Cancer Stem Cells: A Step Toward the Cure

Dr. Weeks’ Comment:  Dr Wicha is a brilliant pioneer in the work to remedy cancer STEM cells. If your oncologist is not focusing on cancer STEM cells,  he or she is NOT addressing the really lethal aspect of cancer.

 

Cancer Stem Cells: A Step Toward the Cure

 

.    Bruce M. Boman   Helen F. Graham Cancer Center, Newark, DE; Thomas Jefferson University, Philadelphia, PA

.    Max S. Wicha

SOURCE:              2008 by American Society of Clinical Oncology

 

The goal of this article is to introduce general concepts of CSC biology to clinicians, and to provide a framework for their understanding the CSC-based approach to the development of novel diagnostics, therapeutics, and prevention strategies in oncology.

This new approach differs from the traditional one because the latter is based on the long-held concept that tumor formation and growth are due to increased proliferation of cells in cancers compared with normal tissues. This traditional view is supported by molecular oncology research during the last 50 years, which showed that tumors undergo a series of genetic events (mutations) that result in the activation or overexpression of genes promoting proliferation (eg, oncogenes), the silencing of genes involved in inhibition of proliferation (eg, tumor suppressor genes), and the development of the ability of cancer cells to elude apoptosis. The consequence is the development and unchecked growth of tumors and their progression to metastases. Based on this traditional concept of tumor growth, it has been disappointing that therapies designed to kill proliferating cells often fail to cure cancer patients.

Furthermore, during tumor development, tissues accumulate a series of mutations over years or even decades, but, because of tissue renewal, most cells in tissues are lost or eliminated in just a brief time, typically days or weeks, and with their loss, any mutations they have acquired would also be lost. Thus, new mechanisms need to be considered to explain human tumorigenesis.

One explanation is that tumorigenic mutations occur in cells that are few in number but reside long term in tissues. But that is not sufficient to explain tumorigenesis. To be truly tumorigenic, this population of rare, mutated cells would have to self-renew, clonally expand, and acquire additional mutations. It is now widely believed that these long-lived, uncommon cells are tissue stem cells (SCs) or cells derived from them that acquire the ability to self-renew. When mutated, they can become CSCs. Both normal SCs and CSCs may account for only a small fraction of cells (∼1%) in any given tissue or tumor. By definition, SCs are self-renewing. Mounting evidence presented in the articles in this special issue suggests that CSCs initiate and drive tumor growth.

This is nothing less than a paradigm shift.1 It is a shift to the view that, first, cancers originate in tissue stem or progenitor cells through dysregulation of the self-renewal process. Second, throughout tumorigenesis, CSCs drive tumor growth. Third, current chemotherapeutic agents and radiation therapy largely target proliferating and differentiated cells that form the bulk of the tumor but not the relatively quiescent CSCs. Also, SCs are drug resistant and CSCs appear to be so too. Thus, this quiescence and resistance may account for many treatment failures. Fourth, if this is the case, then the only effective way to treat cancer is to target the CSC population.

Although this view may seem novel, the basic idea that SCs are the cells of clonal origin of malignancies has existed for at least several decades,2 if not longer,3 particularly for leukemias48 and teratomas.911 Early attempts””beginning in the 1970s””to directly measure SCs in tumors used several approaches.12 One was mouse-spleen colony assays (eg, injection of lymphoma cells into mice with the appearance of lymphoma colonies in the spleen). A second was the counting of metastatic lung colonies after intravenous inoculation of tumor cells into mice. A third was tumor colony formation in vitro. However, this latter method produced mixed results for human tumors.13 SCs have also been known for decades to radiation biologists.14,15 More recent evidence, as presented in the articles in this special issue, supports an SC origin for solid tumors as well. In the following paragraphs, we discuss the properties of normal SCs and CSCs, and their possible roles in carcinogenesis and in mediating tumor behavior.

 

BIOLOGIC PROPERTIES OF SCs

The most well-known property of SC populations is their ability to self-renew. Self-renewal is particularly important in tissues with high turnover, such as the GI tract and bone marrow, as well as in tissue repair after injury. Self-renewal involves the ability of normal SC populations to precisely maintain their numbers through a combination of symmetric and asymmetric SC division.16,17 The property of self-renewal is also a hallmark of populations of CSCs. However, in this case, mechanisms involved in self-renewal are dysregulated and seem to lead to CSC overpopulation. The underlying mechanisms for replacing lost SCs (eg, during tissue repair) or in generating excess SC numbers (eg, in tumor development) are believed to relate to increases in symmetric SC division (which produces two SC daughters) relative to asymmetric SC division (which produces one SC daughter and one non-SC daughter). Indeed, we documented this concept quantitatively for colon cancer development using mathematical modeling.17 We showed that only increased symmetric division of cancer SCs could account for the biologic observation that there is an exponential increase in SCs, non-SC proliferative cells, and nonproliferative cell populations in the development of colorectal cancer. Our modeling also showed that only increased symmetric SC division could account for the known long lag phase in colon cancer development, which is typical in the development of many cancers.

Although SCs have the capacity for self-renewal, in fact they are relatively quiescent; that is, they have proliferative capacity but are often not cycling. Indeed, they have been shown to have significantly longer cell cycle times than proliferating non-SCs.18 This is presumably due to the arrest of SCs at a G0-like cell cycle phase or checkpoint.

Another property of SCs is their potential for multilineage differentiation. This is consistent with the CSC hypothesis because cancers typically contain multiple differentiated cell types that correspond to cells in the tissue from which the cancer arose. This suggests a SC origin for these tumors because only multipotent SCs (or perhaps SC-like progenitor cells) could have given rise to these different cell types. Indeed, it has been known for more than a century that tumors are composed of heterogeneous populations of partially differentiated cell types that resemble those in the normal organ.19 This observation gave rise to the idea that tumors are functioning as aberrantly developed complex organs.20 This is consistent with the concept that CSCs, like normal SCs, give rise to a hierarchical organization of cell populations that underlie organogenesis.21 This hierarchy includes CSCs that produce committed progenitor cells that, in turn, produce rapidly proliferating cells, finally resulting in the generation of fully differentiated cells (Table 1). This is discussed further in this issue in the articles by Sell and Leffert22 on liver CSCs and by Lee et al23 on pancreatic CSCs.

The development of various organs during embryogenesis is known to involve specific signaling pathways. Key pathways include sonic hedgehog, Notch, PTEN, BMI-1, WNT, and p53. Indeed, there is growing evidence that these signaling pathways regulate self-renewal of normal SC populations.16 These pathways are known to become dysregulated during development of tumors, which is believed to lead to dysregulation of SC self-renewal and contribute to neoplastic proliferation.21

 

METHODS TO IDENTIFY AND ISOLATE SCs

Progress in the field of SC biology, including CSC biology, has been long hampered by difficulties in identifying, isolating, and characterizing SCs. However, considerable progress has been made recently. At first, SC populations could only be identified functionally using their ability to undergo self-renewal and differentiation into multiple lineages. More recently, CSCs and even normal SCs from certain tissues have been identified and isolated. This became possible in large part due to the identification of SC markers (Table 2), their use in combination with fluorescence-activated cell sorting, and the ability to show that specific cell subpopulations form tumors in xenograft assays in immunocompromised mice.4 The CSCs and the tumors they form demonstrate the capacity for serial passage (transplantability) in immunocompromised mice and for generation of multiple cell lineages in a histopathologic pattern resembling the original tumor. This approach has been used to isolate CSCs from various cancer types (Table 2), as discussed in the corresponding articles in this issue. Interestingly, many CSC markers are shared across multiple tumor types. They also may be expressed on normal tissue SCs in organs from which these tumors originated.

CSCs have also been identified by in vitro sphere-forming assays.24 It has been demonstrated that the ability of cells to form colonies in multicellular spheroids or spherical cell aggregates under nonadherent culture conditions is a characteristic of many cells having self-renewal capacity. These spheroids can be disaggregated and passaged multiple times with retention of sphere-forming ability. Spheroid formation in vitro was used to study CSCs in breast (see article in this issue by Kakarala and Wicha25) and brain (see article in this issue by Dirks26). The practicality, ease, and low cost of this assay makes it adaptable for performing high-throughput screens for identifying drugs that target CSCs and for measuring the effects of radiation on CSCs. Targets can be validated using more labor-intensive xenograft models.

 

ROLE OF CSCs IN CANCER INITIATION, PRIMARY TUMOR GROWTH, AND METASTASES

Our current understanding of human carcinogenesis involves two central ideas. One is that most, if not all, cancers result from the accumulation of specific genetic mutations. The other is that tumorigenesis involves aberrant tissue organization and cell proliferation. The CSC paradigm begins to suggest how these two events are linked. That is, it provides a cellular mechanism that can explain how the genetic and epigenetic changes give rise to the tissue changes. Moreover, evidence suggests that these mechanisms may be operational in all phases of tumorigenesis: initiation, progression, invasion, and metastasis. The idea that cancer initiation involves development of CSC overpopulation was supported by an analysis of tissues from hereditary cancer patients.27,28 For fully developed cancers, support comes from recent reports that many cancers contain a small but significant proportion (∼1%) of CSCs.2935 Given that the total number of cells is exponentially increased (up to 108 to 1013 cells) in fully developed cancers, it can be deduced that the size of the SC subpopulation in those tumors is also exponentially increased. Indeed, this must have occurred during all phases of tumorigenesis.17 The progressive increase in the CSC population throughout tumorigenesis in the colon will be discussed in this issue in the article by Boman and Huang36 on colon cancer SCs. Regarding metastases, there is a growing body of evidence that suggests that metastases develop when distant organs are seeded with CSCs that arise from a primary tumor.37 This implicates CSCs in the seeding and growth of metastatic lesions. This mechanism is discussed, in this issue, in the article on breast CSCs by Kakarala and Wicha.25

 

CSCs IN CANCER DIAGNOSIS, TREATMENT, AND PREVENTION

As noted, current systemic cancer therapies frequently fail to eradicate advanced tumors. Failure of these therapies to effectively target cancer SCs may account for this failure.38 First, most current agents destroy rapidly proliferating cells””cells that represent the bulk of tumor cell populations. If cancer SCs drive tumorigenesis, current chemotherapeutic approaches may be ineffective because cancer SCs, like all SCs, are relatively quiescent; that is, they proliferate only infrequently. Second, SCs tend to be more resistant to chemotherapeutic agents and radiation than are more mature cell types from the same tissue. This is believed to be due to the presence of multidrug resistance, antiapoptotic proteins, and enhanced DNA repair mechanisms. Since this appears to hold true for cancer SCs, we would predict that they would also display resistance to therapeutics. This is discussed, in this issue, in the article by Eyler and Rich.39 Third, most current therapies do not target the signaling pathways that regulate self-renewal, which appear to be either mutated or epigenetically dysregulated. These pathways, including sonic hedgehog, Notch, PTEN, BMI-1, WNT, and p53, are discussed in this issue in the article by Fan and Eberhart.40 There is evidence that these pathways are involved in regulation of self-renewal, and effective therapies will need to target symmetric SC division.17 Targeting these pathways, it should be noted, will probably not be enough””an ideal agent would have to selectively target cancer SCs over normal SCs. Without this selectivity, the effectiveness of treatment might be limited by systemic toxicity.

Control of SC numbers would also be important in cancer chemoprevention. The ideal cancer chemopreventive agent would also selectively target CSCs over normal SCs. This suggests that the development of agents that eliminate or control CSC populations may be an effective strategy for cancer prevention as well as the development of more effective cancer therapies. Indeed, experiments using antibodies against an CSC marker, CD44, eliminated human acute myeloid leukemic SCs in immunodeficient mice transplanted with human acute myeloid leukemia.41

It is also likely that treatment of patients with CSC-targeted therapeutics will require new clinical end points for monitoring therapeutic efficacy. This is the case because one is targeting only a small fraction of cells within the tumor, not the bulk of the tumor. In addition, responses may take a much longer time than is typically seen with agents that are cytotoxic to the rapidly dividing cells that constitute the bulk of the tumor. Rational approaches might also include using cytotoxic chemotherapies to target the proliferating bulk of the tumor in addition to a CSC-directed therapy directed at eliminating this critical cell population. An important end point will likely involve assessing changes in the size of the CSC population in response to treatment. One way to do this might be by monitoring the burden of CSCs in the circulation. Of course patient survival remains the ultimate clinical end point, which will involve appropriate clinical trials.

Markers for CSCs could be used for predicting treatment responses by identifying the presence of specific CSC subtypes that are selectively sensitive to specific agents, particularly biologic agents such as antibodies against specific CSC targets. Microarray and proteomic profiling of CSCs will likely lead to identification of new markers, as well as potential therapeutic targets. Indeed, a recent microarray study identified a genetic signature associated with the BMI-1 driven SC self-renewal pathway.42 Other gene expression studies have identified genes associated with prognosis in breast cancer, and these signatures contained the aldehyde dehydrogenase 1 (ALDH1) SC marker.43,44 This approach to identify new markers is discussed, in this issue, in the article by Glinsky.45 CSC markers may have prognostic value by allowing assessment of the size of the CSC population within any given tumor. Early evidence suggests that the greater the size of the CSC population, the poorer the prognosis, as reflected by tumor aggressiveness and/or patient survival. This is discussed, in this issue, in the article by Kakarala and Wicha.25

 

MATHEMATICAL MODELING OF CSC DYNAMICS AND THERAPEUTIC APPROACHES

Mathematical modeling will be especially useful in understanding the role of CSCs in cancer development and growth, as well as response to therapy. For example, modeling provided a quantitative basis for the ideas that CSCs drive tumor growth, curative anticancer therapy must eradicate or control CSC populations,28,46 and that future therapeutic approaches need to target symmetric division of CSCs.17

Future modeling studies will be important because SCs are involved in homeostasis of normal tissues, which are complex in their structure and in their dynamics, and which involve many interacting cellular pathways. Moreover, the development of tumors is a complicated biologic process, and this will add to the overall complexity. Indeed, a recent article advocates the use of mathematical studies on SCs as a requisite to advance the field.47

Mathematical modeling will also be required to understand responses to treatment when using anti-CSC therapies, especially in predicting how treatments might affect the dynamics of tumor growth and overall tumor responses to treatment. This is exemplified by recent modeling studies that have provided explanations for the role of CSCs in development of resistance to imatinib therapy in patients with chronic myeloid leukemia.4851 Such modeling will also likely be useful in developing treatment and dose schedules, as well as criteria for monitoring therapeutic responses. The use of mathematical modeling is discussed further throughout this issue, especially in the article by Michor.52

 

OVERTURE TO THIS SPECIAL ISSUE

To fully appreciate the CSC-based approach to cancer treatment, diagnosis, and prevention requires a discussion of different cancer types and the CSCs that drive tumor growth in each type. The rest of this JCO special issue is devoted to that end. This includes 13 articles that focus on the roles SCs may play in each type of malignancy. Together, these articles illustrate the commonality of CSCs in multiple tumor types. This suggests that approaches involving similar techniques and molecular markers may be applicable across tumor types. Furthermore, because pathways of CSC self-renewal and cellular survival also seem to be preserved across different tumor types, strategies designed to target CSCs may also have broad clinical utility. This suggests the need to develop pathway-based rather than tumor type-based approaches to CSC-based therapy. This issue also includes five articles covering select research and clinical topics related to CSCs and clinical oncology, including molecular profiling and biomarkers, mathematical models, chemotherapy resistance, radiation resistance, and imaging approaches. All of these articles provide evidence that SCs or their self-renewing progeny may be the cells of origin of specific cancer types, and demonstrate that significant progress has been made in identifying and characterizing each of those CSC populations. The goal of this issue is to inform clinicians and clinical investigators about progress in the field of CSCs. If the hypotheses central to this emerging field prove to be correct, then they represent an important paradigm shift in our understanding of carcinogenesis. The great promise of this work is that it may lead to more effective strategies for cancer prevention and treatment. Given that CSC-targeting agents are now entering clinical trials, we will soon have an indication as to whether this is the case.

AUTHOR CONTRIBUTIONS

Manuscript writing: Bruce M. Boman, Max S. Wicha

Acknowledgments

We acknowledge the valuable discussions regarding the manuscript of Jeremy Z. Fields, PhD, Director of Scientific Research, CA*TX Inc, Gladwyne, PA.

 

 

REFERENCES

1. ↵ Wicha MS, Liu S, Dontu G: Cancer stem cells: An old idea””A paradigm shift. Cancer Res 66:1883-1890, 2006 Abstract/FREE Full Text

2. ↵ Mackillop WJ, Ciampi A, Till JE, et al: A stem cell model of human tumor growth: Implications for tumor cell clonogenic assays. J Natl Cancer Inst 70:9-16, 1983 Medline

3. ↵ Julius Cohnheim: (1839-1884) experimental pathologist. JAMA 206:1561-1562, 1968 Abstract/FREE Full Text

4. ↵ Lapidot T, Sirard C, Vormoor J, et al: A cell initiating human acute myeloid leukemia after transplantation into SCID mice. Nature 367:645-648, 1994 CrossRefMedline

5. Ailles LE, Gerhard B, Kawagoe H, et al: Blood 94:1761-1772, 1999 Abstract/FREE Full Text

6. Fialkow PJ, Singer JW: Chronic leukemias, in DeVita VT Jr, Hellman S, Rosenberg SA (eds): Cancer: Principles & Practice of Oncology. Philadelphia, PA, JB Lippincott, 1989, pp 1836-1852

7. Greaves MH, Mariri G, Newman RA, et al: Selective expression of the common acute lymphoblastic Leukemia gp100 antigen on immature lymphoid cells and their malignant counterparts. Blood 61:628-639, 1983 Abstract/FREE Full Text

8. ↵ Minden MD: Stem cells in acute leukemia, in Golde DW, Takaku F (eds): Hemopoietic Stem Cells. New York, NY, Marcel Dekker, 1985, pp 274-289

9. ↵ Stevens LC: Origin of testicular teratomas from primordial germ cells. J Natl Cancer Inst 38:549-552, 1967 Medline

  1. Pierce GB, Stevens LC, Nakane PK: Ultrastructural analysis of the early development of teratocarcinomas. J Natl Cancer Inst 39:755-773, 1967 Medline
  2. ↵ Pierce GB, Damjanov I: The pathology of cancer, in McKinnel RG, Parchment RE, Damjanov I, et al (eds): The Biologic Basis of Cancer. New York, NY, Cambridge University Press, 2006, pp 14-50
  3. ↵ Hamburger AW, Salmon SE: Primary bioassay of human tumor stem cells. Science 197:461-463, 1977 Abstract/FREE Full Text
  4. ↵ Selby P, Buick RN, Tannock I: A critical appraisal of the “human tumor stem-cell assay.” N Engl J Med 308:129-134, 1983 Medline
  5. ↵ Kummermehr J, Trott K-R: Tumor stem cells, in Potten CS (ed): Stem Cells. London, United Kingdom, Academic Press, 1997, pp 363-400
  6. ↵ Potten CS, Hendry JH: Stem cells in murine small intestine, in Potten CS (ed): Stem Cells: Their Identification and Characterization. Edinburgh, United Kingdom, Churchill-Livingstone, 1983, pp 155-199
  7. ↵ Molofsky AV, Pardal R, Morrison SJ: Diverse mechanisms regulate stem cell self-renewal. Curr Opin Cell Biol 16:700-707, 2004 CrossRefMedline
  8. ↵ Boman BM, Wicha M, Fields JZ, et al: Symmetric division of cancer stem cells: A key mechanism in tumor growth that should be targeted in future therapeutic approaches. Clin Pharmacol Ther 81:893-898, 2007 CrossRefMedline
  9. ↵ Boman BM, Fields JZ, Cavanaugh KL, et al: How dysregulated colonic crypt dynamics cause stem cell overpopulation and initiate colon cancer. Cancer Res 68:3304-3313, 2008 Abstract/FREE Full Text
  10. ↵ Pierce GB, Nakane PK, Martinez-Hernandez A, et al: Ultrastructural comparison of differentiation of stem cells of murine adenocarcinomas of colon and breast with their normal counterparts. J Natl Cancer Inst 58:1329-1345, 1977 Medline
  11. ↵ Brabletz T, Jung A, Spaderna S, et al: Migrating cancer stem cells: An integrated concept of malignant tumour progression. Nat Rev Cancer 5:744-749, 2005 CrossRefMedline
  12. ↵ Reya T, Morrison SJ, Clarke MF, et al: Stem cells, cancer, and cancer stem cells. Nature 414:105-111, 2001 CrossRefMedline
  13. ↵ Sell S, Leffert HL: Liver cancer stem cells. J Clin Oncol 26:2800-2805, 2008 Abstract/FREE Full Text
  14. ↵ Lee CJ, Dosch J, Simeone DM: Pancreatic cancer stem cells. J Clin Oncol 26:2806-2812, 2008 Abstract/FREE Full Text
  15. ↵ Nicolis SK: Cancer stem cells and “stemness” genes in neuro-oncology. Neurobiol Dis 25:217-229, 2007 CrossRefMedline
  16. ↵ Kakarala M, Wicha MS: Implications of the cancer stem-cell hypothesis for breast cancer prevention and therapy. J Clin Oncol 26:2813-2820, 2008 Abstract/FREE Full Text
  17. ↵ Dirks PB: Brain tumor stem cells: Bringing order to the chaos of brain cancer. J Clin Oncol 26:2916-2924, 2008 Abstract/FREE Full Text
  18. ↵ Boman BM, Walters R, Fields JZ, et al: Colonic crypt changes during adenoma development in familial adenomatous polyposis: Immunohistochemical evidence for expansion of the crypt base cell population. Am J Pathol 165:1489-1498, 2004 Medline
  19. ↵ Boman BM, Fields JZ, Bonham-Carter O, et al: Computer modeling implicates stem cell overproduction in colon cancer initiation. Cancer Res 61:8408-8411, 2001 Abstract/FREE Full Text
  20. ↵ Singh SK, Clarke ID, Terasaki M, et al: Identification of a cancer stem cell in human brain tumors. Cancer Res 63:5821-5828, 2003 Abstract/FREE Full Text
  21. Singh SK, Hawkins C, Clarke ID, et al: Identification of human brain tumour initiating cells. Nature 432:396-401, 2004 CrossRefMedline
  22. Al-Hajj M, Wicha MS, Benito-Hernandez A, et al: Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100:3983-3988, 2003 Abstract/FREE Full Text
  23. O’Brien CA, Pollett A, Gallinger S, et al: A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445:106-110, 2007 CrossRefMedline
  24. Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al: Identification and expansion of human colon-cancer-initiating cells. Nature 445:111-115, 2007 CrossRefMedline
  25. Ginestier C, Hur M, Charafe-Jauffret E, et al: ALDH1 is a marker of normal and malignant breast stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1:555-567, 2007 CrossRefMedline
  26. ↵ Dalerba P, Dylla SJ, Park IK, et al: Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A 104:10158-10163, 2007 Abstract/FREE Full Text
  27. ↵ Boman BM, Huang E: Human colon cancer stem cells: A new paradigm in gastrointestinal oncology. J Clin Oncol 26:2828-2838, 2008 Abstract/FREE Full Text
  28. ↵ Balic M, Lin H, Young L, et al: Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin Cancer Res 12:5615-5621, 2006 Abstract/FREE Full Text
  29. ↵ Pardal R, Clarke MF, Morrison SJ: Applying the principles of stem-cell biology to cancer. Nat Rev Cancer 3:895-902, 2003 CrossRefMedline
  30. ↵ Eyler CE, Rich JN: Survival of the fittest: Cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol 26:2839-2845, 2008 Abstract/FREE Full Text
  31. ↵ Fan X, Eberhart CG: Medulloblastoma stem cells. J Clin Oncol 26:2821-2827, 2008 Abstract/FREE Full Text
  32. ↵ Jin L, Hope KJ, Zhai Q, et al: Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med 12:1167-1174, 2006 CrossRefMedline
  33. ↵ Glinsky GV, Olga Berezovska O, Glinskii AB: Microarray analysis identifies a death from cancer signature predicting therapy failure in patients with multiple types of cancer. J Clin Invest 115:1503-1521, 2005 CrossRefMedline
  34. ↵ Alexe G, Alexe S, Axelrod DE, et al: Breast cancer prognosis by combinatorial analysis of gene expression data. Breast Cancer Res 8:R41, 2006 CrossRefMedline
  35. ↵ van’t Veer LJ, Dai H, van de Vijver, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature 415:530-536, 2002 CrossRefMedline
  36. ↵ Glinsky GV: “Stemness” genomics law governs clinical behavior of human cancer: Implications for decision making in disease management. J Clin Oncol 26:2846-2853, 2008 Abstract/FREE Full Text
  37. ↵ Dingli D, Michor F: Successful therapy must eradicate cancer stem cells. Stem Cells 24:2603-2610, 2006 CrossRefMedline
  38. ↵ Kern S, Shibata D: The fuzzy math of solid tumor stem cells: A perspective? Cancer Res 67:9885-9888, 2007
  39. ↵ Roeder I, Horn M, Glauche I, et al: Dynamics modeling of imatinib-treated chronic myeloid leukemia: Funcational insights and clinical implications. Nat Med 12:1181-1184, 2006 CrossRefMedline
  40. Iwasa Y, Nowak MA, Michor F: Evolution of resistance during clonal expansion. Genetics 172:2557-2566, 2006 Abstract/FREE Full Text
  41. Komarova NL, Wodarz D: Effect of cellular quiescence on the success of targeted CML therapy. PLOS One 10:e990, 2007
  42. ↵ Iwasa Y, Nowak MA, Michor F: Evolution of resistance during clonal expansion. Genetics 172:2257-2566, 2006
  43. ↵ Michor F: Mathematical models of cancer stem cells. J Clin Oncol 26:2854-2861, 2008 Abstract/FREE Full Text

 

Leave a Comment

Your email address will not be published. Required fields are marked *