Cancer stem cell

Cancer stem cells (CSCs) are cancer cells (found within tumors or hematological cancers) that possess characteristics associated with normal stem cells, specifically the ability to give rise to all cell types found in a particular cancer sample. CSCs are therefore tumorigenic (tumor-forming), perhaps in contrast to other non-tumorigenic cancer cells. CSCs may generate tumors through the stem cell processes of self-renewal and differentiation into multiple cell types. Such cells are proposed to persist in tumors as a distinct population and cause relapse and metastasis by giving rise to new tumors. Therefore, development of specific therapies targeted at CSCs holds hope for improvement of survival and quality of life of cancer patients, especially for sufferers of metastatic disease.

Existing cancer treatments have mostly been developed based on animal models, where therapies able to promote tumor shrinkage were deemed effective. However, animals could not provide a complete model of human disease. In particular, in mice, whose life spans do not exceed two years, tumor relapse is exceptionally difficult to study.

The efficacy of cancer treatments is, in the initial stages of testing, often measured by the ablation fraction of tumor mass (fractional kill). As CSCs would form a very small proportion of the tumor, this may not necessarily select for drugs that act specifically on the stem cells. The theory suggests that conventional chemotherapies kill differentiated or differentiating cells, which form the bulk of the tumor but are unable to generate new cells. A population of CSCs, which gave rise to it, could remain untouched and cause a relapse of the disease.

Contents

Evidence

The existence of CSCs is a subject of debate within medical research, because many studies have not been successful in discovering the similarities and differences between normal tissue stem cells and cancer stem cells.[1] Cancer cells must be capable of continuous proliferation and self-renewal in order to retain the many mutations required for carcinogenesis, and to sustain the growth of a tumor since differentiated cells (constrained by the Hayflick Limit) cannot divide indefinitely . However, it is debated whether such cells represent a minority. If most cells of the tumor are endowed with stem cell properties, there is no incentive to focus on a specific sub population. There is also debate on the cell of origin of CSCs - whether they originate from stem cells that have lost the ability to regulate proliferation, or from more differentiated population of progenitor cells that have acquired abilities to self-renew (which is related to the issue of stem cell plasticity).

The first conclusive evidence for CSCs was published in 1997 in Nature Medicine. Bonnet and Dick[2] isolated a subpopulation of leukaemic cells that express a specific surface marker CD34, but lacks the CD38 marker. The authors established that the CD34+/CD38- subpopulation is capable of initiating tumors in NOD/SCID mice that is histologically similar to the donor.

In cancer research experiments, tumor cells are sometimes injected into an experimental animal to establish a tumor. Disease progression is then followed in time and novel drugs can be tested for their ability to inhibit it. However, efficient tumor formation requires thousands or tens of thousands of cells to be introduced. Classically, this has been explained by poor methodology (i.e. the tumor cells lose their viability during transfer) or the critical importance of the microenvironment, the particular biochemical surroundings of the injected cells. Supporters of the cancer stem cell paradigm argue that only a small fraction of the injected cells, the CSCs, have the potential to generate a tumor. In human acute myeloid leukemia the frequency of these cells is less than 1 in 10,000.[2]

Further evidence comes from histology, the study of the tissue structure of tumors. Many tumors are very heterogeneous and contain multiple cell types native to the host organ. Heterogeneity is commonly retained by tumor metastases. This implies that the cell that produced them had the capacity to generate multiple cell types. In other words, it possessed multidifferentiative potential, a classical hallmark of stem cells.[2]

The existence of leukaemic stem cells prompted further research into other types of cancer. CSCs have recently been identified in several solid tumors, including cancers of the:

Importance

Not only is finding the source of cancer cells necessary for successful treatments, but if current treatments of cancer do not properly destroy enough CSCs, the tumor will reappear. Including the possibility that the treatment of for instance, chemotherapy, will leave only chemotherapy-resistant CSCs, then the ensuing tumor will most likely also be resistant to chemotherapy. If the cancer tumor is detected early enough, enough of the tumor can be killed off and marginalized with traditional treatment. But as the tumor size increases, it becomes more and more difficult to remove the tumor without conferring resistance and leaving enough behind for the tumor to reappear.

Some treatments with chemotherapy, such as paclitaxel in ovarian cancer (a cancer usually discovered in late stages), may actually induce chemoresistance (55-75% relapse <2 years[16]). It potentially does this by destroying only the cancer cells susceptible to the drug (targeting those that are CD44-positive, a trait which has been associated with increased survival time in some ovarian cancers), and allowing the cells which are unaffected by paclitaxel (CD44-negative) to regrow, even after a reduction in over a third of the total tumor size.[17] There are studies, though, which show how paclitaxel can be used in combination with other ligands to affect the CD44-positive cells.[18] While paclitaxel alone, as of late, does not cure the cancer, it is effective at extending the survival time of the patients.[16]

Mechanistic and mathematical models

Once the pathways to cancer are hypothesized, it is possible to develop predictive mathematical biology models,[19] e.g., based on the cell compartment method. For instance, the growths of the abnormal cells from their normal counterparts can be denoted with specific mutation probabilities. Such a model has been employed to predict that repeated insult to mature cells increases the formation of abnormal progeny, and hence the risk of cancer.[20] Considerable work needs to be done, however, before the clinical efficacy of such models[21] is established.

Origins

The origin of cancer stem cells is still an area of ongoing research. Several camps have formed within the scientific community regarding the issue, and it is possible that several answers are correct, depending on the tumor type and the phenotype the tumor presents. One important distinction that will often be raised is that the cell of origin for a tumor can not be demonstrated using the cancer stem cell as a model. This is because cancer stem cells are isolated from end-stage tumors. Therefore, describing a cancer stem cell as a cell of origin is often an inaccurate claim, even though a cancer stem cell is capable of initiating new tumor formation.

With that caveat mentioned, various theories define the origin of cancer stem cells. In brief, they may be: mutants in developing stem or progenitor cells, mutants in adult stem cells or adult progenitor cells, or mutant cells that acquire stem like attributes. These theories often do focus on a tumor's cell of origin and as such must be approached with skepticism.

Some researchers favor the theory that the cancer stem cell is caused by a mutation in stem cell niche populations during development. The logical progression claims that these developing stem populations are mutated and then expand such that the mutation is shared by many of the descendants of the mutated stem cell. These daughter stem cells are then much closer to becoming tumors, and since there are many of them there is more chance of a mutation that can cause cancer.[22]

Another theory associates adult stem cells with the formation of tumors. This is most often associated with tissues with a high rate of cell turnover (such as the skin or gut). In these tissues, it has long been expected that stem cells are responsible for tumor formation. This is a consequence of the frequent cell divisions of these stem cells (compared to most adult stem cells) in conjunction with the extremely long lifespan of adult stem cells. This combination creates the ideal set of circumstances for mutations to accumulate; accumulation of mutations is the primary factor that drives cancer initiation. In spite of the logical backing of the theory, only recently has evidence appeared that this association represents an actual phenomenon. It is important to bear in mind that, due to the heterogeneous nature of evidence it is possible that any individual cancer could come from an alternative origin.

A third possibility often raised is the potential de-differentiation of mutated cells such that these cells acquire stem cell like characteristics. This is often used as a potential alternative to any specific cell of origin, as it suggests that any cell might become a cancer stem cell.

Another related concept is the concept of tumor hierarchy. This concept claims that a tumor is a heterogeneous population of mutant cells, all of which share some mutations but will vary in specific phenotype. In this model, the tumor is made up of several types of stem cells, one optimal to the specific environment and several less successful lines. These secondary lines can become more successful in some environments, allowing the tumor to adapt to its environment, including the methods by which tumors can be treated. If this situation is accurate, it has severe repercussions on the realism of a cancer stem cell specific treatment regime.[23] Within a tumor hierarchy model, it would be extremely difficult to pinpoint the cancer stem cell's origin.

Implications for cancer treatment

The existence of CSCs has several implications in terms of future cancer treatment and therapies. These include disease identification, selective drug targets, prevention of metastasis, and development of new intervention strategies.

Normal somatic stem cells are naturally resistant to chemotherapeutic agents- they have various pumps (such as MDR) that pump out drugs, DNA repair proteins and they also have a slow rate of cell turnover (chemotherapeutic agents naturally target rapidly replicating cells). CSCs that have mutated from normal stem cells may also express proteins that would increase their resistance towards chemotherapeutic agents. These surviving CSCs then repopulate the tumor, causing relapse. By selectively targeting CSCs, it would be possible to treat patients with aggressive, non-resectable tumors, as well as preventing the tumor from metastasizing. The hypothesis suggests that upon CSC elimination, cancer would regress due to differentiation and/or cell death. What fraction of tumor cells are CSCs and therefore need to be eliminated is not clear yet.[24]

A number of studies have investigated the possibility of identifying specific markers that may distinguish CSCs from the bulk of the tumor (as well as from normal stem cells).[4] Proteomic and genomic signatures of tumors are also being investigated . In 2009, scientists identified one compound, Salinomycin, that selectively reduces the proportion of breast CSCs in mice by more than 100-fold relative to Paclitaxel, a commonly used chemotherapeutic agent.[25]

Pathways

The design of new drugs for the treatment of CSCs will likely require an understanding of the cellular mechanisms that regulate cell proliferation. The first advances in this area were made with hematopoietic stem cells (HSCs) and their transformed counterparts in leukemia, the disease for which the origin of CSCs is best understood. It is now becoming increasingly clear that stem cells of many organs share the same cellular pathways as leukemia-derived HSCs.

Additionally, a normal stem cell may be transformed into a cancer stem cell through disregulation of the proliferation and differentiation pathways controlling it or by inducing oncoprotein activity.

Bmi-1

The Polycomb group transcriptional repressor Bmi-1 was discovered as a common oncogene activated in lymphoma[26] and later shown to specifically regulate HSCs.[27] The role of Bmi-1 has also been illustrated in neural stem cells.[28] The pathway appears to be active in CSCs of pediatric brain tumors.[29]

Notch

The Notch pathway has been known to developmental biologists for decades. Its role in control of stem cell proliferation has now been demonstrated for several cell types including hematopoietic, neural and mammary[30] stem cells. Components of the Notch pathway have been proposed to act as oncogenes in mammary[31] and other tumors.

Sonic hedgehog and Wnt

These developmental pathways are also strongly implicated as stem cell regulators.[32] Both Sonic hedgehog (SHH) and Wnt pathways are commonly hyperactivated in tumors and are required to sustain tumor growth. However, the Gli transcription factors that are regulated by SHH take their name from gliomas, where they are commonly expressed at high levels. A degree of crosstalk exists between the two pathways and their activation commonly goes hand-in-hand.[33] This is a trend rather than a rule. For instance, in colon cancer hedgehog signalling appears to antagonise Wnt.[34]

Sonic hedgehog blockers are available, such as cyclopamine. There is also a new water soluble cyclopamine that may be more effective in cancer treatment. There is also DMAPT, a water soluble derivative of parthenolide (induces oxidative stress, inhibits NF-κB signaling[35]) for AML (leukemia), and possibly myeloma and prostate cancer. A clinical trial of DMAPT is to start in England in late 2007 or 2008. Finally, the enzyme telomerase may qualify as a study subject in CSC physiology.[36]; GRN163L (Imetelstat) was recently started in trials to target myeloma stem cells. If it is possible to eliminate the cancer stem cell, then a potential cure may be achieved if there are no more CSCs to repopulate a cancer.

References

  1. ^ Gupta PB, Chaffer CL, Weinberg RA (2009). "Cancer stem cells: mirage or reality?". Nat Med 15 (9): 1010–2. doi:10.1038/nm0909-1010. PMID 19734877. 
  2. ^ a b c Bonnet D, Dick JE (July 1997). "Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell". Nature medicine 3 (7): 730–7. doi:10.1038/nm0797-730. PMID 9212098. 
  3. ^ Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB (September 2003). "Identification of a cancer stem cell in human brain tumors". Cancer research 63 (18): 5821–8. PMID 14522905. http://cancerres.aacrjournals.org/cgi/content/abstract/63/18/5821. 
  4. ^ a b Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (April 2003). "Prospective identification of tumorigenic breast cancer cells". Proceedings of the National Academy of Sciences of the United States of America 100 (7): 3983–8. doi:10.1073/pnas.0530291100. PMC 153034. PMID 12629218. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=153034. 
  5. ^ O'Brien CA, Pollett A, Gallinger S, Dick JE (January 2007). "A human colon cancer cell capable of initiating tumour growth in immunodeficient mice". Nature 445 (7123): 106–10. doi:10.1038/nature05372. PMID 17122772. 
  6. ^ Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH, Nephew KP (June 2008). "Identification and characterization of ovarian cancer-initiating cells from primary human tumors". Cancer research 68 (11): 4311–20. doi:10.1158/0008-5472.CAN-08-0364. PMC 2553722. PMID 18519691. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2553722. 
  7. ^ Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM (February 2007). "Identification of pancreatic cancer stem cells". Cancer research 67 (3): 1030–7. doi:10.1158/0008-5472.CAN-06-2030. PMID 17283135. 
  8. ^ Maitland NJ, Collins AT (June 2008). "Prostate cancer stem cells: a new target for therapy". J. Clin. Oncol. 26 (17): 2862–70. doi:10.1200/JCO.2007.15.1472. PMID 18539965. 
  9. ^ Lang Sh, Frame F, Collins A (January 2009). "Prostate cancer stem cells". J. Pathol. 217 (2): 299–306. doi:10.1002/path.2478. PMC 2673349. PMID 19040209. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2673349. 
  10. ^ Schatton T, Murphy GF, Frank, NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, Fuhlbrigge RC, Kupper TS, Sayegh MH, Frank MH (Jan. 2008). "Identification of cells initiating human melanomas". Nature 451 (7176): 345–9. doi:10.1038/nature06489. PMID 18202660. http://www.nature.com/nature/journal/v451/n7176/full/nature06489.html. 
  11. ^ Boiko AD, Razorenova OV, van de Rijn M, Swetter SM, Johnson DL, Ly DP, Butler PD, Yang GP, Joshua B, Kaplan MJ, Longaker MT, Weissman IL (Jul. 2010). "Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271". Nature 466 (7302): 133–7.. doi:10.1038/nature09161. PMC 2898751. PMID 20596026. http://www.nature.com/nature/journal/v466/n7302/full/nature09161.html. 
  12. ^ Schmidt P, Kopecky C, Hombach A, Zigrino P, Mauch C, Abken H. (Feb 2011). "Eradication of melanomas by targeted elimination of a minor subset of tumor cells.". PNAS 108 (6): 2474–9. doi:10.1073/pnas.1009069108. PMC 3038763. PMID 21282657. http://www.pnas.org.ezp-prod1.hul.harvard.edu/content/108/6/2474.long. 
  13. ^ Civenni G, Walter A, Kobert N, Mihic-Probst D, Zipser M, Belloni B, Seifert B, Moch H, Dummer R, van den Broek M, Sommer L. (Mar. 2011). "Human CD271-Positive Melanoma Stem Cells Associated with Metastasis Establish Tumor Heterogeneity and Long-Term Growth". Cancer Res. 71 (8): 3098–109. doi:10.1158/0008-5472.CAN-10-3997. PMID 21393506. http://cancerres.aacrjournals.org/content/early/2011/03/09/0008-5472.CAN-10-3997.long. 
  14. ^ Matsui W, Huff CA, Wang Q, et al. (March 2004). "Characterization of clonogenic multiple myeloma cells". Blood 103 (6): 2332–6. doi:10.1182/blood-2003-09-3064. PMID 14630803. 
  15. ^ Matsui W, Wang Q, Barber JP, et al. (January 2008). "Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance". Cancer Res. 68 (1): 190–7. doi:10.1158/0008-5472.CAN-07-3096. PMC 2603142. PMID 18172311. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2603142. 
  16. ^ a b England's National Institute of Health and Clinical Excellence. Full guidance on the use of paclitaxel in the treatment of ovarian cancer, 22 January 2003.
  17. ^ Mor, G. Yale Medical School. Presentation of unpublished data at University of Missour, Kansas City Medical School. Jan 7, 2008.
  18. ^ Auzenne E, Ghosh SC, Khodadadian M, Rivera B, Farquhar D, Price RE, Ravoori M, Kundra V, Freedman RS, Klostergaard J (June 2007). "Hyaluronic acid-paclitaxel: antitumor efficacy against CD44(+) human ovarian carcinoma xenografts". Neoplasia 9 (6): 479–86. PMC 1899257. PMID 17603630. http://www.neoplasia.com/abstract.php?msid=1127. 
  19. ^ Preziosi, Luigi (2003). Cancer Modelling and Simulation. Boca Raton: CRC Press. ISBN 1-58488-361-8. 
  20. ^ Ganguly R, Puri IK (February 2006). "Mathematical model for the cancer stem cell hypothesis". Cell proliferation 39 (1): 3–14. doi:10.1111/j.1365-2184.2006.00369.x. PMID 16426418. 
  21. ^ Ganguly R, Puri IK (June 2007). "Mathematical model for chemotherapeutic drug efficacy in arresting tumour growth based on the cancer stem cell hypothesis". Cell proliferation 40 (3): 338–354. doi:10.1111/j.1365-2184.2007.00434.x. PMID 17531079. 
  22. ^ Wang Y, Yang J, Zheng H, Tomasek GJ, Zhang P, McKeever PE, Lee EY, Zhu Y (June 2009). "Expression of mutant p53 proteins implicates a lineage relationship between neural stem cells and malignant astrocytic glioma in a murine model". Cancer Cell 15 (6): 514–26. doi:10.1016/j.ccr.2009.04.001. PMC 2721466. PMID 19477430. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2721466. 
  23. ^ Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM (October 2006). "Cancer stem cells--perspectives on current status and future directions: AACR Workshop on cancer stem cells". Cancer research 66 (19): 9339–44. doi:10.1158/0008-5472.CAN-06-3126. PMID 16990346. 
  24. ^ "Cancer stem cells: Invitation to a second round". http://www.nature.com/nature/journal/v466/n7302/full/466040a.html. 
  25. ^ "Cell - Identification of Selective Inhibitors of Cancer Stem Cells by High-Throughput Screening". http://www.cell.com/abstract/S0092-8674(09)00781-8. 
  26. ^ Haupt Y, Bath ML, Harris AW, Adams JM (November 1993). "bmi-1 transgene induces lymphomas and collaborates with myc in tumorigenesis". Oncogene 8 (11): 3161–4. PMID 8414519. 
  27. ^ Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, Morrison SJ, Clarke MF (May 2003). "Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells". Nature 423 (6937): 302–5. doi:10.1038/nature01587. PMID 12714971. 
  28. ^ Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ (October 2003). "Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation". Nature 425 (6961): 962–7. doi:10.1038/nature02060. PMC 2614897. PMID 14574365. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=2614897. 
  29. ^ Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI (December 2003). "Cancerous stem cells can arise from pediatric brain tumors". Proceedings of the National Academy of Sciences of the United States of America 100 (25): 15178–83. doi:10.1073/pnas.2036535100. PMC 299944. PMID 14645703. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=299944. 
  30. ^ Dontu G, Jackson KW, McNicholas E, Kawamura MJ, Abdallah WM, Wicha MS (2004). "Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells". Breast cancer research : BCR 6 (6): R605–15. doi:10.1186/bcr920. PMC 1064073. PMID 15535842. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=1064073. 
  31. ^ Diévart A, Beaulieu N, Jolicoeur P (October 1999). "Involvement of Notch1 in the development of mouse mammary tumors". Oncogene 18 (44): 5973–81. doi:10.1038/sj.onc.1202991. PMID 10557086. 
  32. ^ Beachy PA, Karhadkar SS, Berman DM (November 2004). "Tissue repair and stem cell renewal in carcinogenesis". Nature 432 (7015): 324–31. doi:10.1038/nature03100. PMID 15549094. 
  33. ^ Zhou BP, Hung MC (June 2005). "Wnt, hedgehog and snail: sister pathways that control by GSK-3beta and beta-Trcp in the regulation of metastasis". Cell cycle (Georgetown, Tex.) 4 (6): 772–6. doi:10.4161/cc.4.6.1744. PMID 15917668. http://www.landesbioscience.com/journals/cc/abstract.php?id=1744. 
  34. ^ Akiyoshi T, Nakamura M, Koga K, Nakashima H, Yao T, Tsuneyoshi M, Tanaka M, Katano M (July 2006). "Gli1, downregulated in colorectal cancers, inhibits proliferation of colon cancer cells involving Wnt signalling activation". Gut 55 (7): 991–9. doi:10.1136/gut.2005.080333. PMC 1856354. PMID 16299030. http://www.pubmedcentral.nih.gov/articlerender.fcgi?tool=pmcentrez&artid=1856354. 
  35. ^ She M, Chen X (2009). "Targeting signal pathways active in cancer stem cells to overcome drug resistance". Chin J Lung Cancer 12 (1): 3–7. doi:10.3779/j.issn.1009-3419.2009.01.001. PMID 20712949. http://www.lungca.org/index.php?journal=01&page=article&op=view&path%5B%5D=10.3779%2Fj.issn.1009-3419.2009.01.001. 
  36. ^ Bollmann FM (August 2008). "The many faces of telomerase: emerging extratelomeric effects". BioEssays 30 (8): 728–32. doi:10.1002/bies.20793. PMID 18623070. 

Further reading

External links