Current Issues in Carcinogenesis

IV Vysotskaya1, VP Letyagin2, MA Shabanov2, VYu Kirsanov1, EA Kim1, NV Levkina1

1 IM Sechenov First Moscow State Medical University, 8 bld. 2 Trubetskaya str., Moscow, Russian Federation, 119991

2 NN Blokhin National Medical Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Prof. Irina Viktorovna Vysotskaya, MD, PhD, 8 bld. 2 Trubetskaya str., Moscow, Russian Federation, 119991; e-mail: vysotskaya.irina@mail.ru

For citation: Vysotskaya IV, Letyagin VP, Shabanov MA, et al. Current Issues in Carcinogenesis. Clinical oncohematology. 2019;12(1):101–6.

DOI: 10.21320/2500-2139-2019-12-1-101-106


ABSTRACT

The review presents current data on the major pathogenetic mechanisms underlying uncontrolled growth and dissemination of tumor and its resistance to conventional treatment. Cell genetic instability associated with accumulation of mutations in genes controlling cell growth and differentiation is a key factor in tumor proliferation. Due understanding and detailed analysis of carcinogenesis processes provide the basis for creation of new anticancer drugs which in turn enables optimization and individualization of cancer treatment.

Keywords: carcinogenesis, initiation, promotion, repair, proto-oncogenes, suppressor genes, Ras, TP53, targeted therapy.

Received: June 27, 2018

Accepted: December 20, 2018

Read in PDF 


REFERENCES

  1. Лихтенштейн А.В., Шапот В.С. Опухолевый рост: ткани, клетки, молекулы. Патологическая физиология. 1997;3:35–48.

    [Likhtenshtein AV, Shapot VS. Tumor growth: tissues, cells, molecules. Patologicheskaya fiziologiya. 1997;3:35–48. (In Russ)]

  2. Вельшер Л.З., Матякин Е.Г., Дудицкая Т.К., Поляков Б.И. Онкология: учебник для вузов. М.: ГЭОТАР-Медиа, 2009. 512 с.

    [Vel’sher LZ, Matyakin EG, Duditskaya TK, Polyakov BI. Onkologiya: uchebnik dlya vuzov. (Oncology: textbook for universities.) Moscow: GEOTAR-Media Publ.; 512 p. (In Russ)]

  3. Имянитов Е.Н., Хансон К.П. Молекулярная онкология: клинические аспекты. СПб.: МАПО, 2007. 211 с.

    [Imyanitov EN, Khanson KP. Molekulyarnaya onkologiya: klinicheskie aspekty. (Molecular oncology: clinical aspects.) Saint Petersburg: MAPO Publ.; 2007. 211 p. (In Russ)]

  4. Черезов А.Е. Общая теория рака: тканевый подход. М.: Изд-во МГУ, 1997. 252 с.

    [Cherezov AE. Obshchaya teoriya raka: tkanevyi podkhod. (General theory of cancer: tissue approach.) Moscow: MGU Publ.; 1997. 252 p. (In Russ)]

  5. Рукавишников А.И. Азбука рака. Волгоград: Изд-во ВГМУ, 360 с.

    [Rukavishnikov AI. Azbuka raka. (The ABCs of cancer.) Volgograd: VGMU Publ.; 2007. 360 p. (In Russ)]

  6. Лекции по общей патологической анатомии. Под ред. М.А. Пальцева. М., 2003. 254 с.

    [Pal’tsev MA, ed. Lektsii po obshchei patologicheskoi anatomii. (Lectures on general pathological anatomy.) Moscow; 20 254 p. (In Russ)]

  7. Ермоленко А.Е. Этиологическая классификация опухолей и механизмы канцерогенеза. Математическая морфология: электронная математика и медико-биологический журнал. 2012;1(2):24–34.

    [Ermolenko AE. The etiological classification of tumors and the mechanisms of carcinogenesis. Matematicheskaya morfologiya: Elektronnyi matematicheskii i mediko-biologicheskii zhurnal. 2012;1(2):24–34. (In Russ)]

  8. Патологоанатомическая диагностика опухолей человека: руководство в 2 томах. Под ред. Н.А. Краевского, А.В. Смольянникова, Д.С. Саркисова. М.: Медицина, 1993.

    [Kraevskii, NA, Smol’yannikov AV, Sarkisov DS, Patologoanatomicheskaya diagnostika opukholei cheloveka: rukovodstvo v 2 tomakh (Pathology diagnostics of human tumors: guidelines in 2 volumes.) Moscow: Meditsina Publ.; 1993. (In Russ)]

  9. Knudson AG. Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA. 1971;68(4):820–3. doi: 10.1073/pnas.68.4.820.

  10. Гарькавцева Р.Ф., Гарькавцев И.В. Молекулярно-генетические аспекты злокачественных новообразований. Вестник РАМН. 1999;2:38–44.

    [Gar’kavtseva RF, Gar’kavtsev IV. Molecular genetic aspects of malignant neoplasms. Vestnik RAMN. 1999;2:38–44. (In Russ)]

  11. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability – an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 2010;11(3):220–8. doi: 1038/nrm2858.

  12. Павлов В.Н., Измайлов А.А., Викторова Т.В. и др. Генетические факторы риска развития рака мочевого пузыря. Экспериментальная и клиническая урология. 2010;2:25–31.

    [Pavlov VN, Izmailov AA, Viktorova TV, et al. Genetic risk factors in the development of bladder cancer. Eksperimental’naya i klinicheskaya urologiya. 2010;2:25–31. (In Russ)]

  13. Webb СР, Vande Woude GF. Genes that regulate metastases and angiogenesis. J Neurooncol. 2000;50(1–2):71–87.

  14. Новик А.А., Новик Т.А., Камилова А.А., Цыган В.Н. Генетика в клинической медицине. СПб.: ВМедА, 2001. 219 с.

    [Novik AA, Novik TA, Kamilova AA, Tsygan VN. Genetika v klinicheskoi meditsine. (Genetics in clinical medicine.) Saint Petersburg: VMedA Publ.; 2001. 219 p. (In Russ)]

  15. Аничков Н.М., Плотникова Н.А. О морфологии и классификации опухолеподобных и раковых поражений предстательной железы. Архив патологии. 2001;63(5):44–50.

    [Anichkov NM, Plotnikova NA. On the morphology and classification of prostate tumors and cancerous lesions. Arkhiv patologii. 2001;63(5):44–50. (In Russ)]

  16. Лихтенштейн А.В. Исследования рака: бег с препятствиями. Биохимия. 2014;79(5):493–500.

    [Likhtenshtein AV. Cancer research: a hurdle race. Biokhimiya. 2014;79(5):493–500. (In Russ)]

  17. Копнин Б.П. Мишени действия онкогенов и опухолевых супрессоров: ключ к пониманию базовых механизмов канцерогенеза. Биохимия. 2000;65(1):5–33.

    [Kopnin BP. Targets of oncogenes and tumor suppressors: the key to understanding basic mechanisms of carcinogenesis. 2000;65(1):5–33. (In Russ)]

  18. Рак предстательной железы. Под ред. Н.Е. Кушлинского, Ю.Н. Соловьева, М.Ф. Трапезниковой. М.: Изд-во РАМН, 2002. 432 c.

    [Kushlinskii NE, Solov’ev YuN, Trapeznikova MF, eds. Rak predstatel’noi zhelezy. (Prostate cancer.) Moscow: RAMN Publ.; 432 р. (In Russ)]

  19. Кушлинский Н.Е., Немцова М.В. Молекулярно-биологические характеристики злокачественных новообразований. Вестник РАМН. 2014;69(1–2):5–15. doi: 10.15690/vramn.v69i1-2.934.

    [Kushlinskii NE, Nemtsova MV. Molecular biological characteristics of cancer. Annals of the Russian academy of medical sciences. 2014;69(1–2):5–15. doi: 10.15690/vramn.v69i1-2.934. (In Russ)]

  20. Зильбер Л.А. Специфические антигены опухолей. Успехи в изучении рака. М.: Изд-во иностранной литературы, 1960. С. 186–277.

    [Zil’ber LA. Spetsificheskie antigeny opukholei. Uspekhi v izuchenii raka. (Tumor-specific antigens. Advances in cancer research.) Moscow: Inostrannaya Literatura Publ.; рр. 186–277. (In Russ)]

  21. Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 2007;7(11):834–46. doi: 11038/nrc2256.

  22. Coghlin C, Murray GI. Current and emerging concepts in tumour metastasis. J Pathol. 2010;222(1):1–15. doi: 1002/path.2727.

  23. Egeblad M, Nakasone ES, Werb Z. Tumors as organs: complex tissues that interface with the entire organism. Dev Cell. 2010;18(6):884–901. doi: 1016/j.devcel.2010.05.012.

  24. Williams GM. Mechanisms of chemical carcinogenesis and application in human cancer risk assessment. Toxicology. 2001;166(1–2):3–10. doi:1016/s0300-483x(01)00442-5.

  25. Аничков Н.М. Биологические и клинико-морфологические аспекты учения о метастазировании злокачественных опухолей. Медицинский академический журнал. 2003;1:3–13.

    [Anichkov NM. Biological and morphological aspects of the doctrine of metastasis of malignant tumors. Meditsinskii akademicheskii zhurnal. 2003;1:3–13. (In Russ)]

  26. Худолей В.В. Канцерогены: характеристики, закономерности, механизмы действия. СПб.: Изд-во СПбГУ, 1999. 419 с.

    [Khudolei VV. Kantserogeny: kharakteristiki, zakonomernosti, mekhanizmy deistviya. (Carcinogens: characteristics, patterns, mechanisms of action.) Saint Petersburg: SPbGU Publ.; 1999. 419 p. (In Russ)]

  27. Aльтштейн А.Д. Вирусный канцерогенез и роль вирусов в возникновении опухолей человека. В кн.: Канцерогенез. Под ред. Д.Г. Заридзе. М.: Медицина, 2004. С. 251–74.

    [Al’tshtein AD. Viral carcinogenesis and the role of viruses in the occurrence of human tumors. In: Zaridze DG, ed. Kantserogenez. (Carcinogenesis.) Moscow: Meditsina Publ.; 2004. рр. 251–74. (In Russ)]

  28. Киселев Ф.Л. Онкогенный потенциал вирусов и механизмы его проявления. В кн.: Канцерогенез. Под ред. Д.Г. Заридзе. М.: Медицина. С. 274–87.

    [Kiselev FL. Oncogenic potential of viruses and mechanisms of its manifestation. In: Zaridze DG, ed. Kantserogenez. (Carcinogenesis.) Moscow: Meditsina Publ.; 2004. рр. 274–87. (In Russ)]

  29. Hausen H. Infections causing human cancers. New York: Wiley-VCH, Weinheim; 2006. 517 p.

  30. Fernandez AF, Rosales C, Lopez-Nieva P. et al. The dynamic DNA methylomes of double-stranded DNA viruses associated with human cancer. Genome Res. 2009;19(3):438–51. doi: 1101/gr.083550.108.

  31. Fernandez A, Esteller M. Viral epigenomes in human tumorigenesis. Oncogene. 2010;29(10):1405–20. doi: 1038/onc.2009.517.

  32. Junttila MR, Evan GL. p53 – a Jack of all trades but master of none. Nat Rev Cancer. 2009;9(11);821–9. doi: 1038/nrc2728.

  33. Bertram JS. The molecular biology of cancer. Mol Aspects Med. 2001;21(6):167–223. doi: 10.1016/s0098-2997(00)00007-8.

  34. Плешкан В.В., Алексеенко И.В., Зиновьева М.В. и др. Промоторы со специфической активностью в раковых клетках при генной терапии меланомы. Acta Naturae. 2011;3(2):14–23.

    [Pleshkan VV, Alekseenko IV, Zinov’eva MV. Promoters with cancer cell-specific activity for melanoma gene therapy. Acta Naturae. 2011;3(2):14–23. (In Russ)]

  35. Burdall SE, Hanby AM, Lansdown MR, Speirs V. Breast cancer cell lines: friend or foe? Breast Cancer Res. 2003;5(2):89–95. doi: 10.1186/bcr577.

  36. Ковалева О.В., Назарова О.Р., Матвеев В.Б., Грачев А.Н. Молекулярные особенности почечно-клеточного рака: ранняя диагностика и перспективы терапии. Успехи молекулярной онкологии. 2014;1(2):36–43.

    [Kovaleva OV, Nazarova OR, Matveev VB, Grachev AN. Molecular features of renal cell carcinoma: early diagnosis and perspectives for therapy. Uspekhi molekulyarnoi onkologii. 2014;1(2):36–43. (In Russ)]

  37. Berger AH, Knudson AG, Pandolfi PP. A continuum model for tumour suppression. 2011;476(7359):163–9. doi: 10.1038/nature10275.

  38. Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 2009;23(5):537–48. doi: 1101/gad.1756509.

  39. Berdasco M, Esteller M. Aberrant epigenetic landscape in cancer: how cellular identity goes awry. Dev Cell. 2010;19(5):698–711. doi: 1016/j.devcel.2010.10.005.

  40. Артамонова Е.В., Огнерубов Н.А., Тупицын Н.Н., Летягин В.П. Рак молочной железы: иммунологические факторы прогноза. Воронеж: Изд-во ВГУ, 2005. 240 с.

    [Artamonova EV, Ognerubov NA, Tupitsyn NN, Letyagin VP. Rak molochnoi zhelezy: immunologicheskie faktory prognoza. (Breast cancer: immunological prognostic) Voronezh: VGU Publ.; 2005. 240 p. (In Russ)]

  41. Артамонова Е.В. Иммунологическая микрогетерогенность РМЖ: Дис.… канд. мед. наук. М., 1992.

    [Artamonova EV. Immunologicheskaya mikrogeterogennost’ raka molochnoi zhelezy. (Immunological microheterogeneity of breast cancer.) [dissertation] Moscow; 1992. (In Russ)]

  42. Тешелова В.Т. Канцерогенез и активация периферических лимфоцитов. Успехи современной биологии. 2003;123(5):495–505.

    [Teshelova VT. Carcinogenesis and activation of peripheral lymphocytes. Uspekhi sovremennoi biologii. 2003;123(5):495–505. (In Russ)]

  43. Bindea G, Mlecnik B, Fridman WH. Natural immunity to cancer in humans. Curr Opin Immunol. 2010;22(2):215–22. doi: 1016/j.coi.2010.02.006.

  44. Grivennikov S, Greten FR, Karin M. Immunity, inflammation and cancer. Cell. 2010;140(6):883–99. doi: 1016/j.cell.2010.01.025.

  45. Mougiakakos D, Choudhury A, Lladser A, et al. Regulatory T cells in cancer. Adv Cancer Res. 2010;107:57–117. doi: 1016/S0065-230X(10)07003-X.

  46. Brabletz T, Jung A, Spaderna S, et al. Migrating cancer stem cells – an integrated concept of malignant tumour progression. Nat Rev Cancer. 2005;5(9):744–9. doi: 1038/nrc1694.

  47. Gupta PB, Chaffer CL, Weinberg RA. Cancer stem cells: mirage or reality? Nat Med. 2009;15(9):1010–2. doi: 1038/nm0909-1010.

  48. Semenza GL. Tumor metabolism: cancer cells give and take lactate. J Clin Invest. 2008;118(12):3835–7. doi: 1172/JCI37373.

  49. Sleeman JP. The metastatic niche and stromal progression. Cancer Metast Rev. 2012;31(3–4):429–40. doi: 1007/s10555-012-9373-9.

  50. Straus DC, Thomas JM. Transmission of donor melanoma by organ transplantation. Lancet Oncol. 2010;11(8):790–6. doi: 1016/S1470-2045(10)70024-3.

  51. Gazdar AF, Girard L, Lockwood WW, et al. Lung cancer cell lines as tools for biomedical discovery and research. J Natl Cancer Inst. 2010;102(17):1310–21. doi: 1093/jnci/djq279.

Autophagy: cell death or survival strategy?

O.V. Kovaleva, M.S. Shitova, and I.B. Zborovskaya

N.N. Blokhin Russian Cancer Research Center, RAMS, Moscow, Russian Federation


ABSTRACT

The interaction between proliferation, differentiation, and programmed cell death is an integral part of the life-sustaining activity of multicellular organisms. The imbalance between these processes underlies the development of many human diseases. Understanding molecular mechanisms of these processes is important for identifying new diagnostic and therapeutic targets. During the last decade, the autophagy processed and its role in the cell life and death became a subject of great scientific interest. Autophagy represents a mechanism of intracellular substance utilization. Autophagy is a process that occurs in all cells under normal conditions. But under certain circumstances, this process can result in the cell death. Impaired autophagy significantly contributes into the development of some diseases (cancer, neurodegenerative and cardiovascular disorders etc.).


Keywords: apoptosis, autophagy, carcinogenesis

Read in PDF (RUS)pdficon


REFERENCES

  1. Galluzzi L., Vitale I., Abrams J.M. et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ. 2012; 19(1): 107–20.
  2. Kerr J.F., Wyllie A.H., Currie A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972; 26: 239–57.
  3. Wong R.S. Apoptosis in cancer: from pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011; 26: 30–87.
  4. Hu Y., Benedict M.A., Ding L., Nunez G. Role of cytochrome c and dATP/ ATP hydrolysis in Apaf-1-mediated caspase-9 activation and apoptosis. EMBO J. 1999; 18: 3586–95.
  5. Saelens X., Festjens N., Vande Walle L. et al. Toxic proteins released from mitochondria in cell death. Oncogene 2004; 23(16): 2861–74.
  6. Altieri D.C. Surviving in apoptosis control and cell cycle regulation in cancer. Prog. Cell Cycle Res. 2003; 5: 447–52.
  7. Tamm I., Wang Y., Sausville E. et al. IAP-family protein surviving inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res. 1998; 58(23): 5315–20.
  8. Padanilam B.J. Cell death induced by acute renal injury: a perspective on the contributions of apoptosis and necrosis. Am. J. Physiol. Renal. Physiol. 2003; 284(4): F608–27.
  9. Hoste E., Kemperman P., Devos M. et al. Caspase-14 is required for filaggrin degradation to natural moisturizing factors in the skin. J. Invest. Dermatol. 2011; 131(11): 2233–41.
  10. Levine B., Klionsky D.J. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev. Cell 2004; 6(4): 463–77.
  11. Okada H., Mak T.W. Pathways of apoptotic and non-apoptotic death in tumor cells. Nat. Rev. Cancer 2004; 4(8): 592–603.
  12. Kaminskyy V., Zhivotovsky B. Proteases in autophagy. Biochimica er Biophysica Acta. 2012; 1824: 44–50.
  13. Mijaljica D., Prescott M., Devenish R.J. Microautophagy in mammalian cells: Revisiting a 40-year-old conundrum. Autophagy 2011; 7: 673–82.
  14. Klionsky DJ., Codogno P., Cuervo A.M. et al. A comprehensive glossary of autophagy-related molecules and processes. Autophagy 2010; 6(4): 438–48.
  15. Massey A.C., Zhang C., Cuervo A.M. Chaperone-mediated autophagy in aging and disease. Curr. Top. Dev. Biol. 2006; 73: 205–35.
  16. Kimmelman A.C. The dynamic nature of autophagy in cancer. Genes Dev. 2011; 25(19): 1999–2010.
  17. Reggiori F., Tucker K.A., Stromhaug P.E., Klionsky D.J. The Atg1-Atg13 complex regulates Atg9 and Atg23 retrieval transport from the pre-autophagosomal structure. Dev. Cell 2004; 6(1): 79–90.
  18. Wang C.W., Klionsky D.J. The molecular mechanism of autophagy. Mol. Med. 2003; 9: 65–76.
  19. Teter S.A., Klionsky D.J. Transport of proteins to the yeast vacuole: autophagy, cytoplasm-to-vacuole targeting, and role of the vacuole in degradation. Semin. Cell Dev. Biol. 2000; 11(3): 173–9.
  20. Chen N., Debnath J. Autophagy and Tumorigenesis. FEBS Lett. 2010; 584(7): 1427–35.
  21. Guertin D.A., Sabatini D.M. Defining the role of mTOR in cancer. Cancer Cell 2007; 12: 9–22.
  22. He C., Klionsky D.J. Regulation Mechanisms and Signaling Pathways of Autophagy. Annu. Rev. Genet. 2009; 43: 67–93.
  23. Zhou H., Huang S. The complexes of mammalian target of rapamycin. Curr. Protein Pept. Sci. 2010; 11: 409–24.
  24. Teter T., Hall M.N. TOR, a central controller of cell growth. Cell 2000; 103(2): 253–62.
  25. Tee A.R., Manning B.D., Roux P.P., Cantley L.C., Blenis J. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr. Biol. 2003; 13: 1259–68.
  26. Shackelford D.B., Shaw R.J. The LKB1-AMPK pathway: metabolism and growth control in tumor suppression. Nat. Rev. Cancer 2009; 9: 563–75.
  27. Liang J., Shao SH., Xu Z.X. et al. The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis. Nat. Cell Biol. 2007; 9: 218–24.
  28. Feng Z., Hu W., Stanchina E. et al. The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res. 2007; 67: 3043–53.
  29. Mortensen M., Watson A.S., Simon A.K. Lack of autophagy in the hematopoietic system leads to loss of hematopoietic stem cell function and dysregulated myeloid proliferation. Autophagy 2011; 7(9): 1069–70.
  30. Mortensen M., Soilleux E.J., Djordjevic G. et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 2011; 208: 455–67.
  31. Pua H.H., Komatsu M., He Y.W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J. Immunol. 2009; 182: 4046–55.
  32. Pua H.H., Dzhagalov I., Chuck M. et al. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 2007; 204: 25–31.
  33. Pua H.H., He Y.W. Maintaining T lymphocyte homeostasis: another duty of autophagy. Autophagy 2007; 3: 266–7.
  34. Miller B.C., Zhao Z., Stephenson L.M. et al. The autophagy gene Atg5 plays an essential role in B lymphocyte development. Autophagy 2008; 4: 309–14.
  35. Novak I., Kirkin V., McEwan D.G. et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010; 11(1): 45–51.
  36. Kundu M., Lindsten T., Yang C.Y. et al. Ulk1 plays a critical role in the autophagic clearance of mitochondria and ribosomes during reticulocyte maturation. Blood 2008; 112: 1493–502.
  37. Raslova H., Kauffmann A., Sekkai D. et al. Interrelation between polyploidization and megakaryocyte differentiation: a gene profiling approach. Blood. 2007; 109: 3225–34.
  38. Delgado M.A., Elmaoued R.A., Davis A.S., Kyei G., Deretic V. Toll-like receptors control autophagy. EMBO J. 2008; 27: 1110–21.
  39. Shi C.S., Kehrl J.H. MyD88 and Trif target Beclin 1 to trigger autophagy in macrophages. J. Biol. Chem. 2008; 283: 33175–82.
  40. Xu Y., Jagannath C., Liu X.D. et al. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity. 2007; 27: 135–44.
  41. Канцерогенез. Под ред. Д.Г. Заридзе. М.: Медицина, 2004. [Kantserogenez. Pod red. D.G. Zaridze (Carcinogenesis. Ed. by: D.G. Zaridze). M.: Meditsina, 2004.]
  42. Liang X.H., Jackson S., Seaman M. et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999; 402: 672–6.
  43. Yue Z., Jin S., Yang C., Levine A.J., Heintz N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. U S A 2003; 100: 15077–82.
  44. Qu X., Yu J., Bhagat G. et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Invest. 2003; 112: 1809–20.
  45. Karantza-Wadsworth V., Patel S., Kravchuk O. et al. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 2007; 21: 1621–35.
  46. Meek D.W. Tumor suppression by p53: a role for the DNA damage response? Nat. Rev. Cancer 2009; 9: 714–23.
  47. Degenhardt K., Mathew R., Beaudoin B. et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006; 10: 51–64.
  48. DeNardo D., Johansson M., Coussens L. Immune cells as mediators of solid tumor metastasis. Cancer Metast. Rev. 2008; 27: 11–8.
  49. DeNardo D.G., Barreto J.B., Andreu P. et al. CD4+ T Cells Regulate Pulmonary Metastasis of Mammary Carcinomas by Enhancing Protumor Properties of Macrophages. Cancer Cell 2009; 16: 91–102.
  50. Bingle L., Brown N.J., Lewis C.E. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J. Pathol. 2002; 196: 254–65.
  51. Young A.R., Narita M., Ferreira M. et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009; 23: 798–803.
  52. Petiot A., Ogier-Denis E., Blommaart E.F., Meijer AJ., Codogno P. Distinct classes of phosphatidylinositol 3¢-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J. Biol. Chem. 2000; 275: 992–8.
  53. Luiken J.J., Aerts J.M., Meijer A.J. The role of the intralysosomal pH in the control of autophagic proteolytic flux in rat hepatocytes. Eur. J. Biochem. 1996; 235: 564–73.
  54. Yamamoto A., Tagawa Y., Yoshimori T. et al. Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E cells. Cell Struct. Funct. 1998; 23: 33–42.
  55. Ito H., Daido S., Kanzawa T., Kondo S., Kondo Y. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int. J. Oncol. 2005; 26: 1401–10.
  56. Lomonaco S.L., Finniss S., Xiang C. et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int. J. Cancer 2009; 125: 717–22.
  57. Shingu T., Fujiwara K., Bogler O. et al. Stage-specific effect of inhibition of autophagy on chemotherapy-induced cytotoxicity. Autophagy 2009; 5: 537–9.
  58. Vazquez-Martin A., Oliveras-Ferraros C., Menendez J.A. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. PLoS One 2009; 4: e6251.
  59. Abedin M.J., Wang D., McDonnell M.A., Lehmann U., Kelekar A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2007; 14: 500–10.
  60. Kim R.H., Coates J.M., Bowles T.L. et al. Arginine deiminase as a novel therapy for prostate cancer induces autophagy and caspase-independent apoptosis. Cancer Res. 2009; 69: 700–8.
  61. Bellodi C., Lidonnici M.R., Hamilton A. et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosomepositive cells, including primary CML stem cells. J. Clin. Invest. 2009; 119: 1109–23.
  62. Carew J.S., Nawrocki S.T., Kahue C.N. et al. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 2007; 10: 313–22.
  63. Ertmer A., Huber V., Gilch S. et al. The anticancer drug imatinib induces cellular autophagy. Leukemia 2007; 21: 936–42.
  64. Kamitsuji Y., Kuroda J., Kimura S. et al. The Bcr-Abl kinase inhibitor INNO-406 induces autophagy and different modes of cell death execution in Bcr-Abl-positive leukemias. Cell Death Differ. 2008; 15: 1712–22.
  65. Goussetis D.J., Altman J.K., Glaser H. et al. Autophagy is a critical mechanism for the induction of the antileukemic effects of arsenic trioxide. J. Biol. Chem. 2010; 285: 29989–97.
  66. Qian W., Liu J., Jin J. et al. Arsenic trioxide induces not only apoptosis but also autophagic cell death in leukemia cell lines via up-regulation of Beclin-1. Leuk. Res. 2007; 31: 329–39.
  67. Charoensuk V., Gati WP., Weinfeld M. et al. Differential cytotoxic effects of arsenic compounds in human acute promyelocytic leukemia cells. Toxicol. Appl. Pharmacol. 2009; 239: 64–70.
  68. Chiarini F., Grimaldi C., Ricci F. et al. Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVPBEZ235 against T-cell acute lymphoblastic leukemia. Cancer Res. 2010; 70: 8097–107.
  69. Crazzolara R., Bradstock K.F., Bendall L.J. RAD001 (everolimus) induces autophagy in acute lymphoblastic leukemia. Autophagy 2009; 5: 727–8.
  70. Crazzolara R., Cisterne A., Thien M. et al. Potentiating effects of RAD001 (everolimus) on vincristine therapy in childhood acute lymphoblastic leukemia. Blood 2009; 113: 3297–306.
  71. Puissant A., Auberger P. AMPK- and p62/SQSTM1-dependent autophagy mediate resveratrol-induced cell death in chronic myelogenous leukemia. Autophagy 2010; 6(5): 655–7.
  72. Puissant A., Robert G., Fenouille N. et al. Resveratrol promotes autophagic cell death in chronic myelogenous leukemia cells via JNK mediated p62/SQSTM1 expression and AMPK activation. Cancer Res. 2010; 70: 1042–52.
  73. Kroemer G., Levine B. Autophagic cell death: the story of a misnomer. Nat. Rev. Mol. Cell Biol. 2008; 9: 1004–10.
  74. Turcotte S., Chan D.A., Sutphin P.D. et al. A molecule targeting VHLdeficient renal cell carcinoma that induces autophagy. Cancer Cell. 2008; 14: 90–102.
  75. Kanzawa T., Germano I.M., Komata T. et al. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ. 2004; 11: 448–57.