Semin Liver Dis 2010; 30(1): 026-034
DOI: 10.1055/s-0030-1247130
© Thieme Medical Publishers

Stem Cells in Hepatocarcinogenesis: Evidence from Genomic Data

Jens U. Marquardt1 , Snorri S. Thorgeirsson1
  • 1Laboratory of Experimental Carcinogenesis (LEC), Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
Further Information

Snorri S ThorgeirssonM.D. Ph.D. 

Laboratory of Experimental Carcinogenesis (LEC), Center for Cancer Research

National Cancer Institute, NIH, 37 Convent Drive, Room 4146, Bethesda, MD 20892

Email: snorri_thorgeirsson@nih.gov

Publication History

Publication Date:
19 February 2010 (online)

Table of Contents #

ABSTRACT

Increasing evidence suggests that many, perhaps all solid tumors contain a subset of cells that possess functional properties similar to the normal tissue stem cells, including self-renewal, unlimited proliferative capacity, and pluripotency. The hierarchical cancer model that places a cancer stem cell (CSC) population at the apex of tumor formation is based on this notion. The cancer stem cell hypothesis posits that CSCs are responsible not only for tumor initiation, but also generation of metastasis and local recurrence after therapy. Current definitions of the CSC are based only on functional properties regardless of potential cellular origin. Histopathology investigations of chronic liver diseases and experimental studies support the existence of CSCs in liver cancer. In particular, recent advances in microarray technologies utilizing integrative comparative genomic analysis of human hepatocellular carcinoma specimens, cancer cell lines, and transgenic models establish the molecular similarities between CSC and normal tissue stem cells and highlight the importance of CSC for the prognosis of liver cancer patients. The results have also uncovered the key “stemness” and oncogenic pathways frequently disrupted during hepatocarcinogenesis providing the basis for identifying novel therapeutic targets against CSC.

The concept that many solid tumors adopt a hierarchical organization is being increasingly recognized. Although the idea that only a small subpopulation of cells within each tumor possesses the capacity to generate tumors in vitro and in vivo is not new, research on the “cancer stem cells” (CSCs) in solid tumors mushroomed only in the last decade.[1] [2] [3] [4] [5] [6] [7] In contrast to the traditional stochastic cancer model, the hierarchical model postulates that cancer is a genetically generated disease that is driven by epigenetic changes originated in a minor cell population (<1%) possessing stemness features (commonly referred to as CSC) and occupying the apex of the tumor hierarchy. Similar to the phenotypic diversity of normal adult tissues maintained by the tissue stem cells, the CSC model is helpful to explain the heterogeneity observed within many clonally derived tumors including liver cancer.[7] [8] [9] [10] Although the CSC may have functional properties similar to the normal tissue stem cells, including self-renewal and differentiation capacity, the cancer stem cell hypothesis does not make any assumptions about a putative origin of these cells.[11]

Although numerous hypotheses have attempted to explain how cancer stem cells and tumor heterogeneity originate,[7] [12] the general concepts of carcinogenesis consider at least three different scenarios: (1) differentiation arrest of adult tissue stem cells and/or progenitor cells, (2) dedifferentiation of mature cells, and (3) transdifferentiation of stem cells from different tissues, for example, bone marrow. A combination of all three scenarios may provide another conceivable explanation. The relative contribution of each postulate may depend on number of factors, including type of cancer, tissue microenvironment, and the contributing mutagen(s).

It is axiomatic that comprehensive definition and characterization of the putative cancer stem cells and their cellular origin will improve both the understanding of tumor biology and the prospects of developing new cancer therapies. Because classical therapeutic regimens target proliferating cells, they are unlikely to target the CSC, which are thought to be low turnover/quiescent cells.[13] [14] [15] A detailed characterization of CSC would include the clarification of the underlying molecular, genetic, and epigenetic mechanisms responsible for tumor initiation, seeding of metastasis, and local recurrence that are currently attributed to the CSC.[12] [16] Transcriptome analyses utilizing microarray technologies have significantly advanced the field of cancer research in the last decade. Progressive application of these technologies for investigating CSCs seems particularly relevant to address several of the unresolved issues regarding the role of CSC in liver cancer.[17] [18]

#

CELLULAR ORGANIZATION OF LIVER AND HEPATOCARCINOGENESIS

The human liver is anatomically divided into different macroscopic and microscopic units: lobes and hepatic lobules. The functional unit or hepatic lobule is limited by six portal triads containing the hepatic artery, portal vein, and bile duct and centered on central vein. Venous blood from the intestine enters the liver from the portal vein and interconnects the portal triads with the systemic circulation while passing through fenestrated endothelial sinusoids that are tightly surrounded by the basolateral side of hepatocytes along with the resident nonparenchymal cells of the liver (e.g., stellate cells, Kupffer cells, immune cells, etc.). The apical sides of the hepatocytes form the bile canaliculi from which bile drains back toward the bile ducts. The most terminal branches of the biliary epithelium, denoted as Canals of Hering, are believed to be the niche for hepatic stem/progenitor cells (a more detailed description of the liver anatomy is provided elsewhere).[19] [20] Three types of liver cells, hepatocytes, cholangiocytes, and adult stem/progenitor cells, are commonly regarded as the primary targets of malignant transformation in the liver.

The extensive regenerative and proliferative capacity of mature hepatocytes is well recognized. Elegant experimentation with serial transplantations has shown that hepatocytes can undergo at least 69 doublings without losing functional potential; additionally, hepatocytes have a long lifespan.[21] [22] [23] Because these characteristics indicate that mature hepatocyte possesses intrinsic stem cell-like traits, it could be a target of malignant transformation.

The notion that hepatic stem/progenitor cells can drive hepatocarcinogenesis and be a source of CSC is historically well documented. Experimental induction of liver stem/progenitor cells in rodents has been extensively studied in liver injury models as well as in liver carcinogenesis. In agreement with the cancer stem cell model, the application of a variety of experimental protocols in rodents resulted in activation and proliferation of adult liver stem cells (often referred to as oval cells) and almost as frequently in carcinogenesis. Oval cells are putative liver stem/progenitor cells and were first described by Opie in 1944, and later by Farber.[24] [25] Oval cells are thought to reside in the Canals of Hering and, at a minimum, give rise to both cholangiocytes and hepatocytes.[26] [27] [28] Activation and proliferation of hepatic progenitor cells have been reported in many precancerous conditions, such as chronic inflammation (hepatitis B and C, alcoholic hepatitis, and steatohepatitis).[29] [30] [31] Therefore, liver stem/progenitor cells could be another source of CSC in HCC.

Although convincing evidence supporting a role for bone marrow-derived stem cells in the process of malignant transformation in liver is yet to be provided, there are data to suggest that bone marrow-derived cells may take part in liver regeneration and therefore should also be taken into consideration.[32] [33] [34] [35] For example, transplantation of adult bone marrow cells has been reported to restore liver tissue and function in fumarylacetoacetate hydrolase (FAH-) deficient animals,[36] although more recent data has demonstrated that the contribution of bone marrow cells to liver recovery was due to the fusion with the host hepatocytes.[37] Together, these observations emphasize that many different cell types may be the potential targets of a transforming event(s), thereby contributing to hepatocarcinogenesis (Fig. [1]). The different cellular origin may provide a potential explanation for the heterogeneity of CSCs and the phenotypic diversity of liver cancer.

Zoom Image

Figure 1 Potential cell types of origin of liver cancer stem cells (CSCs). The scheme illustrates the cellular organization of a portal triad consisting of a portal vein (PV), hepatic artery (A), and bile duct (BD). The most terminal branches of the biliary tree are referred to as Canals of Hering. CSCs can originate from the stem/progenitor cells thought to reside in Canals of Hering. They extensively proliferate upon adequate stimuli commonly present in hepatocarcinogenesis, for example, liver damage. CSCs can also originate from mature hepatocytes known to possess stem-like characteristics, such as proliferation capacity and longevity. Hepatocytes are continuously exposed to exogenous and endogenous stress and could be targets for transforming events. Lastly, bone marrow-derived stem cells could be a source of hepatic CSC through transdifferentiation.

#

EVIDENCE FOR A STEM CELLS ORIGIN IN LIVER CANCER

In the past, a plethora of transgenic mouse models and a variety of different chemical- or oncogene-induced carcinogenesis models were used to directly determine the role of stem cells in hepatocarcinogenesis. The common conclusion of the early studies was that HCC can originate from adult liver stem/progenitor cells.[38] [39] [40] [41] [42] Also, there is evidence for the stem/progenitor cell origin in human HCC. Thus, extensive stem/progenitor cell proliferation frequently referred to as “ductular” reaction can be observed in preneoplastic conditions of the liver, such as chronic inflammation. Similar to oval cell activation in rodents, the extent of ductular reaction correlated with the severity of the underlying liver disease.[43] [44] [45] Furthermore, after malignant conversion a substantial number of HCCs demonstrate “bipotential” characteristics: tumor cells coexpressing biliary and hepatocytic markers, such as cytokeratin 7 (CK7), CK19, OV6, α-fetoprotein (AFP), and albumin.[23] [46] The presence of these markers was associated with a more aggressive phenotype and bad clinical outcome.[47] An integrative study from our laboratory performed on a rat model of HCC further validated CK19 as a prognostic marker of early preneoplastic lesions suggesting a progenitor origin of these tumors. In addition, genomic analysis showed that CK19-associated gene signature stratified HCC patients according to clinical outcome.[48]

Other recent studies suggest that disruption of interleukin-6 (IL-6) and transforming growth factor-beta (TGF-β) signaling pathways in adult liver tissue stem cells was a key event leading to the transformation of pluripotent liver stem cells.[49] Another study reported that c-KIT (encodes a proto-oncogenic receptor tyrosine kinase) expressing proliferating liver progenitor cells were the targets of transforming events in a setting of chronic liver injury.[50] However, convincing evidence for the clonal origin of liver cancer from the stem cell compartment is still missing.

#

GENOMICS IN HEPATOCARCINOGENESIS

Over the last decade, the rapid advances in microarray technology have opened a new genomics era in biology and cancer research in particular. Whole genome transcriptomics and other array-based analyses provided more global and unbiased tools to study mechanisms of carcinogenesis in different tumor types.[51] In the liver, microarray analysis uncovered a variety of molecular signatures, signaling pathways, and gene sets associated with human liver cancer.[52] [53] [54] [55] Significantly, in addition to antiapoptotic and proproliferative signaling, many of the disrupted pathways in HCC have been shown to be involved in stem cell maintenance and self-renewal, for example, Wnt/β-catenin, TGF-β, Met (hepatocyte growth factor receptor), Hedgehog, as well as to be activated in adult liver progenitor cells upon regenerative stimuli.[21] [56] [57] [58] Because most of the HCCs develop in the background of chronic inflammatory driven regenerative cycles, activation of these pathways may be indicative of a stem cell origin at least in some liver tumors.[59] Clear prognostic significance of their disruption in different subtypes of liver cancer further confirms this finding.[60] Recent investigations used a more global approach and systematically combined transcriptional programs from different studies to construct a compendium of genes important for embryonic stem cells (ESCs) and adult tissue stem cells.[61] [62] The authors found activation of ESC-like transcriptional programs in aggressive human epithelial cancers including liver cancer. Interestingly, activation of the MYC oncogene was particularly important both for the tumor initiation and the reactivation of the ESC-like module in normal and cancer cells. This observation confirms the results from murine and human studies indicating the critical role for MYC in hepatocarcinogenesis.[63] [64] More recent study using transcriptomic analyses of cirrhotic and dysplastic nodules and early HCC further demonstrated activation of the MYC-associated gene sets in early HCC consistent with the central role of MYC in driving malignant conversion of preneoplastic liver lesions.[65] The subsequent classification based on the MYC target genes expression clearly discriminated between preneoplastic stages and early HCC.

Comparative genomic investigations of human HCC provided evidence supporting stem/progenitor cell origin in a subset of human liver cancer.[66] [67] In this study, gene expression data from rat fetal hepatoblasts (HB) and adult hepatocytes were integrated with the gene expression data from human HCC. HCCs, which shared gene expression pattern with HB (fetal progenitor cells), were profoundly different from other prognostic subtypes of HCC (Fig. [2]).[53] The gene expression signature of the HB subtype of HCC included hepatic oval cell markers and was strongly associated with worse prognosis, consistent with hepatic progenitor cell origin.[67] The prognostic implication of a progenitor signature was independently confirmed by others.[54] Similarly, classification of HCC based on the gene expression of the onco-fetal molecules, epithelial cell adhesion molecule (EpCAM) and AFP revealed distinct HCC subtypes resembling consecutive stages of hepatocyte differentiation. Gene expression signature of EpCAM+/AFP+ HCC was associated with poor survival and was characterized by activation of WNT–β-catenin, TGF-β, epidermal growth factor (EGF), and p53.

Zoom Image

Figure 2 Genomic evidence for a progenitor cell origin in liver cancer. The comparative functional genomic analysis of 139 human hepatocellular carcinomas (HCCs) and rat hepatoblasts/ hepatocytes identified 22 human HCCs that showed similar expression pattern to stem-like hepatoblasts (HBs; designated as HB type) (adapted from Lee et al[67]). Kaplan-Meier plots showed that patients with HB subtype HCC had worse survival and a higher frequency of recurrence. The study highlights the potential stem cell origin in a subtype of HCC and demonstrates clinical relevance of this finding.

Taken together, the transcriptomic analyses of HCC have significantly contributed to the understanding of the cellular origin of liver cancer and identified signaling pathways likely associated with progenitor cells. Integrative approaches further showed a clear prognostic significance of the progenitor cell-like signatures making them valuable tools for generating novel preventive, diagnostic, and/or therapeutic strategies for HCC patients.

#

PROSPECTIVE ISOLATION AND DEFINITION OF HEPATIC CANCER STEM CELLS

In the last years, considerable progress has been made in the prospective isolation and characterization of potential cancer stem cells in human HCC. Cancer stem cells are defined by at least three distinct properties: (1) self-renewing capacity—the ability to undergo symmetric and asymmetric division, thereby “infinitely” repopulating the cancer stem cell pool; (2) differentiation capacity—the ability to recapitulate all cell types of original tumor; and (3) tumor-initiating capacity—the ability to propagate tumors when transplanted into a proper environment.[11] Based on these definitions, two general approaches have been established for the isolation of CSC. First, and the most commonly employed, is an immunogenic approach based on the prospective isolation of a CSC using cell surface antigens, which are frequently adapted from hematopoietic stem cells. The second approach utilizes characteristics that CSCs share with normal stem cells, such as sphere forming ability, chemoresistance, self-renewal, asymmetric division, and “stemness“ gene signatures. Both approaches have been successfully used to isolate CSCs in liver cancer (Table [1]). All isolated cells showed essential cancer stem cell characteristics, such as self-renewal, multipotency, and extensive proliferation capacity, regardless of using functional and/or antigenic-based isolation procedures. Consistent with the CSC hypothesis and in agreement with observations from other types of tumor, the liver CSCs comprise only a minor highly aggressive subpopulation within an original tumor.[68] [69] [70] [71] [72]

Table 1 Commonly Used Markers to Identify Liver Cancer Stem Cells (CSCs)
Marker/Method Frequency Minimal Cell Number Required for Tumor Initiatiation Other Solid Tumors Reference (for Liver CSC)
SP 0–28% 1000 Lung, breast, pancreas 69,78,82
CD133 0–65% 1000 Breast, colon, pancreas, endometrium, prostate, brain 70
CD133/CD44 0.1–1.9 100 Colon, breast 83
CD133/ALDH1 0–56% 500 Colon, breast 73
CD90 0–2.5% 500 Prostate stroma 72,84
EpCAM 0–99% 200 Breast, pancreas, colon 71
SP, side population; EpCAM, epithelial cell adhesion molecule.

Currently, there exist limited genomic data on prospectively isolated putative liver CSCs. The transmembrane protein Prominin1 (CD133) appears to be useful for isolation of liver CSCs.[70] Available gene expression data of CD133+ cells show activation of stemness genes involved in Wnt/β-catenin, NOTCH, and Hedgehog signaling pathways.[73] Another frequently used CSC marker for isolating liver CSCs is EpCAM, also known as ESA and TACSTD1.[71] [74] [75] As mentioned above, classification of HCC patients based on the EpCAM expression has a prognostic significance. HCC classified as a hepatic progenitor cell subtype (HpSC) showed activation of the stem cell and self-renewal pathways, including TGF-β, Wnt/β-catenin, PI3K/Akt, and expressed stem/progenitor cell and CSC markers, such as KRT19 (CK19), TACSTD1 (EpCAM), AFP, DKK1, DLK1, and PROM1 (CD133). Conversely, the mature hepatocyte subtype (MH) displayed the activation of the gene sets associated with the hepatocyte functions, such as metabolism, complement system, and coagulation.[54] [71] Furthermore, the prospectively isolated EpCAM+ cells exhibited an adult stem cell-like gene expression profile as demonstrated by cluster analysis based on the TaqMan human stem cell pluripotency array, whereas EpCAM cells were closer to MH subtype HCC.

Another technique to isolate cells with CSC properties is based on the capacity to exclude the vital dye Hoechst 33342, which defines the so-called side population (SP).[76] Gene expression analysis by reverse transcription polymerase chain reaction (RT-PCR) performed on the SP cells showed that the genes important for stemness, differentiation, as well as chemoresistance were differentially expressed as compared with the corresponding non-SP.[68] [69] Subsequent work has demonstrated that BMI1, a part of the Polycomb group complex PRC1 (protein regulator of cytokinesis) known for its contribution to stem cell self-renewal, has a critical role in maintaining and propagating SP population in liver cancer, suggesting the potential stem cell origin of CSC.[77] [78] [79] Gene expression profiling of SP cells from two different HCC cell lines further showed activation of oncogenic and stemness pathways, such as Wnt and Jun signaling.

Our transcriptomic analysis of epigenetically modified SP cells using the DNMT1-inhibitor Zebularine provides further evidence for a potential stem cell origin of liver CSC (our unpublished data).[80] Signaling pathways important for developmental processes, such as Wnt/β-catenin and IL6, and stem/progenitor cell markers, including CK19, SOX9, SOX4, DMBT1, MED12, and AMD1, were differentially expressed in the isolated SP cells. Additionally, gene set enrichment analysis (GSEA) revealed strong enrichment of gene sets important both for the tissue stem cells and embryonic stem cells. Consistent with the findings from the module map, integrative analysis of the generated CSC signature showed a clear association with poorly differentiated tumors and a bad prognosis of HCC patients.

Although the available data on genomics of prospectively isolated liver CSCs are still limited and overlap between gene expression data and antigenic characteristics from different studies is low, the findings discussed above emphasize the common patterns supporting the existence and importance of CSCs in human HCC. Furthermore, the prognostic impact of the CSC gene expression signatures may suggest a paradigm shift from the traditional therapies to those aimed at eradicating CSCs.[81] Taken together, the existing evidence from microarray-generated molecular classifications of HCC suggests a potential stem cell origin in a subpopulation of human HCC and underlines prognostic implications of the prospective analysis of putative cancer stem cells. Because the heterogeneity of human liver cancer disease may be related to CSC origin, the hepatic CSC cannot be defined by the expression of a single marker. A combination of different functional and antigenic approaches should be used for CSC identification and characterization (Fig. [3]).

Zoom Image

Figure 3 Integrative genomic analysis and its implication for cancer cell research (CSC). Genomic and epigenomic analysis of putative CSC can be used for cross comparison and validation using independent data from different sources (cell culture, human patients, and mouse models). The data can then be integrated and subjected to different analysis to test whether the isolated cells fulfill CSC criteria, i.e., stemness activation of relevant signaling pathways and gene modules. After validation, CSC can be then used for detailed characterization of the key molecular changes, identification of the cellular origin, and stratification of cancer patients. The obtained data can be used to identify new therapeutic targets to specifically eliminate CSC. HCC, hepatocellular cell carcinoma.

#

ACKNOWLEDGMENTS

Dr. Marquardt thanks Dr. Levy Kopelovich for his help and great support. We thank Dr. Valentina M. Factor for critical review and helpful advice with the manuscript, and the members of the Laboratory of Experimental Carcinogenesis for their valuable contribution and input to this work.

#

ABBREVIATIONS

  • AFP α-fetoprotein

  • CK cytokeratin

  • C-Kit encodes a proto-oncogenic receptor tyrosine kinase

  • CSC cancer stem cell

  • ESC embryonic stem cell

  • EGF epidermal growth factor

  • EpCAM epithelial cell adhesion molecule

  • FAH fumarylacetoacetate hydrolase

  • GSEA gene set enrichment analysis

  • HB hepatoblast

  • HCC hepatocellular carcinoma

  • HpSC hepatic progenitor cell

  • IL interleukin

  • MH mature hepatocyte

  • MET MET proto-oncogene (hepatocyte growth factor receptor)

  • PRC protein regulator of cytokinesis

  • RT-PCR Reverse transcription polymerase chain reaction

  • SP side population

  • TGF transforming growth factor

#

REFERENCES

  • 1 Bergsagel D E, Valeriote F A. Growth characteristics of a mouse plasma cell tumor.  Cancer Res. 1968;  28(11) 2187-2196
  • 2 Heppner G H. Tumor heterogeneity.  Cancer Res. 1984;  44(6) 2259-2265
  • 3 Weisenthal L M, Lippman M E. Clonogenic and nonclonogenic in vitro chemosensitivity assays.  Cancer Treat Rep. 1985;  69(6) 615-632
  • 4 Hewitt H B. Studies of the dissemination and quantitative transplantation of a lymphocytic leukaemia of CBA mice.  Br J Cancer. 1958;  12(3) 378-401
  • 5 Southam C M, Brunschwig A. Quantitative studies of autotransplantation of human cancer. Preliminary report.  Cancer. 1961;  14(5) 971-978
  • 6 Hamburger A W, Salmon S E. Primary bioassay of human tumor stem cells.  Science. 1977;  197(4302) 461-463
  • 7 Reya T, Morrison S J, Clarke M F, Weissman I L. Stem cells, cancer, and cancer stem cells.  Nature. 2001;  414(6859) 105-111
  • 8 Bonnet D, Dick J E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.  Nat Med. 1997;  3(7) 730-737
  • 9 Campbell L L, Polyak K. Breast tumor heterogeneity: cancer stem cells or clonal evolution?.  Cell Cycle. 2007;  6(19) 2332-2338
  • 10 Nowell P C. The clonal evolution of tumor cell populations.  Science. 1976;  194(4260) 23-28
  • 11 Clarke M F, Dick J E, Dirks P B et al.. Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells.  Cancer Res. 2006;  66(19) 9339-9344
  • 12 Jordan C T, Guzman M L, Noble M. Cancer stem cells.  N Engl J Med. 2006;  355(12) 1253-1261
  • 13 Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance.  Nat Rev Cancer. 2005;  5(4) 275-284
  • 14 Ganguly R, Puri I K. Mathematical model for chemotherapeutic drug efficacy in arresting tumour growth based on the cancer stem cell hypothesis.  Cell Prolif. 2007;  40(3) 338-354
  • 15 Zajicek G. Resistance to cancer chemotherapy.  Med Hypotheses. 1986;  19(2) 103-112
  • 16 Polyak K, Weinberg R A. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits.  Nat Rev Cancer. 2009;  9(4) 265-273
  • 17 Farazi P A, DePinho R A. Hepatocellular carcinoma pathogenesis: from genes to environment.  Nat Rev Cancer. 2006;  6(9) 674-687
  • 18 Ramaswamy S. Translating cancer genomics into clinical oncology.  N Engl J Med. 2004;  350(18) 1814-1816
  • 19 Couinaud C. Le Foie: Études Anatomiques et Chirurgicales. Paris; Masson et Cie 1957: 187-208
  • 20 Desmet V J. Organizational principles in the liver: biology and pathobiology. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Shafritz DA The Liver: Biology and Pathobiology. 3rd ed. New York; Raven Press 1994: 3-14
  • 21 Duncan A W, Dorrell C, Grompe M. Stem cells and liver regeneration.  Gastroenterology. 2009;  137(2) 466-481
  • 22 Overturf K, al-Dhalimy M, Ou C N, Finegold M, Grompe M. Serial transplantation reveals the stem-cell-like regenerative potential of adult mouse hepatocytes.  Am J Pathol. 1997;  151(5) 1273-1280
  • 23 Roskams T. Liver stem cells and their implication in hepatocellular and cholangiocarcinoma.  Oncogene. 2006;  25(27) 3818-3822
  • 24 Farber E. Carcinoma of the liver in rats fed ethionine.  AMA Arch Pathol. 1956;  62(6) 445-453
  • 25 Opie E L. The pathogenesis of tumors of the liver produced by butter yellow.  J Exp Med. 1944;  80(3) 231-246
  • 26 Evarts R P, Nagy P, Marsden E, Thorgeirsson S S. A precursor-product relationship exists between oval cells and hepatocytes in rat liver.  Carcinogenesis. 1987;  8(11) 1737-1740
  • 27 Popper H, Schaffner F. The liver and its diseases.  Gastroenterology. 1961;  40 536-552
  • 28 Ruben L N, Balls M. Genetic disparity and cancer induction by normal tissue implants in amphibia.  Science. 1964;  146 1321-1322
  • 29 Roskams T, Desmet V. Ductular reaction and its diagnostic significance.  Semin Diagn Pathol. 1998;  15(4) 259-269
  • 30 Hsia C C, Thorgeirsson S S, Tabor E. Expression of hepatitis B surface and core antigens and transforming growth factor-alpha in “oval cells” of the liver in patients with hepatocellular carcinoma.  J Med Virol. 1994;  43(3) 216-221
  • 31 Hsia C C, Evarts R P, Nakatsukasa H, Marsden E R, Thorgeirsson S S. Occurrence of oval-type cells in hepatitis B virus-associated human hepatocarcinogenesis.  Hepatology. 1992;  16(6) 1327-1333
  • 32 Thorgeirsson S S, Grisham J W. Hematopoietic cells as hepatocyte stem cells: a critical review of the evidence.  Hepatology. 2006;  43(1) 2-8
  • 33 Fiegel H C, Lioznov M V, Cortes-Dericks L et al.. Liver-specific gene expression in cultured human hematopoietic stem cells.  Stem Cells. 2003;  21(1) 98-104
  • 34 Fukuda K, Sugihara A, Nakasho K et al.. The origin of biliary ductular cells that appear in the spleen after transplantation of hepatocytes.  Cell Transplant. 2004;  13(1) 27-33
  • 35 Minguet S, Cortegano I, Gonzalo P et al.. A population of c-Kit(low)(CD45/TER119)- hepatic cell progenitors of 11-day postcoitus mouse embryo liver reconstitutes cell-depleted liver organoids.  J Clin Invest. 2003;  112(8) 1152-1163
  • 36 Lagasse E, Connors H, Al-Dhalimy M et al.. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo.  Nat Med. 2000;  6(11) 1229-1234
  • 37 Willenbring H, Bailey A S, Foster M et al.. Myelomonocytic cells are sufficient for therapeutic cell fusion in liver.  Nat Med. 2004;  10(7) 744-748
  • 38 Coleman W B, Wennerberg A E, Smith G J, Grisham J W. Regulation of the differentiation of diploid and some aneuploid rat liver epithelial (stemlike) cells by the hepatic microenvironment.  Am J Pathol. 1993;  142(5) 1373-1382
  • 39 Dumble M L, Croager E J, Yeoh G C, Quail E A. Generation and characterization of p53 null transformed hepatic progenitor cells: oval cells give rise to hepatocellular carcinoma.  Carcinogenesis. 2002;  23(3) 435-445
  • 40 Garfield S, Huber B E, Nagy P, Cordingley M G, Thorgeirsson S S. Neoplastic transformation and lineage switching of rat liver epithelial cells by retrovirus-associated oncogenes.  Mol Carcinog. 1988;  1(3) 189-195
  • 41 Tsao M S, Grisham J W. Hepatocarcinomas, cholangiocarcinomas, and hepatoblastomas produced by chemically transformed cultured rat liver epithelial cells. A light- and electron-microscopic analysis.  Am J Pathol. 1987;  127(1) 168-181
  • 42 Braun L, Mikumo R, Fausto N. Production of hepatocellular carcinoma by oval cells: cell cycle expression of c-myc and p53 at different stages of oval cell transformation.  Cancer Res. 1989;  49(6) 1554-1561
  • 43 Libbrecht L, Desmet V, Van Damme B, Roskams T. Deep intralobular extension of human hepatic “progenitor cells” correlates with parenchymal inflammation in chronic viral hepatitis: can “progenitor cells” migrate?.  J Pathol. 2000;  192(3) 373-378
  • 44 Libbrecht L, Desmet V, Van Damme B, Roskams T. The immunohistochemical phenotype of dysplastic foci in human liver: correlation with putative progenitor cells.  J Hepatol. 2000;  33(1) 76-84
  • 45 Lowes K N, Brennan B A, Yeoh G C, Olynyk J K. Oval cell numbers in human chronic liver diseases are directly related to disease severity.  Am J Pathol. 1999;  154(2) 537-541
  • 46 Durnez A, Verslype C, Nevens F et al.. The clinicopathological and prognostic relevance of cytokeratin 7 and 19 expression in hepatocellular carcinoma. A possible progenitor cell origin.  Histopathology. 2006;  49(2) 138-151
  • 47 Wu P C, Lai V C, Fang J W, Gerber M A, Lai C L, Lau J Y. Hepatocellular carcinoma expressing both hepatocellular and biliary markers also expresses cytokeratin 14, a marker of bipotential progenitor cells.  J Hepatol. 1999;  31(5) 965-966
  • 48 Andersen J B, Loi R, Perra A et al.. Progenitor-derived hepatocellular carcinoma model in the rat.  Hepatology. 2009;  , December 4 (Epub ahead of print)
  • 49 Tang Y, Kitisin K, Jogunoori W et al.. Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling.  Proc Natl Acad Sci U S A. 2008;  105(7) 2445-2450
  • 50 Knight B, Tirnitz-Parker J E, Olynyk J K. C-kit inhibition by imatinib mesylate attenuates progenitor cell expansion and inhibits liver tumor formation in mice.  Gastroenterology. 2008;  135(3) 969-979, 979, e1
  • 51 Lee J S, Thorgeirsson S S. Genetic profiling of human hepatocellular carcinoma.  Semin Liver Dis. 2005;  25(2) 125-132
  • 52 Iizuka N, Oka M, Yamada-Okabe H et al.. Oligonucleotide microarray for prediction of early intrahepatic recurrence of hepatocellular carcinoma after curative resection.  Lancet. 2003;  361(9361) 923-929
  • 53 Lee J S, Chu I S, Heo J et al.. Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling.  Hepatology. 2004;  40(3) 667-676
  • 54 Yamashita T, Forgues M, Wang W et al.. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma.  Cancer Res. 2008;  68(5) 1451-1461
  • 55 Ye Q H, Qin L X, Forgues M et al.. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning.  Nat Med. 2003;  9(4) 416-423
  • 56 Santoni-Rugiu E, Jelnes P, Thorgeirsson S S, Bisgaard H C. Progenitor cells in liver regeneration: molecular responses controlling their activation and expansion.  APMIS. 2005;  113(11-12) 876-902
  • 57 Yang W, Yan H X, Chen L et al.. Wnt/beta-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells.  Cancer Res. 2008;  68(11) 4287-4295
  • 58 Hoshida Y, Nijman S M, Kobayashi M et al.. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma.  Cancer Res. 2009;  69(18) 7385-7392
  • 59 El-Serag H B, Rudolph K L. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis.  Gastroenterology. 2007;  132(7) 2557-2576
  • 60 Villanueva A, Newell P, Chiang D Y, Friedman S L, Llovet J M. Genomics and signaling pathways in hepatocellular carcinoma.  Semin Liver Dis. 2007;  27(1) 55-76
  • 61 Segal E, Friedman N, Koller D, Regev A. A module map showing conditional activity of expression modules in cancer.  Nat Genet. 2004;  36(10) 1090-1098
  • 62 Wong D J, Liu H, Ridky T W, Cassarino D, Segal E, Chang H Y. Module map of stem cell genes guides creation of epithelial cancer stem cells.  Cell Stem Cell. 2008;  2(4) 333-344
  • 63 Lee J S, Chu I S, Mikaelyan A et al.. Application of comparative functional genomics to identify best-fit mouse models to study human cancer.  Nat Genet. 2004;  36(12) 1306-1311
  • 64 Shachaf C M, Kopelman A M, Arvanitis C et al.. MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer.  Nature. 2004;  431(7012) 1112-1117
  • 65 Kaposi-Novak P, Libbrecht L, Woo H G et al.. Central role of c-Myc during malignant conversion in human hepatocarcinogenesis.  Cancer Res. 2009;  69(7) 2775-2782
  • 66 Lee J S, Thorgeirsson S S. Comparative and integrative functional genomics of HCC.  Oncogene. 2006;  25(27) 3801-3809
  • 67 Lee J S, Heo J, Libbrecht L et al.. A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells.  Nat Med. 2006;  12(4) 410-416
  • 68 Chiba T, Kita K, Zheng Y W et al.. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties.  Hepatology. 2006;  44(1) 240-251
  • 69 Haraguchi N, Utsunomiya T, Inoue H et al.. Characterization of a side population of cancer cells from human gastrointestinal system.  Stem Cells. 2006;  24(3) 506-513
  • 70 Ma S, Chan K W, Hu L et al.. Identification and characterization of tumorigenic liver cancer stem/progenitor cells.  Gastroenterology. 2007;  132(7) 2542-2556
  • 71 Yamashita T, Ji J, Budhu A et al.. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features.  Gastroenterology. 2009;  136(3) 1012-1024
  • 72 Yang Z F, Ho D W, Ng M N et al.. Significance of CD90 + cancer stem cells in human liver cancer.  Cancer Cell. 2008;  13(2) 153-166
  • 73 Ma S, Chan K W, Lee T K et al.. Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations.  Mol Cancer Res. 2008;  6(7) 1146-1153
  • 74 Munz M, Baeuerle P A, Gires O. The emerging role of EpCAM in cancer and stem cell signaling.  Cancer Res. 2009;  69(14) 5627-5629
  • 75 Shmelkov S V, Butler J M, Hooper A T et al.. CD133 expression is not restricted to stem cells, and both CD133 + and CD133- metastatic colon cancer cells initiate tumors.  J Clin Invest. 2008;  118(6) 2111-2120
  • 76 Goodell M A, Brose K, Paradis G, Conner A S, Mulligan R C. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.  J Exp Med. 1996;  183(4) 1797-1806
  • 77 Chiba T, Zheng Y W, Kita K et al.. Enhanced self-renewal capability in hepatic stem/progenitor cells drives cancer initiation.  Gastroenterology. 2007;  133(3) 937-950
  • 78 Chiba T, Miyagi S, Saraya A et al.. The polycomb gene product BMI1 contributes to the maintenance of tumor-initiating side population cells in hepatocellular carcinoma.  Cancer Res. 2008;  68(19) 7742-7749
  • 79 Park I K, Qian D, Kiel M et al.. BMI-1 is required for maintenance of adult self-renewing haematopoietic stem cells.  Nature. 2003;  423(6937) 302-305
  • 80 Marquardt J U, Raggi C, Andersen J B et al.. Gene expression signature of putative cancer stem cells predicts survival of HCC patients. Paper presented at: 61st AASLD Annual Meeting October 29–November 2, 2009 Boston, MA;
  • 81 Klonisch T, Wiechec E, Hombach-Klonisch S et al.. Cancer stem cell markers in common cancers - therapeutic implications.  Trends Mol Med. 2008;  14(10) 450-460
  • 82 Shi G M, Xu Y, Fan J et al.. Identification of side population cells in human hepatocellular carcinoma cell lines with stepwise metastatic potentials.  J Cancer Res Clin Oncol. 2008;  134(11) 1155-1163
  • 83 Zhu Z, Hao X, Yan M et al.. Cancer stem/progenitor cells are highly enriched in CD133(+)CD44(+) population in hepatocellular carcinoma.  Int J Cancer. 2009;  , August 26 (Epub ahead of print)
  • 84 Yang Z F, Ngai P, Ho D W et al.. Identification of local and circulating cancer stem cells in human liver cancer.  Hepatology. 2008;  47(3) 919-928

Snorri S ThorgeirssonM.D. Ph.D. 

Laboratory of Experimental Carcinogenesis (LEC), Center for Cancer Research

National Cancer Institute, NIH, 37 Convent Drive, Room 4146, Bethesda, MD 20892

Email: snorri_thorgeirsson@nih.gov

#

REFERENCES

  • 1 Bergsagel D E, Valeriote F A. Growth characteristics of a mouse plasma cell tumor.  Cancer Res. 1968;  28(11) 2187-2196
  • 2 Heppner G H. Tumor heterogeneity.  Cancer Res. 1984;  44(6) 2259-2265
  • 3 Weisenthal L M, Lippman M E. Clonogenic and nonclonogenic in vitro chemosensitivity assays.  Cancer Treat Rep. 1985;  69(6) 615-632
  • 4 Hewitt H B. Studies of the dissemination and quantitative transplantation of a lymphocytic leukaemia of CBA mice.  Br J Cancer. 1958;  12(3) 378-401
  • 5 Southam C M, Brunschwig A. Quantitative studies of autotransplantation of human cancer. Preliminary report.  Cancer. 1961;  14(5) 971-978
  • 6 Hamburger A W, Salmon S E. Primary bioassay of human tumor stem cells.  Science. 1977;  197(4302) 461-463
  • 7 Reya T, Morrison S J, Clarke M F, Weissman I L. Stem cells, cancer, and cancer stem cells.  Nature. 2001;  414(6859) 105-111
  • 8 Bonnet D, Dick J E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.  Nat Med. 1997;  3(7) 730-737
  • 9 Campbell L L, Polyak K. Breast tumor heterogeneity: cancer stem cells or clonal evolution?.  Cell Cycle. 2007;  6(19) 2332-2338
  • 10 Nowell P C. The clonal evolution of tumor cell populations.  Science. 1976;  194(4260) 23-28
  • 11 Clarke M F, Dick J E, Dirks P B et al.. Cancer stem cells–perspectives on current status and future directions: AACR Workshop on cancer stem cells.  Cancer Res. 2006;  66(19) 9339-9344
  • 12 Jordan C T, Guzman M L, Noble M. Cancer stem cells.  N Engl J Med. 2006;  355(12) 1253-1261
  • 13 Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance.  Nat Rev Cancer. 2005;  5(4) 275-284
  • 14 Ganguly R, Puri I K. Mathematical model for chemotherapeutic drug efficacy in arresting tumour growth based on the cancer stem cell hypothesis.  Cell Prolif. 2007;  40(3) 338-354
  • 15 Zajicek G. Resistance to cancer chemotherapy.  Med Hypotheses. 1986;  19(2) 103-112
  • 16 Polyak K, Weinberg R A. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits.  Nat Rev Cancer. 2009;  9(4) 265-273
  • 17 Farazi P A, DePinho R A. Hepatocellular carcinoma pathogenesis: from genes to environment.  Nat Rev Cancer. 2006;  6(9) 674-687
  • 18 Ramaswamy S. Translating cancer genomics into clinical oncology.  N Engl J Med. 2004;  350(18) 1814-1816
  • 19 Couinaud C. Le Foie: Études Anatomiques et Chirurgicales. Paris; Masson et Cie 1957: 187-208
  • 20 Desmet V J. Organizational principles in the liver: biology and pathobiology. In: Arias IM, Boyer JL, Fausto N, Jakoby WB, Shafritz DA The Liver: Biology and Pathobiology. 3rd ed. New York; Raven Press 1994: 3-14
  • 21 Duncan A W, Dorrell C, Grompe M. Stem cells and liver regeneration.  Gastroenterology. 2009;  137(2) 466-481
  • 22 Overturf K, al-Dhalimy M, Ou C N, Finegold M, Grompe M. Serial transplantation reveals the stem-cell-like regenerative potential of adult mouse hepatocytes.  Am J Pathol. 1997;  151(5) 1273-1280
  • 23 Roskams T. Liver stem cells and their implication in hepatocellular and cholangiocarcinoma.  Oncogene. 2006;  25(27) 3818-3822
  • 24 Farber E. Carcinoma of the liver in rats fed ethionine.  AMA Arch Pathol. 1956;  62(6) 445-453
  • 25 Opie E L. The pathogenesis of tumors of the liver produced by butter yellow.  J Exp Med. 1944;  80(3) 231-246
  • 26 Evarts R P, Nagy P, Marsden E, Thorgeirsson S S. A precursor-product relationship exists between oval cells and hepatocytes in rat liver.  Carcinogenesis. 1987;  8(11) 1737-1740
  • 27 Popper H, Schaffner F. The liver and its diseases.  Gastroenterology. 1961;  40 536-552
  • 28 Ruben L N, Balls M. Genetic disparity and cancer induction by normal tissue implants in amphibia.  Science. 1964;  146 1321-1322
  • 29 Roskams T, Desmet V. Ductular reaction and its diagnostic significance.  Semin Diagn Pathol. 1998;  15(4) 259-269
  • 30 Hsia C C, Thorgeirsson S S, Tabor E. Expression of hepatitis B surface and core antigens and transforming growth factor-alpha in “oval cells” of the liver in patients with hepatocellular carcinoma.  J Med Virol. 1994;  43(3) 216-221
  • 31 Hsia C C, Evarts R P, Nakatsukasa H, Marsden E R, Thorgeirsson S S. Occurrence of oval-type cells in hepatitis B virus-associated human hepatocarcinogenesis.  Hepatology. 1992;  16(6) 1327-1333
  • 32 Thorgeirsson S S, Grisham J W. Hematopoietic cells as hepatocyte stem cells: a critical review of the evidence.  Hepatology. 2006;  43(1) 2-8
  • 33 Fiegel H C, Lioznov M V, Cortes-Dericks L et al.. Liver-specific gene expression in cultured human hematopoietic stem cells.  Stem Cells. 2003;  21(1) 98-104
  • 34 Fukuda K, Sugihara A, Nakasho K et al.. The origin of biliary ductular cells that appear in the spleen after transplantation of hepatocytes.  Cell Transplant. 2004;  13(1) 27-33
  • 35 Minguet S, Cortegano I, Gonzalo P et al.. A population of c-Kit(low)(CD45/TER119)- hepatic cell progenitors of 11-day postcoitus mouse embryo liver reconstitutes cell-depleted liver organoids.  J Clin Invest. 2003;  112(8) 1152-1163
  • 36 Lagasse E, Connors H, Al-Dhalimy M et al.. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo.  Nat Med. 2000;  6(11) 1229-1234
  • 37 Willenbring H, Bailey A S, Foster M et al.. Myelomonocytic cells are sufficient for therapeutic cell fusion in liver.  Nat Med. 2004;  10(7) 744-748
  • 38 Coleman W B, Wennerberg A E, Smith G J, Grisham J W. Regulation of the differentiation of diploid and some aneuploid rat liver epithelial (stemlike) cells by the hepatic microenvironment.  Am J Pathol. 1993;  142(5) 1373-1382
  • 39 Dumble M L, Croager E J, Yeoh G C, Quail E A. Generation and characterization of p53 null transformed hepatic progenitor cells: oval cells give rise to hepatocellular carcinoma.  Carcinogenesis. 2002;  23(3) 435-445
  • 40 Garfield S, Huber B E, Nagy P, Cordingley M G, Thorgeirsson S S. Neoplastic transformation and lineage switching of rat liver epithelial cells by retrovirus-associated oncogenes.  Mol Carcinog. 1988;  1(3) 189-195
  • 41 Tsao M S, Grisham J W. Hepatocarcinomas, cholangiocarcinomas, and hepatoblastomas produced by chemically transformed cultured rat liver epithelial cells. A light- and electron-microscopic analysis.  Am J Pathol. 1987;  127(1) 168-181
  • 42 Braun L, Mikumo R, Fausto N. Production of hepatocellular carcinoma by oval cells: cell cycle expression of c-myc and p53 at different stages of oval cell transformation.  Cancer Res. 1989;  49(6) 1554-1561
  • 43 Libbrecht L, Desmet V, Van Damme B, Roskams T. Deep intralobular extension of human hepatic “progenitor cells” correlates with parenchymal inflammation in chronic viral hepatitis: can “progenitor cells” migrate?.  J Pathol. 2000;  192(3) 373-378
  • 44 Libbrecht L, Desmet V, Van Damme B, Roskams T. The immunohistochemical phenotype of dysplastic foci in human liver: correlation with putative progenitor cells.  J Hepatol. 2000;  33(1) 76-84
  • 45 Lowes K N, Brennan B A, Yeoh G C, Olynyk J K. Oval cell numbers in human chronic liver diseases are directly related to disease severity.  Am J Pathol. 1999;  154(2) 537-541
  • 46 Durnez A, Verslype C, Nevens F et al.. The clinicopathological and prognostic relevance of cytokeratin 7 and 19 expression in hepatocellular carcinoma. A possible progenitor cell origin.  Histopathology. 2006;  49(2) 138-151
  • 47 Wu P C, Lai V C, Fang J W, Gerber M A, Lai C L, Lau J Y. Hepatocellular carcinoma expressing both hepatocellular and biliary markers also expresses cytokeratin 14, a marker of bipotential progenitor cells.  J Hepatol. 1999;  31(5) 965-966
  • 48 Andersen J B, Loi R, Perra A et al.. Progenitor-derived hepatocellular carcinoma model in the rat.  Hepatology. 2009;  , December 4 (Epub ahead of print)
  • 49 Tang Y, Kitisin K, Jogunoori W et al.. Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling.  Proc Natl Acad Sci U S A. 2008;  105(7) 2445-2450
  • 50 Knight B, Tirnitz-Parker J E, Olynyk J K. C-kit inhibition by imatinib mesylate attenuates progenitor cell expansion and inhibits liver tumor formation in mice.  Gastroenterology. 2008;  135(3) 969-979, 979, e1
  • 51 Lee J S, Thorgeirsson S S. Genetic profiling of human hepatocellular carcinoma.  Semin Liver Dis. 2005;  25(2) 125-132
  • 52 Iizuka N, Oka M, Yamada-Okabe H et al.. Oligonucleotide microarray for prediction of early intrahepatic recurrence of hepatocellular carcinoma after curative resection.  Lancet. 2003;  361(9361) 923-929
  • 53 Lee J S, Chu I S, Heo J et al.. Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling.  Hepatology. 2004;  40(3) 667-676
  • 54 Yamashita T, Forgues M, Wang W et al.. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma.  Cancer Res. 2008;  68(5) 1451-1461
  • 55 Ye Q H, Qin L X, Forgues M et al.. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning.  Nat Med. 2003;  9(4) 416-423
  • 56 Santoni-Rugiu E, Jelnes P, Thorgeirsson S S, Bisgaard H C. Progenitor cells in liver regeneration: molecular responses controlling their activation and expansion.  APMIS. 2005;  113(11-12) 876-902
  • 57 Yang W, Yan H X, Chen L et al.. Wnt/beta-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells.  Cancer Res. 2008;  68(11) 4287-4295
  • 58 Hoshida Y, Nijman S M, Kobayashi M et al.. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma.  Cancer Res. 2009;  69(18) 7385-7392
  • 59 El-Serag H B, Rudolph K L. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis.  Gastroenterology. 2007;  132(7) 2557-2576
  • 60 Villanueva A, Newell P, Chiang D Y, Friedman S L, Llovet J M. Genomics and signaling pathways in hepatocellular carcinoma.  Semin Liver Dis. 2007;  27(1) 55-76
  • 61 Segal E, Friedman N, Koller D, Regev A. A module map showing conditional activity of expression modules in cancer.  Nat Genet. 2004;  36(10) 1090-1098
  • 62 Wong D J, Liu H, Ridky T W, Cassarino D, Segal E, Chang H Y. Module map of stem cell genes guides creation of epithelial cancer stem cells.  Cell Stem Cell. 2008;  2(4) 333-344
  • 63 Lee J S, Chu I S, Mikaelyan A et al.. Application of comparative functional genomics to identify best-fit mouse models to study human cancer.  Nat Genet. 2004;  36(12) 1306-1311
  • 64 Shachaf C M, Kopelman A M, Arvanitis C et al.. MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer.  Nature. 2004;  431(7012) 1112-1117
  • 65 Kaposi-Novak P, Libbrecht L, Woo H G et al.. Central role of c-Myc during malignant conversion in human hepatocarcinogenesis.  Cancer Res. 2009;  69(7) 2775-2782
  • 66 Lee J S, Thorgeirsson S S. Comparative and integrative functional genomics of HCC.  Oncogene. 2006;  25(27) 3801-3809
  • 67 Lee J S, Heo J, Libbrecht L et al.. A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells.  Nat Med. 2006;  12(4) 410-416
  • 68 Chiba T, Kita K, Zheng Y W et al.. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties.  Hepatology. 2006;  44(1) 240-251
  • 69 Haraguchi N, Utsunomiya T, Inoue H et al.. Characterization of a side population of cancer cells from human gastrointestinal system.  Stem Cells. 2006;  24(3) 506-513
  • 70 Ma S, Chan K W, Hu L et al.. Identification and characterization of tumorigenic liver cancer stem/progenitor cells.  Gastroenterology. 2007;  132(7) 2542-2556
  • 71 Yamashita T, Ji J, Budhu A et al.. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features.  Gastroenterology. 2009;  136(3) 1012-1024
  • 72 Yang Z F, Ho D W, Ng M N et al.. Significance of CD90 + cancer stem cells in human liver cancer.  Cancer Cell. 2008;  13(2) 153-166
  • 73 Ma S, Chan K W, Lee T K et al.. Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations.  Mol Cancer Res. 2008;  6(7) 1146-1153
  • 74 Munz M, Baeuerle P A, Gires O. The emerging role of EpCAM in cancer and stem cell signaling.  Cancer Res. 2009;  69(14) 5627-5629
  • 75 Shmelkov S V, Butler J M, Hooper A T et al.. CD133 expression is not restricted to stem cells, and both CD133 + and CD133- metastatic colon cancer cells initiate tumors.  J Clin Invest. 2008;  118(6) 2111-2120
  • 76 Goodell M A, Brose K, Paradis G, Conner A S, Mulligan R C. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.  J Exp Med. 1996;  183(4) 1797-1806
  • 77 Chiba T, Zheng Y W, Kita K et al.. Enhanced self-renewal capability in hepatic stem/progenitor cells drives cancer initiation.  Gastroenterology. 2007;  133(3) 937-950
  • 78 Chiba T, Miyagi S, Saraya A et al.. The polycomb gene product BMI1 contributes to the maintenance of tumor-initiating side population cells in hepatocellular carcinoma.  Cancer Res. 2008;  68(19) 7742-7749
  • 79 Park I K, Qian D, Kiel M et al.. BMI-1 is required for maintenance of adult self-renewing haematopoietic stem cells.  Nature. 2003;  423(6937) 302-305
  • 80 Marquardt J U, Raggi C, Andersen J B et al.. Gene expression signature of putative cancer stem cells predicts survival of HCC patients. Paper presented at: 61st AASLD Annual Meeting October 29–November 2, 2009 Boston, MA;
  • 81 Klonisch T, Wiechec E, Hombach-Klonisch S et al.. Cancer stem cell markers in common cancers - therapeutic implications.  Trends Mol Med. 2008;  14(10) 450-460
  • 82 Shi G M, Xu Y, Fan J et al.. Identification of side population cells in human hepatocellular carcinoma cell lines with stepwise metastatic potentials.  J Cancer Res Clin Oncol. 2008;  134(11) 1155-1163
  • 83 Zhu Z, Hao X, Yan M et al.. Cancer stem/progenitor cells are highly enriched in CD133(+)CD44(+) population in hepatocellular carcinoma.  Int J Cancer. 2009;  , August 26 (Epub ahead of print)
  • 84 Yang Z F, Ngai P, Ho D W et al.. Identification of local and circulating cancer stem cells in human liver cancer.  Hepatology. 2008;  47(3) 919-928

Snorri S ThorgeirssonM.D. Ph.D. 

Laboratory of Experimental Carcinogenesis (LEC), Center for Cancer Research

National Cancer Institute, NIH, 37 Convent Drive, Room 4146, Bethesda, MD 20892

Email: snorri_thorgeirsson@nih.gov

Zoom Image

Figure 1 Potential cell types of origin of liver cancer stem cells (CSCs). The scheme illustrates the cellular organization of a portal triad consisting of a portal vein (PV), hepatic artery (A), and bile duct (BD). The most terminal branches of the biliary tree are referred to as Canals of Hering. CSCs can originate from the stem/progenitor cells thought to reside in Canals of Hering. They extensively proliferate upon adequate stimuli commonly present in hepatocarcinogenesis, for example, liver damage. CSCs can also originate from mature hepatocytes known to possess stem-like characteristics, such as proliferation capacity and longevity. Hepatocytes are continuously exposed to exogenous and endogenous stress and could be targets for transforming events. Lastly, bone marrow-derived stem cells could be a source of hepatic CSC through transdifferentiation.

Zoom Image

Figure 2 Genomic evidence for a progenitor cell origin in liver cancer. The comparative functional genomic analysis of 139 human hepatocellular carcinomas (HCCs) and rat hepatoblasts/ hepatocytes identified 22 human HCCs that showed similar expression pattern to stem-like hepatoblasts (HBs; designated as HB type) (adapted from Lee et al[67]). Kaplan-Meier plots showed that patients with HB subtype HCC had worse survival and a higher frequency of recurrence. The study highlights the potential stem cell origin in a subtype of HCC and demonstrates clinical relevance of this finding.

Zoom Image

Figure 3 Integrative genomic analysis and its implication for cancer cell research (CSC). Genomic and epigenomic analysis of putative CSC can be used for cross comparison and validation using independent data from different sources (cell culture, human patients, and mouse models). The data can then be integrated and subjected to different analysis to test whether the isolated cells fulfill CSC criteria, i.e., stemness activation of relevant signaling pathways and gene modules. After validation, CSC can be then used for detailed characterization of the key molecular changes, identification of the cellular origin, and stratification of cancer patients. The obtained data can be used to identify new therapeutic targets to specifically eliminate CSC. HCC, hepatocellular cell carcinoma.