Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1436 2023-05-17 11:11:47 |
2 layout Meta information modification 1436 2023-05-18 03:07:33 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kozlov, A.P. Carcino-Evo-Devo. Encyclopedia. Available online: https://encyclopedia.pub/entry/44424 (accessed on 29 April 2024).
Kozlov AP. Carcino-Evo-Devo. Encyclopedia. Available at: https://encyclopedia.pub/entry/44424. Accessed April 29, 2024.
Kozlov, Andrei P.. "Carcino-Evo-Devo" Encyclopedia, https://encyclopedia.pub/entry/44424 (accessed April 29, 2024).
Kozlov, A.P. (2023, May 17). Carcino-Evo-Devo. In Encyclopedia. https://encyclopedia.pub/entry/44424
Kozlov, Andrei P.. "Carcino-Evo-Devo." Encyclopedia. Web. 17 May, 2023.
Carcino-Evo-Devo
Edit

The term “carcino-evo-devo” was used for the first time as a name for the theory of the evolutionary role of tumors. The new term was coined from two other terms: “carcinoembryonic” and “evo-devo”.

carcinoembryonic evo-devo carcino-evo-devo evolution hereditary tumors

1. Introduction

The theory of the evolutionary role of hereditary tumors, or the carcino-evo-devo theory, was developed in order to explain the sources of additional cell masses, which were necessary for the origins of new cell types, tissues, and organs during the progressive evolution of multicellular organisms. The content of the theory has been published in the theoretical papers of the author [1][2][3][4][5][6][7][8][9][10][11][12][13] and as a monograph in English [14], Russian [15], and Chinese [16]. Many experimental papers from the author’s lab were devoted to the confirmation of nontrivial predictions of the carcino-evo-devo theory [17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39].
The basic statements of the carcino-evo-devo theory are the following:
  • Tumor processes participate in the evolution of development;
  • Hereditary tumors provide evolving multicellular organisms with extra cell masses for the expression of evolutionarily novel genes and gene combinations, and thereby participate in the origins of new cell types, tissues, and organs;
  • Populations of tumor-bearing organisms served as transitional forms in progressive evolution;
  • Tumors may be considered search engines for new gene combinations in the space of biological possibilities.

2. Preceding and Related Concepts

The term “carcino-evo-devo” was first introduced in [14], and it was used for the first time as a name for the theory of the evolutionary role of tumors in [9]. The new term was coined from two other terms: “carcinoembryonic” and “evo-devo”.
The term “carcinoembryonic” is used to designate embryonal proteins produced by tumor cells. The first examples of such proteins were alpha-fetoprotein [40][41] and carcinoembryonic antigen [42]. The concept of carcinoembryonic proteins was further developed in the concept of the convergence of embryonic and cancer signaling pathways [43][44][45][46]. These concepts point to molecular links between embryonal and tumor processes.
The abbreviation “evo-devo” corresponds to evolutionary developmental biology, which examines connections of normal individual development and evolution [47].
The carcino-evo-devo theory interconnects evolutionary, individual, and neoplastic development within one unified consideration. This theory studies the role of hereditary tumors in the evolution of development [9][14].
The previous concepts important for the carcino-evo-devo theory are the “embryonal rest” or “embryonal remnants” theory of cancer [48][49][50]; the morphological laws of the evolution of ontogenesis [51]; and the concept of neoplasia as a disease of differentiation [52].
Several previous ideas may be considered historical background for the theory, such as the idea of the role of “hopeful monsters” in evolution [53][54][55][56], and the idea of the positive role of viruses in evolution [57].
The carcino-evo-devo theory has been developing concurrently with Darwinian medicine [58], evolutionary epidemiology [59], and several branches of evolutionary oncology (e.g., the somatic evolution of tumor cells and selection in tumor cell populations [60][61][62], cancer selection [63], and the ecological hypothesis [64]). Comparative oncology has been in place since the 19th century (reviewed in [14]).
So, the carcino-evo-devo theory is deeply rooted in experimental oncology, developmental and evolutionary biology, and evolutionary medicine.

3. Tumor Features That Suggest the Role of Tumors in the Evolution of Organisms

3.1. Hereditary Tumors, like Any Hereditary Trait, Could Be Used in Evolution

Many tumors are hereditary. The high incidence of certain tumors in different strains of laboratory animals is a well-known phenomenon [14][65][66]. Cellular oncogenes and tumor suppressor genes are inherited as part of germline DNA and thus provide a genetic basis for tumor inheritance. The importance of germline mutations in cancer was recognized after the pioneering works of Alfred Knudson [67][68][69]. Twenty percent of cancer-associated mutations occur in the germline DNA, and ten percent are both somatic and germline mutations [70][71]. There is an inverse association between germline susceptibility risk and somatic genetic variation in human cancer [72][73]. Heritable epimutations associated with hereditary tumors (e.g., in Lynch syndrome) have also been described [74].
Tumor prevalence in wild and captive animals can reach a high percentage (e.g., >50% in Santa Catalina Island foxes) [75][76]. The highest cancer prevalence has been recorded in species or populations with low genetic diversity, which suggests that it may have a genetic basis (reviewed in [75]). In humans, increased cancer prevalence is also associated with reduced genetic diversity [77][78].
Hereditary cancer syndromes connected with inherited mutations in cancer predisposition genes, such as MMR or BRCA genes, account for a considerable proportion (up to 10%) of all cancers in humans [79][80][81][82]. The identification and clinical management of patients with such mutations became routine in modern oncology [83]. For the purpose of the discussion, it is important that hereditary cancer syndromes are much more frequent than noncancer hereditary syndromes, the classical genetic diseases (reviewed in [84]).
Any hereditary trait may be selected and may potentially be evolutionarily meaningful. The fact that many tumors are hereditary suggests that they might participate in the evolution of organisms. For example, germline mutations in BRCA1 and BRCA2 tumor suppressor genes increase the risk of breast and ovarian cancer in humans, and Brca1 and Brca2 genes participate in the embryonic development and differentiation of mammary glands in mice (reviewed in [10]).
When I talk about the evolutionary role of tumors, I mean hereditary tumors. More discussion on hereditary tumors may be found in [14].

3.2. The Widespread Occurrence of Tumors in Multicellular Organisms

Tumors are widespread throughout the phylogenetic tree [14]. New reviews on this topic, which appeared after the publication of the author’s book, support the broad distribution of tumors across the tree of life [85][86].
Cellular oncogenes and tumor suppressor genes are also widespread throughout the phylogenetic tree, and many of them are ancient (oncogenes were already present in sponges).

3.3. A Considerable Portion of Tumors Never Kill Their Hosts

Many (maybe most) tumors never kill their hosts. Benign tumors are widespread in nature. They may represent more than half of all tumors [14]. New publications support this view: up to 80% of mammalian tumors are benign [76][87]. For example, in humans, benign lesions in the nasopharynx accounted for 75% of cases, with malignancies accounting for 25% [88].
At earlier stages of progression, tumors do not kill their hosts. Moreover, the overall direction of tumors in the early stages is toward regression [89]. Tumors can be selected for new functions at the early stages of progression [90].
The list of tumors that do not kill their hosts and could be used in evolution includes fetal, neonatal, and infantile tumors; carcinomas in situ and pseudodiseases; tumors that spontaneously regress; and sustainable tumor masses or extra cell masses in dynamic equilibrium with the organism [14].
When I talk about the evolutionary role of hereditary tumors, I mean benign tumors or tumors at the early stages of progression, or the kinds of tumors mentioned above, but not malignant tumors at the final stages of progression, which kill their hosts.

3.4. Tumors Have Many Features That Could Be Used in Evolution

Tumors are excessive cell masses that are not functionally necessary for the organism. Many unusual genes, which are not expressed in normal tissues, are expressed in tumors. There are also many unusual gene combinations expressed in tumors. Tumor cells can differentiate with the loss of malignancy, and tumors have morphogenetic potential.
All these features are much in demand in evolving multicellular organisms. However, they are not widely appreciated or discussed because the scientific community is more interested in tumors that kill the organism (i.e., malignant tumors, and the medical aspects of malignancy). 
The data exist that tumors were indeed used in evolution (e.g., the positive selection of tumor-associated genes in primate evolution (reviewed in [14])), and examples of organs that possibly originated from tumors [9][10][11][14][91]. The placenta [9][10][14], the mammary gland and prostate [9][10], and mammalian adipose [11] are a few examples of organs that might have developed from tumors. In the author’s book [14], further examples are covered, including the nitrogen-fixing root nodules of legumes, melanomatous cells and macromelanophores of Xiphophorus fish, hoods of Lionhead goldfish, and malignant papillomatosis and symbiovilli of voles.
New data suggest that tumors are connected with evolutionary adaptation [92] and the evolution rate [93].

3.5. “Nothing in Biology Makes Sense except in the Light of Evolution” [94]

Tumors are no exception to Dobzhansky’s maxima, and biologists have started thinking about the place that tumors occupy in evolution. Several monographs have been published over the last decade on tumors and evolution [14][63][95][96]. Until very recently, the role of tumors in evolution was considered completely negative. The “tumors and evolution” relationships were studied mainly in a comparative biological context, or in the context of the evolution of tumor cells in tumor cell populations. However, the hereditary nature of many tumors, the widespread occurrence of tumors, the fact that a considerable proportion of tumors never kill their hosts, and tumor features that could be used in the evolution of multicellular organisms are all features that suggest that hereditary tumors could play some positive role in the evolution of host organisms.

References

  1. Kozlov, A.P. Regulatory Mechanisms as an Expression and the Result of Evolution of Competitive Relations between the Genes. In Salinity Adaptations of Water Organisms; Nauka: Leningrad, Russia, 1976; pp. 237–345.
  2. Kozlov, A.P. Evolution of Living Organisms as a Multilevel Process. J. Theor. Biol. 1979, 81, 1–17.
  3. Kozlov, A.P. The principles of multilevel development of organisms. In Problems of Analysis of Biological Systems; Maximov, V.N., Ed.; Moscow University Press: Moscow, Russia, 1983; pp. 48–62.
  4. Kozlov, A.P. Gene competition and the possible evolutionary role of tumors and cellular oncogenes. In Theoretical and Mathematical Aspects of Morphogenesis; Presnov, E.V., Maresin, V.M., Zotin, A.I., Eds.; Nauka: Moscow, Russia, 1987; pp. 136–140.
  5. Kozlov, A.P. Conservation principles in the system of molecular-biological laws. Trans. Leningr. Soc. Nat. Sci. 1988, 87, 4–21.
  6. Kozlov, A.P. Gene competition and the possible evolutionary role of tumors. Med. Hypotheses 1996, 46, 81–84.
  7. Kozlov, A.P. Tumors and evolution. Vopr. Oncol. 2008, 54, 695–705.
  8. Kozlov, A.P. The Possible Evolutionary Role of Tumors in the Origin of New Cell Types. Med. Hypotheses 2010, 74, 177–185.
  9. Kozlov, A.P. The role of heritable tumors in evolution of development: A new theory of carcino-evo-devo. Acta Nat. 2019, 11, 65–72.
  10. Kozlov, A.P. Mammalian tumor-like organs.1. The role of tumor-like normal organs and atypical tumor organs in the evolution of development (carcino-evo-devo). Infect. Agents Cancer 2022, 17, 2.
  11. Kozlov, A.P. Mammalian tumor-like organs. 2. Mammalian adipose has many tumor features and obesity is a tumor-like process. Infect. Agents Cancer 2022, 17, 15.
  12. Kozlov, A.P. Mammalian adipose has many tumor features, which suggests the possible way of its evolutionary origin. Proceedings of the American Association of Cancer Research Annual Meeting, New Orleans, LA, USA, 8–13 April 2022. Cancer Res. 2022, 82 (Suppl. S12), 6077.
  13. Kozlov, A.P. Biological computation and compatibility search in the possibility space as the mechanism of complexity increase during progressive evolution. Evol. Bioinform. 2022, 18, 1–5.
  14. Kozlov, A.P. Evolution by Tumor Neofunctionalization. The Role of Tumors in the Origin of New Cell Types, Tissues and Organs; Elsevier/Academic Press: Amsterdam, Boston, Heidelberg, London, New York, Oxford, Paris, San Diego, San Francisco, Singapore, Sydney, Tokyo, 2014; pp. 1–248.
  15. Kozlov, A.P. Evolution by Tumor Neofunctionalization: Neoplasms as the Factor of Progressive Evolution; Publishing House of Peter the Great Polytechnic University: St. Petersburg, Russia, 2016; pp. 1–263.
  16. Kozlov, A.P. Evolution by Tumor Neofunctionalization: The Role of Tumors in the Origin of New Cell Types, Tissues and Organs; China Science Publishing & Media Ltd. (Science Press): Beijing, China, 2019; pp. 1–200.
  17. Evtushenko, V.I.; Hanson, K.P.; Barabitskaya, O.V.; Emelyanov, A.V.; Reshetnikov, V.L.; Kozlov, A.P. Determination of the upper limit of rat genome expression. Mol. Biol. 1989, 23, 663–67518.
  18. Baranova, A.V.; Lobashev, A.V.; Ivanov, D.V.; Krukovskaya, L.L.; Yankovsky, N.K.; Kozlov, A.P. In silico screening for tumor-specific expressed sequences in human genome. FEBS Lett. 2001, 508, 143–148.
  19. Krukovskaya, L.L.; Baranova, A.; Tyezelova, T.; Polev, D.; Kozlov, A.P. Experimental study of human expressed sequences newly identified in silico as tumor specific. Tumor Biol. 2005, 26, 17–24.
  20. Kozlov, A.P.; Galachyants, Y.P.; Dukhovlinov, I.V.; Samusik, N.A.; Baranova, A.V.; Polev, D.E.; Krukovskaya, L.L. Evolutionarily new sequences expressed in tumors. Infect. Agents Cancer 2006, 1, 8.
  21. Palena, C.; Polev, D.E.; Tsang, K.Y.; Fernando, R.I.; Litzinger, M.; Krukovskaya, L.L.; Baranova, A.V.; Kozlov, A.P.; Schlom, J. The human T-box mesodermal transcription factor Brachyury is a candidate target for T-cell-mediated cancer immunotherapy. Clin. Cancer Res. 2007, 13, 2471–2478.
  22. Galachyants, Y.; Kozlov, A.P. CDD as a tool for discovery of specifically-expressed transcripts. Russ. J. AIDS Cancer Public Health 2009, 13, 60–61.
  23. Polev, D.; Nosova, J.; Krukovskaya, L.; Baranova, A.; Kozlov, A. Expression of transcripts related to the cluster HS.633957 in human tissues. Mol. Biol. 2009, 43, 97–102.
  24. Krukovskaia, L.L.; Polev, D.E.; Nosova, I.K.; Baranova, A.V.; Koliubaeva, S.N.; Kozlov, A.P. Investigation of transcription factor Brachyury (T) expression in human normal and tumor tissues. Vopr. Onkol. 2008, 54, 739–743.
  25. Krukovskaja, L.L.; Samusik, N.D.; Shilov, Y.S.; Polev, D.Y.; Kozlov, A.P. Tumor-specific expression of the newly-discovered PBOV1 gene. Vopr. Onkol. 2010, 56, 327–332.
  26. Polev, D.E.; Krukovskaya, L.L.; Kozlov, A.P. Expression of the locus Hs.633957 in human digestive system and tumors. Vopr. Oncol. 2011, 57, 48–49.
  27. Samusik, N.A.; Galachyants, Y.P.; Kozlov, A.P. Analysis of evolutionary novelty of tumor-specifically expressed sequences. Russ. J. Genet. Appl. Res. 2011, 1, 138–148.
  28. Dobrynin, P.; Matyunina, E.; Malov, S.V.; Kozlov, A.P. The novelty of human cancer/testis antigen encoding genes in evolution. Int. J. Genom. 2013, 2013, e105108.
  29. Samusik, N.; Krukovskaya, L.; Meln, I.; Shilov, E.; Kozlov, A.P. PBOV1 is a human de novo gene with tumor-specific expression that is associated with a positive clinical outcome of cancer. PLoS ONE 2013, 8, e56162.
  30. Polev, D.E.; Karnaukhova, J.K.; Krukovskaya, L.L.; Kozlov, A.P. ELFN1-AS1, a novel primate gene with possible microRNA function expressed predominantly in tumors. BioMed Res. Int. 2014, 2014, e398097.
  31. Matyunina, E.; Emelyanov, A.; Kozlov, A. Evolutionarily novel genes expressed in fish tumors determine progressive evolutionary characters. Cancer Res. 2015, 75 (Suppl. S15), 1927.
  32. Kozlov, A.P. Expression of evolutionarily novel genes in tumors. Infect. Agents Cancer 2016, 11, 34.
  33. Krukovskaya, L.L.; Polev, D.E.; Kurbatova, T.V.; Karnaukhova, Y.K.; Kozlov, A.P. The study of the tumor specificity of expression of some evolutionarily novel genes. Vopr. Oncol. 2016, 62, 495–500.
  34. Karnaukhova, Y.K.; Polev, D.E.; Krukovskaya, L.L.; Kozlov, A.P. Study of the expression of gene Orthopedia homeobox in various tumor and normal human tissues. Vopr. Oncol. 2017, 63, 128–134.
  35. Makashov, A.; Malov, S.V.; Kozlov, A.P. Oncogenes, tumor suppressor and differentiation genes represent the oldest human gene classes and evolve concurrently. Sci. Rep. 2019, 9, 16410.
  36. Matyunina, E.A.; Emelyanov, A.V.; Kurbatova, T.V.; Makashov, A.A.; Mizgirev, I.V.; Kozlov, A.P. Evolutionary novel genes are expressed in transgenic fish tumors and their orthologs are involved in development of progressive traits in humans. Infect. Agents Cancer 2019, 14, 46.
  37. Kozlov, A.P.; Matyunina, E.A.; Makashov, A.A. The Biomedical Center TSEEN Genes Database. Certificate about Federal Registration of the Database №2021621840. St.Petersburg, 2021. Available online: https://tseendb.org/#/ (accessed on 7 October 2022).
  38. Kozlov, A.P. The theory of carcino-evo-devo and its non-trivial predictions. Genes 2022, 13, 2347.
  39. Karnaukhova, I.K.; Polev, D.E.; Krukovskaya, L.L.; Makashov, A.A.; Masharsky, A.E.; Nazarenko, O.V.; Poverennaya, I.V.; Makeev, V.J.; Kozlov, A.P. A new cancer/testis long noncoding RNA, the OTP-AS1 RNA. Sci. Rep. 2022; in press.
  40. Abelev, G.I.; Perova, S.; Kramkova, N.I.; Postnikova, Z.; Irlin, I. Embryonal serum alpha-globulin and its synthesis by transplantable mouse hepatomas. Transplant. Bull. 1963, 1, 174–180.
  41. Tatarinov, Y.S. Presence of embryonic alpha-globulin in the serum of patient with primary hepatocellular carcinoma. Vopr. Med. Khim. 1964, 10, 90–91.
  42. Gold, P.; Freedman, S.O. Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques. J. Exp. Med. 1965, 121, 439–462.
  43. Topczewska, J.M.; Postovit, L.M.; Margaryan, N.V.; Sam, A.; Hess, A.R.; Wheaton, W.W.; Nickoloff, B.J.; Topczewski, J.; Hendrix, M.J. Embryonic and tumorigenic pathways converge via Nodal signaling: Role in melanoma aggressiveness. Nat. Med. 2006, 12, 925–932.
  44. Hendrix, M.J.C.; Lurie, R.H. The convergence of embryonic and cancer signaling pathways: Role in tumor cell plasticity. FASEB Exp. Biol. Part 1 2007, 21, A32.
  45. Hendrix, M.J.C.; Seftor, E.A.; Seftor, R.E.B.; Kasemeier-Kulesa, J.; Kulesa, P.M.; Postovit, L.M. Reprogramming metastatic tumor cells with embryonic microenvironment. Nat. Rev. Cancer 2007, 7, 246–255.
  46. Hniz, D.; Schuijers, J.; Lin, C.Y.; Weintraub, A.S.; Abraham, B.J.; Lee, T.I.; Bradner, J.E.; Young, R.A. Covergence of developmental and oncogenic signaling pathways at transcriptional super-enhancers. Mol. Cell 2015, 58, 1–9.
  47. Hall, B.K. Evo-Devo: Evolutionary developmental mechanisms. Int. J. Dev. Biol. 2003, 47, 491–495.
  48. Durante, F. Nessofisio-patologicotra la strutturadeineimaterni e la genesi di alcunitumorimaligni. Arch. Memo. Observ. Chir. Prat. 1874, 11, 217.
  49. Cohnheim, J. Vorlesungen uber Allgemein Pathologie; Hirschwald: Berlin, Germany, 1877.
  50. Cohnheim, J. Lectures on General Pathology: A Handbook for Practitioners and Students; The New Sydenham Society: London, UK, 1889; Volume 2.
  51. Severtsov, A.N. Morphological Laws of Evolution. Collection of Works; Publishing House of the Academy of Sciences of the USSR: Moscow, Leningrad, 1949; Volume 5.
  52. Markert, C.L. Neoplasia: A disease of cell differentiation. Cancer Res. 1968, 28, 1908–1914.
  53. Shimkevich, V.M. Malformations and the Origin of Species; Volf Publishing House: St. Petersburg, Russia; Moscow, Russia, 1909.
  54. Goldschmidt, R. The Material Basis of Evolution; Yale University Press: New Haven, CT, USA, 1940.
  55. Gould, S.J. The return of hopeful monsters. Nat. Hist. 1977, 86, 22–30.
  56. Gould, S.J. Is a new and general theory of evolution emerging? Paleobiology 1980, 6, 119–130.
  57. Anderson, N.G. Evolutionary significance of viral infection. Nature 1970, 227, 1346–1347.
  58. Williams, G.C.; Nesse, R.M. The dawn of Darwinian medicine. Q. Rev. Biol. 1991, 66, 88–100.
  59. Ewald, P.W. Evolution of Infectious Disease; Oxford University Press: New York, NY, USA, 1994.
  60. Cairns, J. Mutation, selection and the natural history of cancer. Nature 1975, 255, 197–200.
  61. Nowell, P.C. The clonal evolution of tumor cell populations. Science 1976, 194, 23–28.
  62. Nowell, P.C. Mechanisms of tumor progression. Cancer Res. 1986, 46, 2203–2207.
  63. Graham, J. Cancer Selection. The New Theory of Evolution; Aculeus Press Inc.: Lexington, KY, USA, 1992; pp. 1–226.
  64. Gatenby, R.A. A change of strategy in the war on cancer. Nature 2009, 459, 508–509.
  65. Kuroiwa, Y.; Ando, R.; Kasahara, K.; Nagatani, M.; Yamakawa, S.; Okazaki, S. Transition of historical control data for high incidence tumors in F344 rats. J. Toxicol. Pathol. 2013, 26, 226–230.
  66. Klaunig, J.E.; Dekant, W.; Plotzke, K.; Scialli, A.R. Biological relevance of decamethylcyclopentasiloxane (D5) induced rat uterine endometrial adenocarcinoma tumorigenesis: Mode of action and relevance to humans. Reg. Toxicol. Pharmacol. 2016, 74, S44–S56.
  67. Knudson, A.G., Jr. Mutation and cancer: Statistical study of retinoblastoma. Proc. Natl. Acad. Sci. USA 1971, 68, 820–823.
  68. Knudson, A.G., Jr. Mutagenesis and embryonal carcinogenesis. Natl. Cancer Inst. Monogr. 1979, 51, 19–24.
  69. Knudson, A.G., Jr. Epidemiology of genetically determined cancer. In Genetic Analysis of Tumour Suppression: Ciba Foundation Symposium 142; Bock, G., Marsh, J., Eds.; Wiley: Chichester, UK, 2007; pp. 3–19.
  70. Sondka, Z.; Bamford, S.; Cole, C.G.; Ward, S.A.; Dunham, I.; Forbes, S.A. The COSMIC cancer gene census: Describing genetic dysfunction across all human cancers. Nat. Rev. Cancer 2018, 18, 696–705.
  71. Tollis, M.; Schneider-Utaka, A.K.; Maley, C.C. The evolution of cancer gene duplications across mammals. Mol. Biol. Evol. 2020, 37, 2875–2886.
  72. Zhu, B.; Mukherjee, A.; Machiela, M.J.; Song, L.; Hua, X.; Shi, J.; Garcia-Closas, M.; Chanock, S.J.; Chatterjee, N. An investigation of genetic susceptibility risk with somatic mutation burden in breast cancer. Br. J. Cancer 2016, 115, 752–760.
  73. Liu, Y.; Gusev, A.; Heng, Y.J.; Alexandrov, L.B.; Kraft, P. Somatic mutational profiles and germline polygenic risk scores in human cancer. Genome Med. 2022, 14, 14.
  74. Leclerc, J.; Flament, C.; Lovecchio, T.; Delattre, L.; Ait Yahya, E.; Baert-Desurmont, S.; Burnichon, N.; Bronner, M.; Cabaret, O.; Lejeune, S.; et al. Diversity of genetic events associated with MLH1 promoter methylation in Lynch syndrome families with heritable constitutional epimutation. Genet. Med. 2018, 20, 1589–1599.
  75. Madsen, T.; Arnal, A.; Vittecoq, M.; Bernex, F.; Abadie, J.; Labrut, S.; Garcia, D.; Faugere, D.; Lemberger, K.; Beckmann, C.; et al. Cancer prevalence and etiology in wild and captive animals. Ecol. Evol. Cancer 2017, 2017, 11–46.
  76. Boddy, A.M.; Abegglen, L.M.; Pessier, A.P.; Aktipis, A.; Schiffman, J.D.; Maley, C.C.; Witte, C. Lifetime cancer prevalence and lifetime history traits in mammals. Evol. Med. Public Health 2020, 2020, 187–195.
  77. Rudan, I.; Rudan, D.; Campbell, H.; Carothers, A.; Wright, A.; Smolej-Narancic, N.; Janicijevic, B.; Jin, L.; Chakraborty, R.; Deka, R.; et al. Inbreeding and risk of late onset complex disease. J. Med. Genet. 2003, 40, 925–932.
  78. Assie, G.; LaFramboise, T.; Platzer, P.; Eng, C. Frequency of germline genomic homozygosity associated with cancer cases. JAMA 2008, 299, 1437–1445.
  79. Garber, J.E.; Offit, K. Hereditary cancer predisposition syndromes. J. Clin. Oncol. 2005, 23, 276–292.
  80. Rahner, N.; Steinke, V. Hereditary cancer syndromes. Dtsch. Arztebl. Int. 2008, 105, 706–714.
  81. Wang, Q. Cancer predisposition genes: Molecular mechanisms and clinical impact on personalized cancer care: Examples of Lynch and HBOC syndromes. Acta Pharmacol. Sin. 2016, 37, 143–149.
  82. Vietri, M.T.; D’Elia, G.; Caliendo, G.; Casamassimi, A.; Federico, A.; Passariello, L.; Cioffi, M.; Molinari, A.M. Prevalence of mutations in BRCA and MMR genes in patients affected with hereditary endometrial cancer. Med. Oncol. 2021, 38, 13.
  83. Ryan, P.D. Genetics of hereditary breast cancer. In Principles of Clinical Cancer Genetics; Chung, D., Haber, D., Eds.; Springer: Boston, MA, USA, 2010; pp. 41–51.
  84. Sokolenko, A.P.; Imyanitov, E.N. Molecular diagnostics in clinical oncology. Front. Mol. Biosci. 2018, 5, 76.
  85. Aktipis, C.A.; Boddy, A.M.; Jansen, G.; Hibner, U.; Hochberg, M.E.; Maley, C.C.; Wilkinson, G.S. Cancer across the tree of life: Cooperation and cheating in multicellularity. Philos. Trans. R. Soc. B Biol. Sci. 2015, 370, 20140219.
  86. Albuquerque, T.A.F.; Drummond do Val, L.; Doherty, A.; Pedro de Magalhaes, J. From humans to hydra: Patterns of cancer across the tree of life. Biol. Rev. 2018, 93, 1715–1734.
  87. Boutry, J.; Tissot, S.; Ujvari, B.; Capp, J.P.; Giraudeau, M.; Nedelcu, A.M.; Thomas, F. The evolution and ecology of benign tumors. BBA—Rev. Cancer 2022, 1877, 188643.
  88. Satarkar, R.N.; Srikanth, S. Tumors and tumor-like conditions of the nasal cavity, paranasal sinuses, and nasopharynx: A study of 206 cases. Indian J. Cancer 2016, 53, 478–482.
  89. Clark, W.H. Tumour progression and the nature of cancer. Br. J. Cancer 1991, 64, 631–644.
  90. Vorontsov, N.N. Macromutations and evolution: Fixation of Goldschmidt’s macromutations as species and genus characters. Papillomatosis and appearance of macrovilli in the rodent stomach. Russ. J. Genet. 2003, 39, 422–426.
  91. Kozlov, A.P.; Zabezhinski, M.A.; Popovich, I.G.; Polev, D.E.; Shilov, E.S.; Murashev, B.V. Hyperplastic skin growth on the head of goldfish--comparative oncology aspects. Vopr. Onkol. 2012, 58, 387–393.
  92. Voskarides, K. Combination of 247 genome-wide association studies reveals high cancer risk as a result of evolutionary adaptation. Mol. Biol. Evol. 2017, 35, 473–485.
  93. Voskarides, K.; Dweep, H.; Chrysostomou, C. Evidence that DNA repair genes, a family of tumor suppressor genes, are associated with evolution rate and size of genomes. Hum. Genom. 2019, 13, 26.
  94. Dobzhansky, T. Nothing in biology makes sense except in the light of evolution. Am. Biol. Teach. 1973, 35, 125–129.
  95. Greaves, M. Cancer. The Evolutionary Legacy; Oxford University Press: Oxford, MS, USA, 2000; pp. 1–288.
  96. Aktipis, A. The Cheating Cell: How Evolution Helps Us Understand and Treat Cancer; Princeton University Press: Princeton, NJ, USA, 2020; pp. 1–256.
More
Information
Subjects: Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 249
Revisions: 2 times (View History)
Update Date: 18 May 2023
1000/1000