Tumour heterogeneity

Tumour heterogeneity describes the observation that different tumour cells can show distinct morphological and phenotypic profiles, including cellular morphology, gene expression, metabolism, motility, proliferation, and metastatic potential.[1] This phenomenon occurs both between tumours (inter-tumour heterogeneity) and within tumours (intra-tumour heterogeneity). The heterogeneity of cancer cells introduces significant challenges in designing effective treatment strategies. However, research into understanding and characterizing heterogeneity can allow for a better understanding of the causes and progression of disease. In turn, this has the potential to guide the creation of more refined treatment strategies that incorporate knowledge of heterogeneity to yield higher efficacy.

Tumour heterogeneity has been observed in leukemias,[2] breast,[3] prostate,[4][5][6] colon,[7][8][9] brain,[10] esophagus,[11] head and neck,[12] bladder[13] and gynecological carcinomas,[14] liposarcoma,[15] and multiple myeloma.[16]

Models of Heterogeneity

There are two models used to explain the heterogeneity of tumour cells. These are the cancer stem cell model and the clonal evolution model. The models are not mutually exclusive, and it is believed that they both contribute to heterogeneity in varying amounts across different tumour types.[17]

Ability of cancer cells to form tumours under the cancer stem cell and clonal evolution models of heterogeneity.

Cancer Stem Cells

Main article: Cancer stem cell

The cancer stem cell model asserts that within a population of tumour cells, there is only a small subset of cells that are tumourigenic (able to form tumours). These cells are termed cancer stem cells (CSCs), and are marked by the ability to both self-renew and differentiate into non-tumourigenic progeny. The CSC model posits that the heterogeneity observed between tumour cells is the result of differences in the stem cells from which they originated. Stem cell variability is often caused by epigenetic changes, but can also result from clonal evolution of the CSC population where advantageous genetic mutations can accumulate in CSCs and their progeny (see below).[17]

Evidence of the cancer stem cell model has been demonstrated in multiple tumour types including leukemias,[18][19] glioblastoma,[20] breast cancer,[21] and prostate cancer.[22]

However, the existence of CSCs is still under debate. One reason for this is that markers for CSCs have been difficult to reproduce across multiple tumours. Further, methods for determining tumourigenic potential utilize xenograft models. These methods suffer from inherent limitations such as the need to control immune response in the transplant animal, and the significant difference in environmental conditions from the primary tumour site to the xenograft site (e.g. absence of required exogenous molecules or cofactors).[23] This has caused some doubt about the accuracy of CSC results and the conclusions about which cells have tumourigenic potential.

Clonal Evolution

The clonal evolution model was first proposed in 1976 by Peter Nowell.[24] In this model, tumours arise from a single mutated cell, accumulating additional mutations as it progresses. These changes give rise to additional subpopulations, and each of these subpopulations has the ability to divide and mutate further. This heterogeneity may give rise to subclones that possess an evolutionary advantage over the others within the tumour environment, and these subclones may become dominant in the tumour over time.[25][26] When proposed, this model allowed for the understanding of tumour growth, treatment failure, and tumour aggression that occurs during the natural process of tumour formation.[25]

Branched evolution is more likely to contribute to tumour heterogeneity.

Evolution of the initial tumour cell may occur by two methods:

Linear expansion

Sequentially ordered mutations accumulate in driver genes, tumour suppressor genes, and DNA repair enzymes, resulting in clonal expansion of tumour cells. Linear expansion is less likely to reflect the endpoint of a malignant tumour[27] because the accumulation of mutations is stochastic in heterogeneic tumours.

Branched expansion

Expansion into multiple subclonal populations occurs through a splitting mechanism.[25] This method is more associated with tumour heterogeneity than linear expansion. The acquisition of mutations is random as a result of increased genomic instability with each successive generation. The long-term mutational accumulation may provide a selective advantage during certain stages of tumour progression. The tumour microenvironment may also contribute to tumour expansion, as it is capable of altering the selective pressures that the tumour cells are exposed to.[27]

Types & Causes of Heterogeneity

Multiple types of heterogeneity have been observed between tumour cells, stemming from both genetic and non-genetic variability.[28]

Genetic Heterogeneity

Genetic heterogeneity is a common feature of tumour genomes, and can arise from multiple sources. Some cancers are initiated when exogenous factors introduce mutations, such as ultraviolet radiation (skin cancers) and tobacco (lung cancer). A more common source is genomic instability, which often arises when key regulatory pathways are disrupted in the cells. Some examples include impaired DNA repair mechanisms which can lead to increased replication errors, and defects in the mitosis machinery that allow for large-scale gain or loss of entire chromosomes.[29] Furthermore, it is possible for genetic variability to be further increased by some cancer therapies (e.g. treatment with temozolomide and other chemotherapy drugs).[30][31]

Other Heterogeneity

Tumour cells can also show heterogeneity between their expression profiles. This is often caused by underlying epigenetic changes.[28] Variation in expression signatures have been detected in different regions of tumour samples within an individual. Researchers have shown that convergent mutations affecting H3K36 methyltransferase SETD2 and histone H3K4 demethylase KDM5C arose in spatially separated tumour sections. Similarly, MTOR, a gene encoding a cell regulatory kinase, has shown to be constitutively active, thereby increasing S6 phosphorylation. This active phosphorylation may serve as a biomarker in clear-cell carcinoma.[27]

Mechanochemical heterogeneity is a hallmark of living eukaryotic cells. It has an impact on epigenetic gene regulation. The heterogeneous dynamic mechanochemical processes regulate interrelationships within the group of cellular surfaces through adhesion.[32] Tumour development and spreading is accompanied by change in heterogeneous chaotic dynamics of mechanochemical interaction process in the group cells, including cells within tumour, and is hierarchical for the host of cancer patients.[33] The biological phenomena of mechanochemical heterogeneity maybe used for differential gastric cancer diagnostics against patients with inflammation of gastric mucosa[34] and for increasing antimetastatic activity of dendritic cells based on vaccines when mechanically heterogenized microparticles of tumor cells are used for their loading.[35]

Tumour microenvironment

Heterogeneity between tumour cells can be further increased due to heterogeneity in the tumour microenvironment. Regional differences in the tumour (e.g. availability of oxygen) impose different selective pressures on tumour cells, leading to a wider spectrum of dominant subclones in different spatial regions of the tumour. The influence of microenvironment on clonal dominance is also a likely reason for the heterogeneity between primary and metastatic tumours seen in many patients, as well as the inter-tumour heterogeneity observed between patients with the same tumour type.[36]

Implications & Challenges

Treatment resistance

Heterogeneic tumours may exhibit different sensitivities to cytotoxic drugs among different clonal populations. This is attributed to clonal interactions that may inhibit or alter therapeutic efficacy, posing a challenge for successful therapies in heterogeneic tumours (and their heterogeneic metastases).[1]

Drug administration in heterogeneic tumours will seldom kill all tumour cells. The initial heterogeneic tumour population may bottleneck, such that few drug resistant cells (if any) will survive. This allows resistant tumour populations to replicate and grow a new tumour through the branching evolution mechanism (see above). The resulting repopulated tumour is heterogeneic and resistant to the initial drug therapy used. The repopulated tumour may also return in a more aggressive manner.

The administration of cytotoxic drugs often results in initial tumour shrinkage. This represents the destruction of initial non-resistant subclonal populations within a heterogeneic tumour, leaving only resistant clones. These resistant clones now contain a selective advantage and can replicate to repopulate the tumour. Replication will likely occur through branching evolution, contributing to tumour heterogeneity. The repopulated tumour may appear to be more aggressive. This is attributed to the drug-resistant selective advantage of the tumour cells.

Drug treatment induces a bottleneck effect, where resistant subclones will survive and propagate to re-form a heterogeneous tumour.

Biomarker Discovery

Due to the genetic differences within and between tumours, biomarkers that may predict treatment response or prognosis may not be widely applicable. However, it has been suggested that the level of heterogeneity can itself be used as a biomarker since more heterogeneous tumours may be more likely to contain treatment-resistant subclones.[28] Further research into developing biomarkers that account for heterogeneity is still in progress.

Model Systems

Current model systems typically lack the heterogeneity seen in human cancers.[37] In order to accurately study tumour heterogeneity, we must develop more accurate preclinical models. One such model, the patient derived tumour xenograft, has shown excellent utility in preserving tumour heterogeneity whilst allowing detailed study of the drivers of clonal fitness.[38] However, even this model cannot capture the full complexity of cancer.

Current Strategies

While the problem of identifying, characterizing, and treating tumour heterogeneity is still under active research, some effective strategies have been proposed, including both experimental and computational solutions.

Experimental

Sequencing

See also

References

  1. 1 2 3 4 5 6 7 8 9 Marusyk, A; Polyak, K (2010). "Tumor heterogeneity: Causes and consequences". Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 1805 (1): 105–117. doi:10.1016/j.bbcan.2009.11.002. PMC 2814927Freely accessible. PMID 19931353.
  2. Campbell, P. J.; Pleasance, E. D.; Stephens, P. J.; Dicks, E; Rance, R; Goodhead, I; Follows, G. A.; Green, A. R.; Futreal, P. A.; Stratton, M. R. (2008). "Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing". Proceedings of the National Academy of Sciences. 105 (35): 13081–13086. doi:10.1073/pnas.0801523105. PMC 2529122Freely accessible. PMID 18723673.
  3. Shipitsin, M; Campbell, L. L.; Argani, P; Weremowicz, S; Bloushtain-Qimron, N; Yao, J; Nikolskaya, T; Serebryiskaya, T; Beroukhim, R; Hu, M; Halushka, M. K.; Sukumar, S; Parker, L. M.; Anderson, K. S.; Harris, L. N.; Garber, J. E.; Richardson, A. L.; Schnitt, S. J.; Nikolsky, Y; Gelman, R. S.; Polyak, K (2007). "Molecular definition of breast tumor heterogeneity". Cancer Cell. 11 (3): 259–273. doi:10.1016/j.ccr.2007.01.013. PMID 17349583.
  4. MacIntosh, C. A.; Stower, M; Reid, N; Maitland, N. J. (1998). "Precise microdissection of human prostate cancers reveals genotypic heterogeneity". Cancer Research. 58 (1): 23–28. PMID 9426051.
  5. Alvarado, C; Beitel, L. K.; Sircar, K; Aprikian, A; Trifiro, M; Gottlieb, B (2005). "Somatic mosaicism and cancer: A micro-genetic examination into the role of the androgen receptor gene in prostate cancer". Cancer Research. 65 (18): 8514–8518. doi:10.1158/0008-5472.CAN-05-0399. PMID 16166332.
  6. Konishi, N; Hiasa, Y; Matsuda, H; Tao, M; Tsuzuki, T; Hayashi, I; Kitahori, Y; Shiraishi, T; Yatani, R; Shimazaki, J (1995). "Intratumor cellular heterogeneity and alterations in ras oncogene and p53 tumor suppressor gene in human prostate carcinoma". The American Journal of Pathology. 147 (4): 1112–1122. PMC 1871010Freely accessible. PMID 7573356.
  7. González-García, I; Solé, R. V.; Costa, J (2002). "Metapopulation dynamics and spatial heterogeneity in cancer". Proceedings of the National Academy of Sciences. 99 (20): 13085–13089. doi:10.1073/pnas.202139299. PMC 130590Freely accessible. PMID 12351679.
  8. Samowitz, W. S.; Slattery, M. L. (1999). "Regional reproducibility of microsatellite instability in sporadic colorectal cancer". Genes, Chromosomes and Cancer. 26 (2): 106–114. doi:10.1002/(SICI)1098-2264(199910)26:2<106::AID-GCC2>3.0.CO;2-F. PMID 10469448.
  9. Giaretti, W; Monaco, R; Pujic, N; Rapallo, A; Nigro, S; Geido, E (1996). "Intratumor heterogeneity of K-ras2 mutations in colorectal adenocarcinomas: Association with degree of DNA aneuploidy". The American Journal of Pathology. 149 (1): 237–245. PMC 1865212Freely accessible. PMID 8686748.
  10. Heppner, G. H. (1984). "Tumor heterogeneity". Cancer Research. 44 (6): 2259–2265. PMID 6372991.
  11. Maley, C. C.; Galipeau, P. C.; Finley, J. C.; Wongsurawat, V. J.; Li, X; Sanchez, C. A.; Paulson, T. G.; Blount, P. L.; Risques, R. A.; Rabinovitch, P. S.; Reid, B. J. (2006). "Genetic clonal diversity predicts progression to esophageal adenocarcinoma". Nature Genetics. 38 (4): 468–473. doi:10.1038/ng1768. PMID 16565718.
  12. Califano, J; Van Der Riet, P; Westra, W; Nawroz, H; Clayman, G; Piantadosi, S; Corio, R; Lee, D; Greenberg, B; Koch, W; Sidransky, D (1996). "Genetic progression model for head and neck cancer: Implications for field cancerization". Cancer Research. 56 (11): 2488–2492. PMID 8653682.
  13. Sauter, G; Moch, H; Gasser, T. C.; Mihatsch, M. J.; Waldman, F. M. (1995). "Heterogeneity of chromosome 17 and erbB-2 gene copy number in primary and metastatic bladder cancer". Cytometry. 21 (1): 40–46. doi:10.1002/cyto.990210109. PMID 8529469.
  14. Fujii, H; Yoshida, M; Gong, Z. X.; Matsumoto, T; Hamano, Y; Fukunaga, M; Hruban, R. H.; Gabrielson, E; Shirai, T (2000). "Frequent genetic heterogeneity in the clonal evolution of gynecological carcinosarcoma and its influence on phenotypic diversity". Cancer Research. 60 (1): 114–120. PMID 10646862.
  15. Horvai, A. E.; Devries, S; Roy, R; O'Donnell, R. J.; Waldman, F (2009). "Similarity in genetic alterations between paired well-differentiated and dedifferentiated components of dedifferentiated liposarcoma". Modern Pathology. 22 (11): 1477–1488. doi:10.1038/modpathol.2009.119. PMID 19734852.
  16. Pantou, D; Rizou, H; Tsarouha, H; Pouli, A; Papanastasiou, K; Stamatellou, M; Trangas, T; Pandis, N; Bardi, G (2005). "Cytogenetic manifestations of multiple myeloma heterogeneity". Genes, Chromosomes and Cancer. 42 (1): 44–57. doi:10.1002/gcc.20114. PMID 15495197.
  17. 1 2 Shackleton, M; Quintana, E; Fearon, E. R.; Morrison, S. J. (2009). "Heterogeneity in cancer: Cancer stem cells versus clonal evolution". Cell. 138 (5): 822–829. doi:10.1016/j.cell.2009.08.017. PMID 19737509.
  18. Lapidot, T; Sirard, C; Vormoor, J; Murdoch, B; Hoang, T; Caceres-Cortes, J; Minden, M; Paterson, B; Caligiuri, M. A.; Dick, J. E. (1994). "A cell initiating human acute myeloid leukaemia after transplantation into SCID mice". Nature. 367 (6464): 645–648. doi:10.1038/367645a0. PMID 7509044.
  19. Wang, J. C.; Lapidot, T; Cashman, J. D.; Doedens, M; Addy, L; Sutherland, D. R.; Nayar, R; Laraya, P; Minden, M; Keating, A; Eaves, A. C.; Eaves, C. J.; Dick, J. E. (1998). "High level engraftment of NOD/SCID mice by primitive normal and leukemic hematopoietic cells from patients with chronic myeloid leukemia in chronic phase". Blood. 91 (7): 2406–2414. PMID 9516140.
  20. Singh, S. K.; Hawkins, C; Clarke, I. D.; Squire, J. A.; Bayani, J; Hide, T; Henkelman, R. M.; Cusimano, M. D.; Dirks, P. B. (2004). "Identification of human brain tumour initiating cells". Nature. 432 (7015): 396–401. doi:10.1038/nature03128. PMID 15549107.
  21. Al-Hajj, M; Wicha, M. S.; Benito-Hernandez, A; Morrison, S. J.; Clarke, M. F. (2003). "Prospective identification of tumorigenic breast cancer cells". Proceedings of the National Academy of Sciences. 100 (7): 3983–3988. doi:10.1073/pnas.0530291100. PMC 153034Freely accessible. PMID 12629218.
  22. Maitland, N. J.; Collins, A. T. (2008). "Prostate cancer stem cells: A new target for therapy". Journal of Clinical Oncology. 26 (17): 2862–2870. doi:10.1200/JCO.2007.15.1472. PMID 18539965.
  23. Meacham, C. E.; Morrison, S. J. (2013). "Tumour heterogeneity and cancer cell plasticity". Nature. 501 (7467): 328–337. doi:10.1038/nature12624. PMID 24048065.
  24. Nowell, P. C. (1976). "The clonal evolution of tumor cell populations". Science. 194 (4260): 23–28. doi:10.1126/science.959840. PMID 959840.
  25. 1 2 3 Swanton, C (2012). "Intratumor heterogeneity: Evolution through space and time". Cancer Research. 72 (19): 4875–4882. doi:10.1158/0008-5472.CAN-12-2217. PMC 3712191Freely accessible. PMID 23002210.
  26. Merlo, L. M. F.; Pepper, J. W.; Reid, B. J.; Maley, C. C. (2006). "Cancer as an evolutionary and ecological process". Nature Reviews Cancer. 6 (12): 924–935. doi:10.1038/nrc2013. PMID 17109012.
  27. 1 2 3 Gerlinger, M; Rowan, A. J.; Horswell, S; Larkin, J; Endesfelder, D; Gronroos, E; Martinez, P; Matthews, N; Stewart, A; Tarpey, P; Varela, I; Phillimore, B; Begum, S; McDonald, N. Q.; Butler, A; Jones, D; Raine, K; Latimer, C; Santos, C. R.; Nohadani, M; Eklund, A. C.; Spencer-Dene, B; Clark, G; Pickering, L; Stamp, G; Gore, M; Szallasi, Z; Downward, J; Futreal, P. A.; Swanton, C (2012). "Intratumor heterogeneity and branched evolution revealed by multiregion sequencing". New England Journal of Medicine. 366 (10): 883–892. doi:10.1056/NEJMoa1113205. PMID 22397650.
  28. 1 2 3 Marusyk, A; Almendro, V; Polyak, K (2012). "Intra-tumour heterogeneity: A looking glass for cancer?". Nature Reviews Cancer. 12 (5): 323–334. doi:10.1038/nrc3261. PMID 22513401.
  29. Burrell, R. A.; McGranahan, N; Bartek, J; Swanton, C (2013). "The causes and consequences of genetic heterogeneity in cancer evolution". Nature. 501 (7467): 338–345. doi:10.1038/nature12625. PMID 24048066.
  30. Johnson, B. E.; Mazor, T; Hong, C; Barnes, M; Aihara, K; McLean, C. Y.; Fouse, S. D.; Yamamoto, S; Ueda, H; Tatsuno, K; Asthana, S; Jalbert, L. E.; Nelson, S. J.; Bollen, A. W.; Gustafson, W. C.; Charron, E; Weiss, W. A.; Smirnov, I. V.; Song, J. S.; Olshen, A. B.; Cha, S; Zhao, Y; Moore, R. A.; Mungall, A. J.; Jones, S. J.; Hirst, M; Marra, M. A.; Saito, N; Aburatani, H; Mukasa, A (2014). "Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma". Science. 343 (6167): 189–193. doi:10.1126/science.1239947. PMC 3998672Freely accessible. PMID 24336570.
  31. 1 2 Ding, L; Ley, T. J.; Larson, D. E.; Miller, C. A.; Koboldt, D. C.; Welch, J. S.; Ritchey, J. K.; Young, M. A.; Lamprecht, T; McLellan, M. D.; McMichael, J. F.; Wallis, J. W.; Lu, C; Shen, D; Harris, C. C.; Dooling, D. J.; Fulton, R. S.; Fulton, L. L.; Chen, K; Schmidt, H; Kalicki-Veizer, J; Magrini, V. J.; Cook, L; McGrath, S. D.; Vickery, T. L.; Wendl, M. C.; Heath, S; Watson, M. A.; Link, D. C.; Tomasson, M. H. (2012). "Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing". Nature. 481 (7382): 506–510. doi:10.1038/nature10738. PMC 3267864Freely accessible. PMID 22237025.
  32. G.M.Edelman (1989). "Topobiology". J. Scientific American (260): 76–88.
  33. V.E. Orel; N.N Dzyatkovskaya; M.I. Danko; A.V. Romanov; Y.I. Mel'nik; Y.A. Grinevich; S.V. Martynenko (2004). "Spatial and mechanoemission chaos of mechanically deformed tumor cells". J. Journal of Mechanics in Medicine and Biology. 4 (1): 31–45.
  34. V.E. Orel; A.V. Romanov; N.N. Dzyatkovskaya; Yu.I. Mel’nik (2002). "The device and algorithm for estimation of the mechanoemisson chaos in blood of patients with gastric cancer". J. Medical Engineering & Physics. 24 (5): 365–371.
  35. N. Khranovskaya; V. Orel; Y. Grinevich; O. Alekseenko; A. Romanov; O. Skachkova; N.Dzyatkovskaya; A. Burlaka,; S.Lukin (2012). "Mechanical heterogenization of Lewis lung carcinoma cells can improve antimetastatic effect of dendritic cells". J. Journal of Mechanics in Medicine and Biology. 3 (12): 22. doi:10.1142/S0219519411004757.
  36. Junttila, M. R.; De Sauvage, F. J. (2013). "Influence of tumour micro-environment heterogeneity on therapeutic response". Nature. 501 (7467): 346–354. doi:10.1038/nature12626. PMID 24048067.
  37. Auman, James Todd; McLeod, Howard L. (2010-01-01). "Colorectal Cancer Cell Lines Lack the Molecular Heterogeneity of Clinical Colorectal Tumors". Clinical Colorectal Cancer. 9 (1): 40–47. doi:10.3816/ccc.2010.n.005.
  38. Cassidy, John W.; Caldas, Carlos; Bruna, Alejandra (2015-08-01). "Maintaining Tumor Heterogeneity in Patient-Derived Tumor Xenografts". Cancer Research. 75 (15): 2963–2968. doi:10.1158/0008-5472.CAN-15-0727. ISSN 0008-5472. PMC 4539570Freely accessible. PMID 26180079.
  39. Bai H, Harmancı AS, Erson-Omay AZ, Li J, Coșkun S, Simon M, et al. (Nov 2015). "Integrated genomic characterization of IDH1-mutant glioma malignant progression". Nature Genetics. doi:10.1038/ng.3457.
  40. Bedard, P. L.; Hansen, A. R.; Ratain, M. J.; Siu, L. L. (2013). "Tumour heterogeneity in the clinic". Nature. 501 (7467): 355–364. doi:10.1038/nature12627. PMID 24048068.
  41. Dawson, S. J.; Tsui, D. W.; Murtaza, M; Biggs, H; Rueda, O. M.; Chin, S. F.; Dunning, M. J.; Gale, D; Forshew, T; Mahler-Araujo, B; Rajan, S; Humphray, S; Becq, J; Halsall, D; Wallis, M; Bentley, D; Caldas, C; Rosenfeld, N (2013). "Analysis of circulating tumor DNA to monitor metastatic breast cancer". New England Journal of Medicine. 368 (13): 1199–1209. doi:10.1056/NEJMoa1213261. PMID 23484797.
  42. Gatenby, R. A.; Silva, A. S.; Gillies, R. J.; Frieden, B. R. (2009). "Adaptive therapy". Cancer Research. 69 (11): 4894–4903. doi:10.1158/0008-5472.CAN-08-3658. PMC 3728826Freely accessible. PMID 19487300.
  43. Cibulskis, K; Lawrence, M. S.; Carter, S. L.; Sivachenko, A; Jaffe, D; Sougnez, C; Gabriel, S; Meyerson, M; Lander, E. S.; Getz, G (2013). "Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples". Nature Biotechnology. 31 (3): 213–219. doi:10.1038/nbt.2514. PMC 3833702Freely accessible. PMID 23396013.
  44. Koboldt, D. C.; Zhang, Q; Larson, D. E.; Shen, D; McLellan, M. D.; Lin, L; Miller, C. A.; Mardis, E. R.; Ding, L; Wilson, R. K. (2012). "Var Scan 2: Somatic mutation and copy number alteration discovery in cancer by exome sequencing". Genome Research. 22 (3): 568–576. doi:10.1101/gr.129684.111. PMC 3290792Freely accessible. PMID 22300766.
  45. Saunders, C. T.; Wong, W. S.; Swamy, S; Becq, J; Murray, L. J.; Cheetham, R. K. (2012). "Strelka: Accurate somatic small-variant calling from sequenced tumor-normal sample pairs". Bioinformatics. 28 (14): 1811–1817. doi:10.1093/bioinformatics/bts271. PMID 22581179.
  46. Carter, S. L.; Cibulskis, K; Helman, E; McKenna, A; Shen, H; Zack, T; Laird, P. W.; Onofrio, R. C.; Winckler, W; Weir, B. A.; Beroukhim, R; Pellman, D; Levine, D. A.; Lander, E. S.; Meyerson, M; Getz, G (2012). "Absolute quantification of somatic DNA alterations in human cancer". Nature Biotechnology. 30 (5): 413–421. doi:10.1038/nbt.2203. PMID 22544022.
  47. Shah, S. P.; Roth, A; Goya, R; Oloumi, A; Ha, G; Zhao, Y; Turashvili, G; Ding, J; Tse, K; Haffari, G; Bashashati, A; Prentice, L. M.; Khattra, J; Burleigh, A; Yap, D; Bernard, V; McPherson, A; Shumansky, K; Crisan, A; Giuliany, R; Heravi-Moussavi, A; Rosner, J; Lai, D; Birol, I; Varhol, R; Tam, A; Dhalla, N; Zeng, T; Ma, K; Chan, S. K. (2012). "The clonal and mutational evolution spectrum of primary triple-negative breast cancers". Nature. 486 (7403): 395–399. doi:10.1038/nature10933. PMC 3863681Freely accessible. PMID 22495314.
  48. Navin, N; Kendall, J; Troge, J; Andrews, P; Rodgers, L; McIndoo, J; Cook, K; Stepansky, A; Levy, D; Esposito, D; Muthuswamy, L; Krasnitz, A; McCombie, W. R.; Hicks, J; Wigler, M (2011). "Tumour evolution inferred by single-cell sequencing". Nature. 472 (7341): 90–94. doi:10.1038/nature09807. PMID 21399628.
  49. Zare, Habil (2014). "Inferring clonal composition from multiple sections of a breast cancer". PLOS Computational Biology. 10: e1003703. doi:10.1371/journal.pcbi.1003703.
  50. Fischer, Andrej (2014). "High-definition reconstruction of clonal composition in cancer". Cell Reports. 7: 1740–1752. doi:10.1016/j.celrep.2014.04.055.
  51. Deshwar, Amit (2015). "Monitoring chronic lymphocytic leukemia progression by whole genome sequencing reveals heterogeneous clonal evolution patterns". Genome Biology. 16: 1–20. doi:10.1182/blood-2012-05-433540.
  52. Roth, Andrew (2014). "PyClone: statistical inference of clonal population structure in cancer". Nature Methods. 11: 396–398. doi:10.1038/nmeth.2883.
  53. Marass, Francesco (2015). "A phylogenetic latent feature model for clonal deconvolution". arXiv:1604.01715Freely accessible.
  54. Matsui, Yusuke (2016). "phyC: Clustering cancer evolutionary trees" (PDF). Bioinformatics.
  55. Jiang, Yuchao; Qiu, Yu; Minn, Andy J.; Zhang, Nancy R. (29 August 2016). "Assessing intratumor heterogeneity and tracking longitudinal and spatial clonal evolutionary history by next-generation sequencing". Proceedings of the National Academy of Sciences: 2373. doi:10.1073/pnas.1522203113. Retrieved 6 September 2016.
This article is issued from Wikipedia - version of the 9/7/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.