‘needle’ that enters the abnormal ‘organ’ and kill the ‘organ’ by replacing and depriving its initiating cells. The key to the success of this proposal/hypothesis is whether those injected stem-like cells could actually seed in the tumor mass and then replace CSCs in the presence of drug one, which we believe offers survival advantage for injected stem-like cells over the original CSCs (Fig.  1 ). Recent studies by Kim et al. ( 2009 ) showed that mainly mediated by tumor-derived attractants IL-6, IL-8 and proteins matrix metalloproteinase-1 (MMP-1) and fascin-1, circulating tumor cells (CTCs) can re-seed their tumors of origin. The process of tumor-self seeding was believed a process that may select more aggressive cancer cells for the tumor colonization and breeding of more metastatic cancer cell progenies (Kim et al. 2009 ; Hahnfeldt 2010 ; Dondossola et al. 2012 ). Given the tumor formation ability of CTCs and the fact that CSCs may constitute the majority of CTCs, it is reasonable to believe that the re-infiltrated CTCs may have some stem-like properties (Toloudi et al. 2011 ). However, the amount of stem-like cells is limited by the availability of the niche that provides essential signals maintaining the self-renewal and pluripotency ability of these stem-like cells. Then, some sort of competition may exist between the more aggressive re-seeding CTCs and the original residing CSCs for the limited niche, as what happens between CTCs and the hematopoietic stem cells (HSCs) when CTCs try to establish footholds in bone marrow (Shiozawa et al. 2011 ). This kind of competition for the limited stem cell niche constitutes the major foundation of our hypothesis and we reasonably hold that in the presence of drug one that favors the out-growth of the engineered stem-like cells, injected stem-like cells may finally replace the original CSCs in the niche, as depicted in Fig.  1 .

Proposal 2. Manipulate the CSCs to our advantage: creating functional organs from CSCs?

In the last decade, conceptual progresses in cancer biology highlighted another emerging hallmark of human malignancies-their ability to evade immune destruction (Hanahan and Weinberg 2011 ). In this case, the so-called donor-related malignancy, in which immunosuppressed organ transplant recipients could potentially develop donor derived cancers, indicating that in immunocompetent donors the cancer cells were held in a dormant state by a fully functional immune system, whereas in the immunosuppressed recipients these cancer cells could be reactivated and progress (Strauss and Thomas 2010 ). Theoretically, cancer cells paralyze anti-tumor immune surveillance by secreting immunosuppressive factors like TGF-β and chemokine ligand (CCL21), by recruiting inflammatory cells like regulatory T cells (Tregs) and myeloid-derived suppressor cells or by evading the evolutionally deprived immune system of the host (Hanahan and Weinberg 2011 ; Ostrand-Rosenberg and Sinha 2009 ; Mougiakakos et al. 2010 ; Shields et al. 2010 ; Yang et al. 2010 ).

Although not well understood, recent studies suggested that CSCs possessed the functional and phenotypic characteristics that helped cancer cells evade host antitumor immunity. For example, a novel type of CSCs, ABCB5 + malignant melanoma initiating cells (MMIC) were reported refractory to current immunotherapeutic treatment strategies (Schatton and Frank 2009 ). And interestingly, CSCs derived from glioblastoma multiform patients could suppress immune responses by impeding T-cell proliferation and activation, inducing T-cell apoptosis and inhibiting the phagocytosis ability of MΦs/microglia (Wei et al. 2010a , b ; Wu et al. 2010 ).

Two hallmarks of CSCs are revealed here: CSCs are pluripotent stem-like cells with self-renewal ability and they are immune suppressive. The ideal combination of these two characteristics promises significant therapeutic implications beyond cancer therapy and we propose/hypothesize here that CSCs could be potentially engineered to create functional tissues and organs for transplantation (Fig.  3 ). Ideally, these organs are immune-tolerated by universal recipients. Here we tried to define the word ‘universal’ as ‘a group of individuals of the same species’. This definition constitutes the major highlight of this proposal.

An external file that holds a picture, illustration, etc.
Object name is 10616_2012_9517_Fig3_HTML.jpg

Schematic representation illustrating proposal 2. Given that cancer stem cells have characteristics of normal stem cells, we hypothesize that we could possibly direct the differentiation of CSCs to normal organs. Since cancer cells could efficiently evade the immuno-surveillance of the host, we propose that the functional organ might also be well tolerated by the recipients, even though those CSCs are derived from other individuals

Technically, directed differentiation of normal embryonic and iPS cells into specific organ cell types is relatively well studied at least for certain tissues. For instance, iPS cells had been engineered to differentiate into liver hepatocytes and pancreatic endocrine (Cai et al. 2007 ; D’Amour et al. 2006 ; Song et al. 2009 ; Zhang et al. 2009 ). Moreover, a recent study reported the successful directed differentiation of human iPS cells into functional intestinal tissue in vitro by manipulating a temporal series of growth factors (Spence et al. 2011 ). CSCs could also be differentiated into functional cells, since in glioblastoma multiforme and medulloblastoma, stem-like neurosphere-forming glioma stem cells were manipulated to give rise to neuronal and astroglial-like cells (Hemmati et al. 2003 ; Singh et al. 2003 ). In this context, the so-call differentiation therapy is now widely studied. Retinoic acid (RA, Vitamin A), for example, is one of the most thoroughly studied drugs that are capable of inducing CSC differentiation (Sell 2004 ; Tang and Gudas 2011 ; Gudas and Wagner 2011 ). However, mainly due to retinoid resistance and some serious side effects, the clinical outcome of RA treatment is not satisfactory for many solid tumors (Sell 2004 ). On another side, given their pluripotent and immunosuppressive properties, it might be possible to produce CSCs into functional organs for clinical transplantation. So, instead of killing them, we foresee here a promising application of these novel stem-like cells to the advantage of our healthcare.

Conclusion and prospects

Drugs that target CSCs implicate enormous therapeutic potential for cancer therapy. However, this enthusiasm needs to be tempered. The stark reality that drugable signaling pathways, such as Wnt, Hedgehog, and Notch, that are aberrantly modulated in CSCs are also critical for normal adult stem cell maintenance calls for re-evaluation of current drug-screening methods. However, it is becoming increasingly apparent that the concept of CSCs in tumor progression highlights the significance of stem cell based drug delivery system. In spite of that, the nomination or even the validity of the CSC hypothesis remains critically questioned, mainly due to the problems discussed in this article, the concept is a great leap forward on how to detect the process of tumorigenesis, how to develop efficient therapies and most importantly, if the tumor colonies are maintained by the rare CSCs, and how to prevent their initiation in the first place. The rarity of CSCs in the cancer cell populations, their instability when cultured in vitro and their similarity with normal tissue stem cells thwart the identification and clinical application of CSC specific agents. Meanwhile, the clinical applicability of the stem cell based drug delivery system was seriously compromised by its inherent therapeutic uncertainty.

Instead of killing CSCs by environmental hostile agents, we proposed a novel stem cell based strategy in proposal 1 in which CSCs were replaced by implanted stem-like cells. By doing so, we could circumvent undesirable disadvantages that come along with stem cell based anti-tumor therapy. Moreover, can we manipulate the CSCs to our advantage? Based on pluripotent and immunosuppressive characteristics of CSCs, we hypothesized a novel application of CSCs in proposal 2. A rational question concerning the relationship between the proposal 1 and 2 is that when we know how to direct the differentiation of CSCs into functional tissue cells, why don’t we use a similar strategy to induce CSCs differentiation in vivo and then cure the cancer? Presumably, the directed differentiation of CSCs as highlighted in the proposal 2 is a multiple-step process, happens in a spatial and temporal manner, and consequently, and at least in the actual state of knowledge and development, the differentiation strategy may be inapplicable for cancer treatment in vivo, although, hopefully, this may lead to the development of other more potent drugs that are suitable for the differentiation therapy.

According to our knowledge, the proposals we provided here are not yet experimentally explored. Here, with the help of the world renowned journal, we provide these strategies for possible exploration.

Acknowledgments

This work was supported partially by “National Natural Science Foundation of China” (No.31272100 and No.30901774), the “National High Technology Research and Development Program of China (863 Program)” (No.2012AA020809), the Innovative Research Program for Graduates of Shanxi Province (20113018) and the Program for the Top Young Academic Leaders of Higher Learning Institutions of Shanxi.

Conflict of interest

The authors state that they have no conflict of interest.

  • Ailles LE, Weissman IL. Cancer stem cells in solid tumors. Curr Opin Biotechnol. 2007; 18 :460–466. doi: 10.1016/j.copbio.2007.10.007. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Albarenque SM, Zwacka RM, Mohr A. Both human and mouse mesenchymal stem cells promote breast cancer metastasis. Stem Cell Res. 2011; 7 :163–171. doi: 10.1016/j.scr.2011.05.002. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumourigenic breast cancer cells. Proc Natl Acad Sci USA. 2003; 100 :3983–3988. doi: 10.1073/pnas.0530291100. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bjerkvig R, Tysnes BB, Aboody KS, Najbauer J, Terzis AJ. Opinion: the origin of the cancer stem cell: current controversies and new insights. Nat Rev Cancer. 2005; 5 :899–904. doi: 10.1038/nrc1740. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Bonnett D, Dick JE. Human acute myeloid leukaemia is organized as a hierarchy that originates from a primate haematopoietic cell. Nat Med. 1997; 3 :730–737. doi: 10.1038/nm0797-730. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Burness ML, Sipkins DA. The stem cell niche in health and malignancy. Semin Cancer Biol. 2010; 20 :107–115. doi: 10.1016/j.semcancer.2010.05.006. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Cai J, Zhao Y, Liu Y, Ye F, Song Z, Qin H, Meng S, Chen Y, Zhou R, Song X, Guo Y, Ding M, Deng H. Directed differentiation of human embryonic stem cells into functional hepatic cells. Hepatology. 2007; 45 :1229–1239. doi: 10.1002/hep.21582. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Chaffer CL, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA, Rodrigues LO, Brooks M, Reinhardt F, Su Y, Polyak K, Arendt LM, Kuperwasser C, Bierie B, Weinberg RA. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci USA. 2011; 108 :7950–7955. doi: 10.1073/pnas.1102454108. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Cho RW, Clarke MF. Recent advances in cancer stem cells. Curr Opin Genet Dev. 2008; 18 :48–53. doi: 10.1016/j.gde.2008.01.017. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM. Cancer stem cells–perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res. 2006; 66 :9339–9344. doi: 10.1158/0008-5472.CAN-06-3126. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • D’Amour KA, Bang AG, Eliazer S, Kelly OG, Agulnick AD, Smart NG, Moorman MA, Kroon E, Carpenter M, Baetge EE. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006; 24 :1392–1401. doi: 10.1038/nbt1259. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Dondossola E, Crippa L, Colombo B, Ferrero E, Corti A. Chromogranin A regulates tumor self-seeding and dissemination. Cancer Res. 2012; 72 :449–459. doi: 10.1158/0008-5472.CAN-11-2944. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Dong D, Dubeau L, Bading J, Nguyen K, Luna M, Yu H, Gazit-Bornstein G, Gordon EM, Gomer C, Hall FL, Gambhir SS, Lee AS. Spontaneous and controllable activation of suicide gene expression driven by the stress-inducible grp78 promoter resulting in eradication of sizable human tumors. Hum Gene Ther. 2004; 15 :553–561. doi: 10.1089/104303404323142006. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008; 15 :504–514. doi: 10.1038/sj.cdd.4402283. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Gudas LJ, Wagner JA. Retinoids regulate stem cell differentiation. J Cell Physiol. 2011; 226 :322–330. doi: 10.1002/jcp.22417. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, Lander ES. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell. 2009; 138 :645–659. doi: 10.1016/j.cell.2009.06.034. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hahnfeldt P. Significance of tumor self-seeding as an augmentation to the classic metastasis paradigm. Future Oncol. 2010; 6 :681–685. doi: 10.2217/fon.10.43. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144 :646–674. doi: 10.1016/j.cell.2011.02.013. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI. Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci USA. 2003; 100 :15178–15183. doi: 10.1073/pnas.2036535100. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Hwang-Verslues WW, Kuo WH, Chang PH, Pan CC, Wang HH, Tsai ST, Jeng YM, Shew JY, Kung JT, Chen CH, Lee EY, Chang KJ, Lee WH. Multiple lineages of human breast cancer stem/progenitor cells identified by profiling with stem cell markers. PLoS ONE. 2009; 4 :e8377. doi: 10.1371/journal.pone.0008377. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Jordan CT, Guzman ML, Noble M. Cancer Stem Cells. N Engl J Med. 2006; 355 :1253–1261. doi: 10.1056/NEJMra061808. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R, Weinberg RA. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007; 449 :557–563. doi: 10.1038/nature06188. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, Massagué J. Tumor self-seeding by circulating cancer cells. Cell. 2009; 139 :1315–1326. doi: 10.1016/j.cell.2009.11.025. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994; 367 :645–648. doi: 10.1038/367645a0. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Maenhaut C, Dumont JE, Roger PP, van Staveren WC. Cancer stem cells: a reality, a myth, a fuzzy concept or a misnomer? An analysis. Carcinogenesis. 2010; 31 :149–158. doi: 10.1093/carcin/bgp259. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Matthess Y, Kappel S, Spänkuch B, Zimmer B, Kaufmann M, Strebhardt K. Conditional inhibition of cancer cell proliferation by tetracycline-responsive, H1 promoter-driven silencing of PLK1. Oncogene. 2005; 24 :2973–2980. doi: 10.1038/sj.onc.1208472. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Mougiakakos D, Choudhury A, Lladser A, Kiessling R, Johansson CC. Regulatory T cells in cancer. Adv Cancer Res. 2010; 107 :57–117. doi: 10.1016/S0065-230X(10)07003-X. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Ostrand-Rosenberg S, Sinha P. Myeloid-derived suppressor cells: linking inflammation and cancer. J Immunol. 2009; 182 :4499–4506. doi: 10.4049/jimmunol.0802740. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008; 456 :593–598. doi: 10.1038/nature07567. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001; 414 :105–111. doi: 10.1038/35102167. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Rosen JM, Jordan CT. The increasing complexity of the cancer stem cell paradigm. Science. 2009; 324 :1670–1673. doi: 10.1126/science.1171837. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Schatton T, Frank MH. Antitumor immunity and cancer stem cells. Ann N Y Acad Sci. 2009; 1176 :154–169. doi: 10.1111/j.1749-6632.2009.04568.x. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Sell S. Stem cell origin of cancer and differentiation therapy. Crit Rev Oncol Hematol. 2004; 51 :1–28. doi: 10.1016/j.critrevonc.2004.04.007. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Shackleton M. Normal stem cells and cancer stem cells: similar and different. Semin Cancer Biol. 2010; 20 :85–92. doi: 10.1016/j.semcancer.2010.04.002. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Shah K (2012) Mesenchymal stem cells engineered for cancer therapy. Adv Drug Deliv Rev 64:739–748 [ PMC free article ] [ PubMed ]
  • Shields JD, Kourtis IC, Tomei AA, Roberts JM, Swartz MA. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science. 2010; 328 :749–752. doi: 10.1126/science.1185837. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, Kim JK, Patel LR, Ying C, Ziegler AM, Pienta MJ, Song J, Wang J, Loberg RD, Krebsbach PH, Pienta KJ, Taichman RS. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest. 2011; 121 :1298–12312. doi: 10.1172/JCI43414. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003; 63 :5821–5828. [ PubMed ] [ Google Scholar ]
  • Song Z, Cai J, Liu Y, Zhao D, Yong J, Duo S, Song X, Guo Y, Zhao Y, Qin H, Yin X, Wu C, Che J, Lu S, Ding M, Deng H. Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Res. 2009; 19 :1233–1242. doi: 10.1038/cr.2009.107. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Speer BS, Shoemaker NB, Salyers AA. Bacterial resistance to tetracycline: mechanisms, transfer, and clinical significance. Clin Microbiol Rev. 1992; 5 :387–399. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM, Shroyer NF, Wells JM. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011; 470 :105–109. doi: 10.1038/nature09691. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Strauss DC, Thomas JM. Transmission of donor melanoma by organ transplantation. Lancet Oncol. 2010; 11 :790–796. doi: 10.1016/S1470-2045(10)70024-3. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Takaishi S, Okumura T, Tu S, Wang SS, Shibata W, Vigneshwaran R, Gordon SA, Shimada Y, Wang TC. Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells. 2009; 27 :1006–1020. doi: 10.1002/stem.30. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 2011; 8 :97–106. doi: 10.1038/nrclinonc.2010.196. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Tang XH, Gudas LJ. Retinoids, retinoic acid receptors, and cancer. Annu Rev Pathol. 2011; 6 :345–364. doi: 10.1146/annurev-pathol-011110-130303. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Toloudi M, Apostolou P, Chatziioannou M, Papasotiriou I. Correlation between cancer stem cells and circulating tumor cells and their value. Case Rep Oncol. 2011; 4 :44–54. doi: 10.1159/000324403. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Tsai KS, Yang SH, Lei YP, Tsai CC, Chen HW, Hsu CY, Chen LL, Wang HW, Miller SA, Chiou SH, Hung MC, Hung SC. Mesenchymal stem cells promote formation of colorectal tumors in mice. Gastroenterology. 2011; 141 :1046–1056. doi: 10.1053/j.gastro.2011.05.045. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008; 8 :755–768. doi: 10.1038/nrc2499. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Voog J, Jones DL. Cell stem cell—stem cells and the niche: a dynamic duo. Cell Stem Cell. 2010; 6 :103–115. doi: 10.1016/j.stem.2010.01.011. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, Henry V, Colman H, Priebe W, Sawaya R, Lang FF, Heimberger AB. Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway. Mol Cancer Ther. 2010; 9 :67–78. doi: 10.1158/1535-7163.MCT-09-0734. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Wei J, Barr J, Kong LY, Wang Y, Wu A, Sharma AK, Gumin J, Henry Hyperlink V, Colman H, Sawaya R, Lang FF, Heimberger AB. Glioma-associated cancer-initiating cells induce immunosuppression. Clin Cancer Res. 2010; 16 :461–473. doi: 10.1158/1078-0432.CCR-09-1983. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ] Retracted
  • Wu A, Wei J, Kong LY, Wang Y, Priebe W, Qiao W, Sawaya R, Heimberger AB. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol. 2010; 12 :1113–1125. doi: 10.1093/neuonc/noq082. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Yang L, Pang Y, Moses HL. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010; 31 :220–227. doi: 10.1016/j.it.2010.04.002. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Zhang D, Jiang W, Liu M, Sui X, Yin X, Chen S, Shi Y, Deng H. Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell Res. 2009; 19 :429–438. doi: 10.1038/cr.2009.28. [ PubMed ] [ CrossRef ] [ Google Scholar ]
  • Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature. 2011; 474 :212–215. doi: 10.1038/nature10135. [ PubMed ] [ CrossRef ] [ Google Scholar ]

Cancer Stem Cell (CSC) Hypothesis

  • Download PDF Copy

Hidaya Aliouche, B.Sc.

Cancer cells are considered to be aberrant versions of their tissues of origin. However, their ability to proliferate and resistance to therapy led to the theory that cancers contain a heterogeneous population of cells. One theory that has gained traction since a hallmark paper was published in 2001 is the cancer stem cell (CSC) hypothesis.

By Kateryna Kon

Kateryna Kon | Shutterstock

The first assumption of the CSC hypothesis is that the cells in any given cancer mass are unequal, or heterogeneous. This describes a tumor in which cancer cells are hierarchically organized, and contain a small subpopulation of cancer stem cells (CSCs) that can proliferate extensively to sustain the progression and growth of the tumor.

The rationale behind this theory is that stem cells possess two properties, or hallmarks; (1) indefinite self-renewal and (2) pluripotency. The latter refers to the differentiation of the stem cell to become a specific cell type which is unipotent (can only produce daughter cells of the same type).

The second assumption of the CSC hypothesis is that CSCs are physically identifiable, possessing distinctive surface markers which differentiate them from other cancer cells.

Thirdly, the CSCs in a tumor comprises a mixture of cells which are either tumor-promoting (tumorigenic) or benign (non-tumorigenic). The tumorigenic population of CSCs is often considered to be tumor-initiating cells (TICs), aiding tumor relapse and resistance to therapy. For this reason, TICs are important therapeutic targets in drug development.

Evidence for CSCs

The most persuasive evidence for the existence of stem cells within a tumor comes from the analysis of cell surface markers. In 2003, Al-Hajj and colleagues identified that, in 8/9 primary breast tumor samples, the tumorigenic cells were CD44 + /CD24 -/low /ESA + /lin - i.e. positive for marker CD44, negative or low for CD24, positive for the marker Epithelial Specific Antigen (ESA), and negative for the lineage (lin) marker. This identified the breast cancer stem cells.

Singh et al . revealed that the tumorigenic brain cancer stem cell population was characterized by CD133 + . These cells could produce new tumor colonies termed neurospheres and could be reproduced in human brain tumors when injected into mice. Similarly, human colorectal cancer cells possessed the same qualities of tumor colony initiation and tumor reproduction. This evidence, whilst robust, has been contested.

Evidence Against CSCs?

The CSC hypothesis has been directly refuted by work that has demonstrated that tumors can grow rapidly; if CSCs existed, and were drivers of formation, they should be infrequent and identifiable by surface markers.

Related Stories

  • New method efficiently converts stem cells to endothelial cells for cardiovascular research
  • Stem cell models unlock secrets of early human embryo development
  • Primitive endoderm in early embryos may be key to creating new effective fertility treatments

In 2007, Andreas Strasser found that a high proportion (1 in 10) of leukemia cells transplanted into mice could initiate lymphomas. Similarly, Sean Morrison found 25% of unselected melanoma cells (that is, without any surface marker) could form tumors in mice. These findings have been refuted since, with an increasing body of evidence pointing to the existence of CSCs possessing the qualities of infrequency, isolation by surface markers, the ability to form tumors following xenotransplantation.

Origins of CSCS: the connection between Epithelial-Mesenchymal Transition (EMT) and CSCs

The Epithelial-Mesenchymal Transition (EMT) is the process in which differentiated cells de-differentiate to acquire stem cell properties. Oncogenic mutations are posited to drive this process and promote self-renewal.

This mechanistic link is palpable, as it suggests that a large cell population could possess tumorigenic potential (as found by Strasser and Morrison), but only a small subset would initiate the tumor. The evidence for this comes from the work of Thomas Brabletz (2005) and Mani, which was published in Cell in 2008.

More specifically, factors that control gene expression called transcription factors, were found to be upregulated in CSCs. These transcription factors activated transcriptional programs that are seen in stem cells; the factors that are known to cause EMT, SNAIL and TWIST were identified.

Furthermore, extrinsic drivers of CSC production were found to promote the transcription of factors needed for the EMT. This was demonstrated by Scheel et al., who found that mammary cells underwent EMT by extrinsic signaling molecules TGF-β and Wnt.

The EMT program in mammary cells subsequently induced migration and sphere formation. In agreement, inhibition of extrinsic factors in transformed cells prevented tumorigenesis and metastatic spread. Thus, circulating tumor cells have features of both EMT and cancer stem cells and play a substantial role in resistance and metastasis .

Implications for current and emerging therapies

The notion that EMT induction leads to the formation of CSCs has been supported by research in more recent years and suggested that targeting the EMT process can represent a potential therapeutic target to treat cancer. Conventional chemotherapy primarily targets the numerous highly proliferative cells to reduce the tumor size, promoting the increased frequency of CSCs that arise through EMT.

This subsequently replenishes these lost cells, and tumor recurrence results. Direct target of CSCs, through interference with signaling pathways and transcription factors implicated in EMT, and thus CSC production - termed ablation - would prevent cancer stem cells from providing differentiated cells to populate the tumor mass.

Such inhibition of tumor regeneration would ultimately lead to the regression of the tumor. Efforts are currently underway and include blocking stem cell self-renewal pathways, targeting the microenvironment that sustains CSCs, inducing CSC programmed cell death, as well as forcing CSCs to differentiate into non-proliferative cell types amongst targeting the EMT pathway.

  • https://www.nature.com/articles/35102167
  • https://www.ncbi.nlm.nih.gov/pubmed/9212098
  • http://www.pnas.org/content/100/7/3983.long
  • https://www.ncbi.nlm.nih.gov/pubmed/18485877
  • https://www.nature.com/articles/nrc1694
  • https://www.ncbi.nlm.nih.gov/pubmed/21663795
  • http://science.sciencemag.org/content/317/5836/337.long
  • https://www.nature.com/articles/nature07567
  • https://www.nature.com/articles/nature03128

Last Updated: Dec 6, 2018

Hidaya Aliouche

Hidaya Aliouche

Hidaya is a science communications enthusiast who has recently graduated and is embarking on a career in the science and medical copywriting. She has a B.Sc. in Biochemistry from The University of Manchester. She is passionate about writing and is particularly interested in microbiology, immunology, and biochemistry.

Please use one of the following formats to cite this article in your essay, paper or report:

Aliouche, Hidaya. (2018, December 06). Cancer Stem Cell (CSC) Hypothesis. News-Medical. Retrieved on August 26, 2024 from https://www.news-medical.net/life-sciences/Cancer-Stem-Cell-Hypothesis.aspx.

Aliouche, Hidaya. "Cancer Stem Cell (CSC) Hypothesis". News-Medical . 26 August 2024. <https://www.news-medical.net/life-sciences/Cancer-Stem-Cell-Hypothesis.aspx>.

Aliouche, Hidaya. "Cancer Stem Cell (CSC) Hypothesis". News-Medical. https://www.news-medical.net/life-sciences/Cancer-Stem-Cell-Hypothesis.aspx. (accessed August 26, 2024).

Aliouche, Hidaya. 2018. Cancer Stem Cell (CSC) Hypothesis . News-Medical, viewed 26 August 2024, https://www.news-medical.net/life-sciences/Cancer-Stem-Cell-Hypothesis.aspx.

Suggested Reading

Precise protein design offers hope for damaged hearts and kidneys

  • Trending Stories
  • Latest Interviews
  • Top Life Sciences Articles

Study reveals genetic link between Alzheimer's disease, lipid metabolism, and coronary artery disease

Global and Local Efforts to Take Action Against Hepatitis

Lindsey Hiebert and James Amugsi

In this interview, we explore global and local efforts to combat viral hepatitis with Lindsey Hiebert, Deputy Director of the Coalition for Global Hepatitis Elimination (CGHE), and James Amugsi, a Mandela Washington Fellow and Physician Assistant at Sandema Hospital in Ghana. Together, they provide valuable insights into the challenges, successes, and the importance of partnerships in the fight against hepatitis.

Global and Local Efforts to Take Action Against Hepatitis

Addressing Important Cardiac Biology Questions with Shotgun Top-Down Proteomics

In this interview conducted at Pittcon 2024, we spoke to Professor John Yates about capturing cardiomyocyte cell-to-cell heterogeneity via shotgun top-down proteomics.

Addressing Important Cardiac Biology Questions with Shotgun Top-Down Proteomics

A Discussion with Hologic’s Tim Simpson on the Future of Cervical Cancer Screening

Tim Simpson

Hologic’s Tim Simpson Discusses the Future of Cervical Cancer Screening.

A Discussion with Hologic’s Tim Simpson on the Future of Cervical Cancer Screening

Latest Life Science News

Asgard archaea's role in eukaryotic immunity revealed

Newsletters you may be interested in

Bowel Cancer

Your AI Powered Scientific Assistant

Hi, I'm Azthena, you can trust me to find commercial scientific answers from News-Medical.net.

A few things you need to know before we start. Please read and accept to continue.

  • Use of “Azthena” is subject to the terms and conditions of use as set out by OpenAI .
  • Content provided on any AZoNetwork sites are subject to the site Terms & Conditions and Privacy Policy .
  • Large Language Models can make mistakes. Consider checking important information.

Great. Ask your question.

Azthena may occasionally provide inaccurate responses. Read the full terms .

While we only use edited and approved content for Azthena answers, it may on occasions provide incorrect responses. Please confirm any data provided with the related suppliers or authors. We do not provide medical advice, if you search for medical information you must always consult a medical professional before acting on any information provided.

Your questions, but not your email details will be shared with OpenAI and retained for 30 days in accordance with their privacy principles.

Please do not ask questions that use sensitive or confidential information.

Read the full Terms & Conditions .

Provide Feedback

cancer stem cell hypothesis wiki

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

The cancer stem cell hypothesis: failures and pitfalls

Affiliation.

  • 1 Department of Neurosurgery, University of Florida, Gainesville, Florida, USA. [email protected]
  • PMID: 21135745
  • DOI: 10.1227/NEU.0b013e3181ff9eb5

Based on the clonal evolution model and the assumption that the vast majority of tumor cells are able to propagate and drive tumor growth, the goal of cancer treatment has traditionally been to kill all cancerous cells. This theory has been challenged recently by the cancer stem cell (CSC) hypothesis, that a rare population of tumor cells, with stem cell characteristics, is responsible for tumor growth, resistance, and recurrence. Evidence for putative CSCs has been described in blood, breast, lung, prostate, colon, liver, pancreas, and brain. This new hypothesis would propose that indiscriminate killing of cancer cells would not be as effective as selective targeting of the cells that are driving long-term growth (ie, the CSCs) and that treatment failure is often the result of CSCs escaping traditional therapies.The CSC hypothesis has gained a great deal of attention because of the identification of a new target that may be responsible for poor outcomes of many aggressive cancers, including malignant glioma. As attractive as this hypothesis sounds, especially when applied to tumors that respond poorly to current treatments, we will argue in this article that the proposal of a stemlike cell that initiates and drives solid tissue cancer growth and is responsible for therapeutic failure is far from proven. We will present the point of view that for most advanced solid tissue cancers such as glioblastoma multiforme, targeting a putative rare CSC population will have little effect on patient outcomes. This review will cover problems with the CSC hypothesis, including applicability of the hierarchical model, inconsistencies with xenotransplantation data, and nonspecificity of CSC markers.

PubMed Disclaimer

Similar articles

  • Insights into the cell of origin in breast cancer and breast cancer stem cells. Lindeman GJ, Visvader JE. Lindeman GJ, et al. Asia Pac J Clin Oncol. 2010 Jun;6(2):89-97. doi: 10.1111/j.1743-7563.2010.01279.x. Asia Pac J Clin Oncol. 2010. PMID: 20565420 Review.
  • Die hard: are cancer stem cells the Bruce Willises of tumor biology? Fábián A, Barok M, Vereb G, Szöllosi J. Fábián A, et al. Cytometry A. 2009 Jan;75(1):67-74. doi: 10.1002/cyto.a.20690. Cytometry A. 2009. PMID: 19051297 Review.
  • Cancer stem cells and glioma. Das S, Srikanth M, Kessler JA. Das S, et al. Nat Clin Pract Neurol. 2008 Aug;4(8):427-35. doi: 10.1038/ncpneuro0862. Epub 2008 Jul 15. Nat Clin Pract Neurol. 2008. PMID: 18628751 Review.
  • Cancer stem cells and their potential implications for the treatment of solid tumors. Grotenhuis BA, Wijnhoven BP, van Lanschot JJ. Grotenhuis BA, et al. J Surg Oncol. 2012 Aug 1;106(2):209-15. doi: 10.1002/jso.23069. Epub 2012 Feb 27. J Surg Oncol. 2012. PMID: 22371125 Review.
  • Cancer stem cells in hepatocellular carcinoma: Recent progress and perspective. Chiba T, Kamiya A, Yokosuka O, Iwama A. Chiba T, et al. Cancer Lett. 2009 Dec 28;286(2):145-53. doi: 10.1016/j.canlet.2009.04.027. Epub 2009 May 22. Cancer Lett. 2009. PMID: 19464789 Review.
  • Drug tolerant persister cell plasticity in cancer: A revolutionary strategy for more effective anticancer therapies. He J, Qiu Z, Fan J, Xie X, Sheng Q, Sui X. He J, et al. Signal Transduct Target Ther. 2024 Aug 14;9(1):209. doi: 10.1038/s41392-024-01891-4. Signal Transduct Target Ther. 2024. PMID: 39138145 Free PMC article. Review.
  • Cancer-associated fibroblasts and prostate cancer stem cells: crosstalk mechanisms and implications for disease progression. Chen H, Fang S, Zhu X, Liu H. Chen H, et al. Front Cell Dev Biol. 2024 Jul 18;12:1412337. doi: 10.3389/fcell.2024.1412337. eCollection 2024. Front Cell Dev Biol. 2024. PMID: 39092186 Free PMC article. Review.
  • Spontaneous Fusion with Transformed Mesenchymal Stromal Cells Results in Complete Heterogeneity in Prostate Cancer Cells. Wang R, Hu P, Wang F, Lyu J, Ou Y, Edderkaoui M, Zhang Y, Lewis MS, Pandol SJ, Zhau HE, Chung LWK. Wang R, et al. Cancers (Basel). 2024 Feb 27;16(5):951. doi: 10.3390/cancers16050951. Cancers (Basel). 2024. PMID: 38473313 Free PMC article.
  • Spatial distribution and functional relevance of FGFR1 and FGFR2 expression for glioblastoma tumor invasion. Alshahrany N, Begum A, Siebzehnrubl D, Jimenez-Pascual A, Siebzehnrubl FA. Alshahrany N, et al. Cancer Lett. 2023 Sep 1;571:216349. doi: 10.1016/j.canlet.2023.216349. Epub 2023 Aug 12. Cancer Lett. 2023. PMID: 37579831 Free PMC article.
  • Epithelial Ovarian Cancer: A Five Year Review. Arnaoutoglou C, Dampala K, Anthoulakis C, Papanikolaou EG, Tentas I, Dragoutsos G, Machairiotis N, Zarogoulidis P, Ioannidis A, Matthaios D, Perdikouri EI, Giannakidis D, Sardeli C, Petousis S, Oikonomou P, Nikolaou C, Charalampidis C, Sapalidis K. Arnaoutoglou C, et al. Medicina (Kaunas). 2023 Jun 21;59(7):1183. doi: 10.3390/medicina59071183. Medicina (Kaunas). 2023. PMID: 37511995 Free PMC article. Review.

Publication types

  • Search in MeSH

LinkOut - more resources

Full text sources.

  • Ovid Technologies, Inc.
  • Wolters Kluwer

Research Materials

  • NCI CPTC Antibody Characterization Program

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

COMMENTS

  1. Cancer stem cell

    The cancer stem cell model, also known as the Hierarchical Model proposes that tumors are hierarchically organized (CSCs lying at the apex [6] (Fig. 3).) Within the cancer population of the tumors there are cancer stem cells (CSC) that are tumorigenic cells and are biologically distinct from other subpopulations [7] They have two defining features: their long-term ability to self-renew and ...

  2. The cancer stem cell hypothesis: a work in progress

    That adult stem cells give rise to cancer is an attractive hypothesis, given that the classic multistep model of carcinogenesis requires a long-lived cell in which multiple genetic hits can occur ...

  3. Warburg hypothesis

    The Warburg hypothesis ( / ˈvɑːrbʊərɡ / ), sometimes known as the Warburg theory of cancer, postulates that the driver of tumorigenesis is an insufficient cellular respiration caused by insult to mitochondria. [ 1] The term Warburg effect in oncology describes the observation that cancer cells, and many cells grown in vitro, exhibit ...

  4. The cancer stem cell hypothesis: in search of definitions, markers, and

    The cancer stem cell hypothesis largely ignores the inherent properties of malignant cells: genomic instability and the ability to undergo rapid evolutionary changes. For example, when 'tumor spheres' derived from highly vascular glioblastomas were transplanted into mice, the initial tumors demonstrated low-grade glioma phenotype without ...

  5. The cancer stem cell hypothesis: a work in progress

    There is a growing body of evidence that supports the idea that malignant tumors are initiated and maintained by a population of tumor cells that share similar biologic properties to normal adult stem cells. This model, the cancer stem cell (CSC) hypothesis, is based on the observation that tumors, like adult tissues, arise from cells that exhibit the ability to self-renew as well as give rise ...

  6. PDF The cancer stem cell hypothesis: a work in progress

    by a population of tumor cells that share similar biologic properties to normal adult stem cells. This model, the cancer stem cell (CSC) hypothesis, is based on the observation that tumors, like ...

  7. The cancer stem cell hypothesis: a work in progress

    That adult stem cells give rise to cancer is an attractive hypothesis, given that the classic multistep model of carcinogenesis requires a long-lived cell in which multiple genetic hits can occur. 1 In fact, investigators had long ago postulated that cancers arise from cells with stem cell properties or directly from tissue-specific stem cells ...

  8. The cancer stem cell hypothesis: a work in progress

    Abstract. There is a growing body of evidence that supports the idea that malignant tumors are initiated and maintained by a population of tumor cells that share similar biologic properties to normal adult stem cells. This model, the cancer stem cell (CSC) hypothesis, is based on the observation that tumors, like adult tissues, arise from cells ...

  9. The cancer stem cell hypothesis: a guide to potential ...

    Common cancer theories hold that tumor is an uncontrolled somatic cell proliferation caused by the progressive addition of random mutations in critical genes that control cell growth. Nevertheless, various contradictions related to the mutation theory have been reported previously. These events may …

  10. Somatic evolution in cancer

    The first malignant cell, that gives rise to the tumor, is often labeled a cancer stem cell. [87] The cancer stem-cell hypothesis relies on the fact that a lot of tumors are heterogeneous - the cells in the tumor vary by phenotype and functions. [87] [88] [89] Current research shows that in many cancers there is apparent hierarchy among cells.

  11. Implications of the Cancer Stem-Cell Hypothesis for Breast Cancer

    The cancer stem-cell hypothesis proposes that cancers arise in breast, stem, and/or progenitor cells through dysregulation of the normally tightly regulated process of self-renewal. It is important to emphasize that this hypothesis does not require that all cancers arise from normal tissue stem cells. Indeed, there is substantial evidence in ...

  12. The Cancer Stem-Cell Hypothesis

    The cancer stem-cell (CSC) hypothesis suggests that there is a small sub-set of cancer cells that are responsible for tumor initiation and growth, possessing properties such as indefinite self-renewal, slow replication, intrinsic resistance to chemotherapy and radiotherapy, and an ability to give rise to differentiated progeny.

  13. Cancer stem cell hypothesis: a brief summary and two proposals

    The investigation and development of the cancer stem cell (CSC) model has received much focus during these years. CSC is characterized as a small fraction of cancer cells that have an indefinite ability for self-renewal and pluripotency and are responsible for initiating and sustaining of the bulk of cancer. So, whether current treatment ...

  14. The cancer stem cell hypothesis: in search of definitions ...

    In breast cancer, putative cancer stem cells with CD24 −/low /CD44 + phenotype constituted 12-60% of the tumor cells, whereas in colon cancer, CD133+ putative cancer stem cells ranged from 3.8 ...

  15. PDF The Cancer Stem Cell Hypothesis: Challenges and Implications

    Over the past ten years, the cancer stem cell (CSC) hypothesis has produced an enormous amount of interest, optimism, and debate in the cancer research community. The biological principle at the heart of the CSC hypothesis is simple: in cancers there exist a small, distinct population of cells, "cancer stem cells", that have the ability to ...

  16. The cancer stem cell theory: Is it correct?

    The cancer stem cell hypothesis posits that tumor growth is driven by a rare subpopulation of cells, designated cancer stem cells (CSC). Studies supporting this theory are based in large part on xenotransplantation experiments wherein human cancer cells are grown in immunocompromised mice and only CSC, often constituting less than 1% of the malignancy, generate tumors.

  17. Cancer stem cells: advances in knowledge and implications for cancer

    Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity.

  18. Cancer stem cell hypothesis: a brief summary and two proposals

    Central to the heterogeneity of human malignancies is the existence of cancer stem cells (CSCs), a rare fraction of slow-cycling cancer cells that have the ability of self-renewal, pluripotency and chemo-resistance. In the presumptive CSC model, only the CSCs are tumorigenic and can produce all of the cells necessary to repopulate a tumor ...

  19. The cancer stem cell hypothesis: in search of definitions ...

    Cancer is a disease of genes. Inherited or somatic alterations in genes are what make a normal cell ignore growth-controlling signals and form a tumor that eventually leads to the destruction of the organism. Based on accumulated knowledge on the genetic composition of cancer cells, the clonal evolution model of tumorigenesis was established ...

  20. Cancer Stem Cell (CSC) Hypothesis

    Cancer Stem Cell (CSC) Hypothesis. Cancer cells are considered to be aberrant versions of their tissues of origin. However, their ability to proliferate and resistance to therapy led to the theory ...

  21. The cancer stem cell hypothesis: failures and pitfalls

    This theory has been challenged recently by the cancer stem cell (CSC) hypothesis, that a rare population of tumor cells, with stem cell characteristics, is responsible for tumor growth, resistance, and recurrence. Evidence for putative CSCs has been described in blood, breast, lung, prostate, colon, liver, pancreas, and brain.

  22. Metastasis

    Metastasis is a pathogenic agent's spread from an initial or primary site to a different or secondary site within the host's body; [1] the term is typically used when referring to metastasis by a cancerous tumor. [2] The newly pathological sites, then, are metastases (mets). [3] [4] It is generally distinguished from cancer invasion, which is the direct extension and penetration by cancer ...

  23. PDF The cancer stem cell hypothesis: in search of definitions ...

    The cancer stem cell hypothesis: in search of definitions, markers, and relevance Michail Shipitsin and Kornelia Polyak Cancer is a disease of genes. Inherited or somatic alterations in genes are ...