Chronic Lymphocytic Leukemia: A Genomic Landscape and Cellular Signal Pathways
ISSN (print) 1997-6933     ISSN (online) 2500-2139
2025-1
PDF_2025-18-1-21-37 (Russian)

Keywords

chronic lymphocytic leukemia
signaling pathway
molecular genetics
epigenetics

How to Cite

Mikhaleva M.A., Kunevich E.O., Kuvshinov A.Y., Voloshin S.V. Chronic Lymphocytic Leukemia: A Genomic Landscape and Cellular Signal Pathways. Clinical Oncohematology. 2025;(1):21–37. doi:10.21320/2500-2139-2025-18-1-21-37.

Keywords

Abstract

Genomic diagnostic methods, developed and improved in recent decades, allow a deeper understanding of pathogenic diversity of chronic lymphocytic leukemia (CLL) across all aspects from the concepts of tumor cellular origin and its reactive microenvironment to molecular landscape and genetic prognostic markers. This review discusses clinically significant molecular genetic abnormalities to be considered for risk stratification of CLL patients and personalized treatment decision making. It provides a current view of molecular landscape of CLL including information on cell signaling mechanisms and clinically significant biomarkers. This review also focuses on heterogeneity of CLL clinical course reflecting biological events at the multi-omics level: genome, epigenome, transcriptome, proteome, and metabolome. It also covers the latest technologies and emphasizes the relevance of multi-omic profiling for giving rise to new CLL subclassifications.

PDF_2025-18-1-21-37 (Russian)

References

  1. 1. Hallek M. Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am J Hematol. 2019;94(11):1266–87. doi: 10.1002/ajh.25595.
  2. 2. Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood. 2018;131(25):2745–60. doi: 10.1182/blood-2017-09-806398.
  3. 3. Chiorazzi N, Rai KR, Ferrarini M. Chronic lymphocytic leukemia. N Engl J Med. 2005;352(8):804–15. doi: 10.1056/NEJMra041720.
  4. 4. Klein U, Dalla-Favera R. New insights into the phenotype and cell derivation of B cell chronic lymphocytic leukemia. Curr Top Microbiol Immunol. 2005;294:31–49. doi: 10.1007/3-540-29933-5_3.
  5. 5. Stevenson FK, Caligaris-Cappio F. Chronic lymphocytic leukemia: revelations from the B-cell receptor. Blood. 2004;103(12):4389–95. doi: 10.1182/blood-2003-12-4312.
  6. 6. Swerdlow SH, Campo E, Harris NL, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th ed. Lyon: IARC Press; 2008.
  7. 7. Bosch F, Dalla-Favera R. Chronic lymphocytic leukaemia: from genetics to treatment. Nat Rev Clin Oncol. 2019;16(11):684–701. doi: 10.1038/s41571-019-0239-8.
  8. 8. Klein U, Tu Y, Stolovitzky GA, et al. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med. 2001;194(11):1625–38. doi: 10.1084/jem.194.11.1625.
  9. 9. Seifert M, Sellmann L, Bloehdorn J, et al. Cellular origin and pathophysiology of chronic lymphocytic leukemia. J Exp Med. 2012;209(12):2183–98. doi: 10.1084/jem.20120833.
  10. 10. Kulis M, Heath S, Bibikova M, et al. Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia. Nat Genet. 2012;44(11):1236–42. doi: 10.1038/ng.2443.
  11. 11. Ferrer G, Montserrat E. Critical molecular pathways in CLL therapy. Mol Med. 2018;24(1):9. doi: 10.1186/s10020-018-0001-1.
  12. 12. Ten Hacken E, Burger JA. Microenvironment interactions and B-cell receptor signaling in Chronic Lymphocytic Leukemia: Implications for disease pathogenesis and treatment. Biochim Biophys Acta. 2016;1863(3):401–13. doi: 10.1016/j.bbamcr.2015.07.009.
  13. 13. O’Donnell A, Pepper C, Mitchell S, Pepper A. NF-kB and the CLL microenvironment. Front Oncol. 2023;13:1169397. doi: 10.3389/fonc.2023.1169397.
  14. 14. Juliusson G, Oscier DG, Fitchett M, et al. Prognostic subgroups in B-cell chronic lymphocytic leukemia defined by specific chromosomal abnormalities. N Engl J Med. 1990;323(11):720–4. doi: 10.1056/NEJM199009133231105.
  15. 15. Dohner H, Stilgenbauer S, Benner A, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343(26):1910–6. doi: 10.1056/NEJM200012283432602.
  16. 16. Puiggros A, Blanco G, Espinet B. Genetic abnormalities in chronic lymphocytic leukemia: where we are and where we go. Biomed Res Int. 2014;2014:435983. doi: 10.1155/2014/435983.
  17. 17. Klein U, Lia M, Crespo M, et al. The DLEU2/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia. Cancer Cell. 2010;17(1):28–40. doi: 10.1016/j.ccr.2009.11.019.
  18. 18. Kasar S, Salerno E, Yuan Y, et al. Systemic in vivo lentiviral delivery of miR-15a/16 reduces malignancy in the NZB de novo mouse model of chronic lymphocytic leukemia. Genes Immun. 2012;13(2):109–19. doi: 10.1038/gene.2011.58.
  19. 19. Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proceedings of the National Academy of Sciences. 2005;102(39):13944–9. doi: 10.1073/pnas.0506654102.
  20. 20. Jeromin S, Weissmann S, Haferlach C, et al. SF3B1 mutations correlated to cytogenetics and mutations in NOTCH1, FBXW7, MYD88, XPO1 and TP53 in 1160 untreated CLL patients. Leukemia. 2014;28(1):108–17. doi: 10.1038/leu.2013.263.
  21. 21. Larrayoz M, Rose-Zerilli MJJ, Kadalayil L, et al. Non-coding NOTCH1 mutations in chronic lymphocytic leukemia; their clinical impact in the UK CLL4 trial. Leukemia. 2017;31(2):510–4. doi: 10.1038/leu.2016.298.
  22. 22. Brown JR, Hanna M, Tesar B, et al. Integrative genomic analysis implicates gain of PIK3CA at 3q26 and MYC at 8q24 in chronic lymphocytic leukemia. Clin Cancer Res. 2012;18(14):3791–802. doi: 10.1158/1078-0432.CCR-11-2342.
  23. 23. Zenz T, Mertens D, Kuppers R, et al. From pathogenesis to treatment of chronic lymphocytic leukaemia. Nat Rev Cancer. 2010;10(1):37–50. doi: 10.1038/nrc2764.
  24. 24. Lampson BL, Gupta A, Tyekucheva S, et al. Rare Germline ATM Variants Influence the Development of Chronic Lymphocytic Leukemia. J Clin Oncol. 2023;41(5):1116–28. doi: 10.1200/JCO.22.00269.
  25. 25. Hallek M, Fischer K, Fingerle-Rowson G, et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 trial. Lancet. 2010;376(9747):1164–74. doi: 10.1016/S0140-6736(10)61381-5.
  26. 26. Zenz T, Vollmer D, Trbusek M, et al. TP53 mutation profile in chronic lymphocytic leukemia: evidence for a disease specific profile from a comprehensive analysis of 268 mutations. Leukemia. 2010;24(12):2072–9. doi: 10.1038/leu.2010.208.
  27. 27. Seiffert M, Dietrich S, Jethwa A, et al. Exploiting biological diversity and genomic aberrations in chronic lymphocytic leukemia. Leuk Lymphoma. 2012;53(6):1023–31. doi: 10.3109/10428194.2011.631638.
  28. 28. Yu L, Kim HT, Kasar SN, et al. Survival of del17p CLL depends on genomic complexity and somatic mutation. Clin Cancer Res. 2017;23(3):735–45. doi: 10.1158/1078-0432.CCR-16-0594.
  29. 29. Mikhaleva M, Tyekucheva S, Mashima K, et al. Higher Mutational Burden Is an Independent Predictor of Shorter Time to First Treatment in Untreated Chronic Lymphocytic Leukemia Patients. Blood. 2023;142(Suppl 1):3270. doi: 10.1182/blood-2023-173738.
  30. 30. Rigolin GM, Cibien F, Martinelli S, et al. Chromosome aberrations detected by conventional karyotyping using novel mitogens in chronic lymphocytic leukemia with “normal” FISH: correlations with clinicobiologic parameters. Blood. 2012;119(10):2310–3. doi: 10.1182/blood-2011-11-395269.
  31. 31. Baliakas P, Iskas M, Gardiner A, et al. Chromosomal translocations and karyotype complexity in chronic lymphocytic leukemia: a systematic reappraisal of classic cytogenetic data. Am J Hematol. 2014;89(3):249–55. doi: 10.1002/ajh.23618.
  32. 32. Cavallari M, Cavazzini F, Bardi A, et al. Biological significance and prognostic/predictive impact of complex karyotype in chronic lymphocytic leukemia. Oncotarget. 2018;9(76):34398–412. doi: 10.18632/oncotarget.26146.
  33. 33. Jaglowski SM, Ruppert AS, Heerema NA, et al. Complex karyotype predicts for inferior outcomes following reduced‐intensity conditioning allogeneic transplant for chronic lymphocytic leukaemia. Br J Haematol. 2012;159(1):82–7. doi: 10.1111/j.1365-2141.2012.09239.x.
  34. 34. Baliakas P, Jeromin S, Iskas M, et al. Cytogenetic complexity in chronic lymphocytic leukemia: definitions, associations, and clinical impact. Blood. 2019;133(11):1205–16. doi: 10.1182/blood-2018-09-873083.
  35. 35. Baliakas P, Espinet B, Mellink C, et al. Cytogenetics in chronic lymphocytic leukemia: ERIC perspectives and recommendations. Hemasphere. 2022;6(4):e707. doi: 10.1097/HS9.0000000000000707.
  36. 36. Hamblin TJ, Davis Z, Gardiner A, et al. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood. 1999;94(6):1848–54.
  37. 37. Damle RN, Wasil T, Fais F, et al. Ig V Gene Mutation Status and CD38 Expression As Novel Prognostic Indicators in Chronic Lymphocytic Leukemia. Blood. 1999;94(6):1840–7.
  38. 38. Ghia P, Stamatopoulos K, Belessi C, et al. ERIC recommendations on IGHV gene mutational status analysis in chronic lymphocytic leukemia. Leukemia. 2007;21(1):1–3. doi: 10.1038/sj.leu.2404457.
  39. 39. Agathangelidis A, Chatzidimitriou A, Chatzikonstantinou T, et al. Immunoglobulin gene sequence analysis in chronic lymphocytic leukemia: the 2022 update of the recommendations by ERIC, the European Research Initiative on CLL. Leukemia. 2022;36(8):1961–8. doi: 10.1038/s41375-022-01604-2.
  40. 40. Rosenquist R, Ghia P, Hadzidimitriou A, et al. Immunoglobulin gene sequence analysis in chronic lymphocytic leukemia: updated ERIC recommendations. Leukemia. 2017;31(7):1477–81. doi: 10.1038/leu.2017.125.
  41. 41. Mansouri L, Thorvaldsdottir B, Sutton LA, et al. Different prognostic impact of recurrent gene mutations in chronic lymphocytic leukemia depending on IGHV gene somatic hypermutation status: a study by ERIC in HARMONY. Leukemia. 2023;37(2):339–47. doi: 10.1038/s41375-022-01802-y.
  42. 42. Бидерман Б.В., Никитин Е.А., Сергиенко Т.Ф. и др. Репертуар генов тяжелой цепи иммуноглобулинов при В-клеточном хроническом лимфолейкозе в России и Беларуси. Онкогематология. 2012;3:38–43. [Biderman B.V., Nikitin E.A., Sergienko T.F., et al. Repertoire of immunoglobulin heavy chain genes in B-cell chronic lymphocytic leukemia in Russia and Belarus. Onkogematologiya. 2012;3:38–43. (In Russ)]
  43. 43. Бидерман Б.В., Судариков А.Б. Гены иммуноглобулинов и стереотипные антигенные рецепторы при хроническом лимфолейкозе и других лимфопролиферативных заболеваниях. Гематология и трансфузиология. 2023;68(1):70–9. doi: 10.35754/0234-5730-2023-68-1-70-79. [Biderman B.V., Sudarikov A.B. Immunoglobulin genes and stereotyped antigenic receptors in chronic lymphocytic leukemia and other lymphoproliferative diseases. Russian journal of hematology and transfusiology. 2023;68(1):70–9. doi: 10.35754/0234-5730-2023-68-1-70-79. (In Russ)]
  44. 44. Burger JA, Barr PM, Robak T, et al. Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study. Leukemia. 2020;34(3):787–98. doi: 10.1038/s41375-019-0602-x.
  45. 45. Stilgenbauer S, Schnaiter A, Paschka P, et al. Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood. 2014;123(21):3247–54. doi: 10.1182/blood-2014-01-546150.
  46. 46. Fischer K, Al-Sawaf O, Bahlo J, et al. Venetoclax and obinutuzumab in patients with CLL and coexisting conditions. N Engl J Med. 2019;380(23):2225–36. doi: 10.1056/NEJMoa1815281.
  47. 47. Brown JR, Eichhorst B, Hillmen P, et al. Zanubrutinib or ibrutinib in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med. 2023;388(4):319–32. doi: 10.1056/NEJMoa2211582.
  48. 48. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating. Proc Nat Acad Sci USA. 1977;74(12):5463–7. doi: 10.1073/pnas.74.12.5463.
  49. 49. Puente XS, Pinyol M, Quesada V, et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature. 2011;475(7354):101–5. doi: 10.1038/nature10113.
  50. 50. Quesada V, Conde L, Villamor N, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet. 2011;44(1):47–52. doi: 10.1038/ng.1032.
  51. 51. Wang L, Lawrence MS, Wan Y, et al. SF3B1 and other novel cancer genes in chronic lymphocytic leukemia. N Engl J Med. 2011;365(26):2497–506. doi: 10.1056/NEJMoa1109016.
  52. 52. Fabbri G, Rasi S, Rossi D, et al. Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation. J Exp Med. 2011;208(7):1389–401. doi: 10.1084/jem.20110921.
  53. 53. Puente XS, Bea S, Valdes-Mas R, et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature. 2015;526(7574):519–24. doi: 10.1038/nature14666.
  54. 54. Landau DA, Carter SL, Stojanov P, et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell. 2013;152(4):714–26. doi: 10.1016/j.cell.2013.01.019.
  55. 55. Landau DA, Stewart C, Reiter JG, et al. Novel Putative Driver Gene Mutations in Chronic Lymphocytic Leukemia (CLL): Results from a Combined Analysis of Whole-Exome Sequencing of 262 Primary CLL Samples. Blood. 2014;124(21):1952. doi: 10.1182/blood.v124.21.1952.1952.
  56. 56. Landau DA, Tausch E, Taylor-Weiner AN, et al. Mutations driving CLL and their evolution in progression and relapse. Nature. 2015;526(7574):525–30. doi: 10.1038/nature15395.
  57. 57. Ljungstrom V, Cortese D, Young E, et al. Whole-exome sequencing in relapsing chronic lymphocytic leukemia: clinical impact of recurrent RPS15 mutations. Blood. 2016;127(8):1007–16. doi: 10.1182/blood-2015-10-674572.
  58. 58. Knisbacher BA, Lin Z, Hahn CK, et al. Molecular map of chronic lymphocytic leukemia and its impact on outcome. Nat Genet. 2022;54(11):1664–74. doi: 10.1038/s41588-022-01140-w.
  59. 59. Mollstedt J, Mansouri L, Rosenquist R. Precision diagnostics in chronic lymphocytic leukemia: Past, present and future. Front Oncol. 2023;13:1146486. doi: 10.3389/fonc.2023.1146486.
  60. 60. Brieghel C, da Cunha-Bang C, Yde CW, et al. The number of signaling pathways altered by driver mutations in chronic lymphocytic leukemia impacts disease outcome. Clin Cancer Res. 2020;26(6):1507–15. doi: 10.1158/1078-0432.CCR-18-4158.
  61. 61. Vogelstein B, Papadopoulos N, Velculescu VE, et al. Cancer genome landscapes. Science. 2013;340(6127):1546–58. doi: 10.1126/science.1235122.
  62. 62. Sanchez-Vega F, Mina M, Armenia J, et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018;173(2):321–37.e10. doi: 10.1016/j.cell.2018.03.035.
  63. 63. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4. doi: 10.1158/2159-8290.CD-12-0095.
  64. 64. NCBI Resource Coordinators. Database resources of the national center for biotechnology information. Nucleic Acids Res. 2016;44(D1):D7–D19. doi: 10.1093/nar/gkv1290.
  65. 65. Tate JG, Bamford S, Jubb HC, et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019;47(D1):D941–D947. doi: 10.1093/nar/gky1015.
  66. 66. Sondka Z, Bamford S, Cole CG, et al. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat Rev Cancer. 2018;18(11):696–705. doi: 10.1038/s41568-018-0060-1.
  67. 67. Futreal PA, Coin L, Marshall M, et al. A census of human cancer genes. Nat Rev Cancer. 2004;4(3):177–83. doi: 10.1038/nrc1299.
  68. 68. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore M). Online Mendelian Inheritance in Man, OMIM®. Available from: https://omim.org/ (accessed 01.10.2024).
  69. 69. Fabregat A, Sidiropoulos K, Viteri G, et al. Reactome diagram viewer: data structures and strategies to boost performance. Bioinformatics. 2018;34(7):1208–14. doi: 10.1093/bioinformatics/btx752.
  70. 70. Kanehisa M, Sato Y. KEGG Mapper for inferring cellular functions from protein sequences. Protein Sci. 2020;29(1):28–35. doi: 10.1002/pro.3711.
  71. 71. Kanehisa M, Sato Y, Kawashima M. KEGG mapping tools for uncovering hidden features in biological data. Protein Sci. 2022;31(1):47–53. doi: 10.1002/pro.4172.
  72. 72. Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. Nat Genet. 2000;25(1):25–9. doi: 10.1038/75556.
  73. 73. Gene Ontology Consortium. The Gene Ontology resource: enriching a GOLD mine. Nucleic Acids Res. 2021;49(D1):D325–D334. doi: 10.1093/nar/gkaa1113.
  74. 74. Chakravarty D, Gao J, Phillips S, et al. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. 2017;2017:PO.17.00011. doi: 10.1200/PO.17.00011.
  75. 75. Bruford EA, Braschi B, Denny P, et al. Guidelines for human gene nomenclature. Nat Genet. 2020;52(8):754–8. doi: 10.1038/s41588-020-0669-3.
  76. 76. Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat Rev Cancer. 2003;3(10):756–67. doi: 10.1038/nrc1186.
  77. 77. Zent CS, Burack WR. Mutations in chronic lymphocytic leukemia and how they affect therapy choice: focus on NOTCH1, SF3B1, and TP53. Hematology. 2014;2014(1):119–24. doi: 10.1182/asheducation-2014.1.119.
  78. 78. Arruga F, Vaisitti T, Deaglio S. The NOTCH pathway and its mutations in mature B cell malignancies. Front Oncol. 2018;8:550. doi: 10.3389/fonc.2018.00550.
  79. 79. Damm F, Mylonas E, Cosson A, et al. Acquired initiating mutations in early hematopoietic cells of CLL patients. Cancer Discov. 2014;4(9):1088–111. doi: 10.1158/2159-8290.CD-14-0104.
  80. 80. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature. 2005;434(7035):843–50. doi: 10.1038/nature03319.
  81. 81. Makinen N, Mehine M, Tolvanen J, et al. MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science. 2011;334(6053):252–5. doi: 10.1126/science.1208930.
  82. 82. Janovska P, Bryja V. Wnt signalling pathways in chronic lymphocytic leukaemia and B‐cell lymphomas. Br J Pharmacol. 2017;174(24):4701–15. doi: 10.1111/bph.13949.
  83. 83. Messina M, Del Giudice I, Khiabanian H, et al. Genetic lesions associated with chronic lymphocytic leukemia chemo-refractoriness. Blood. 2014;123(15):2378–88. doi: 10.1182/blood-2013-10-534271.
  84. 84. Nilsson JA, Cleveland JL. Myc pathways provoking cell suicide and cancer. Oncogene. 2003;22(56):9007–21. doi: 10.1038/sj.onc.1207261.
  85. 85. Rossi D. MYC addiction in chronic lymphocytic leukemia. Leuk Lymphoma. 2013;54(5):905–6. doi: 10.3109/10428194.2012.755179.
  86. 86. Kern D, Regl G, Hofbauer SW, et al. Hedgehog/GLI and PI3K signaling in the initiation and maintenance of chronic lymphocytic leukemia. Oncogene. 2015;34(42):5341–51. doi: 10.1038/onc.2014.450.
  87. 87. Ghia EM, Rassenti LZ, Neuberg DS, et al. Activation of hedgehog signaling associates with early disease progression in chronic lymphocytic leukemia. Blood. 2019;133(25):2651–63. doi: 10.1182/blood-2018-09-873695.
  88. 88. Zhao S, Allis CD, Wang GG. The language of chromatin modification in human cancers. Nat Rev Cancer. 2021;21(7):413–30. doi: 10.1038/s41568-021-00357-x.
  89. 89. Rodriguez D, Bretones G, Quesada V, et al. Mutations in CHD2 cause defective association with active chromatin in chronic lymphocytic leukemia. Blood. 2015;126(2):195–202. doi: 10.1182/blood-2014-10-604959.
  90. 90. Mallm J, Iskar M, Ishaque N, et al. Linking aberrant chromatin features in chronic lymphocytic leukemia to transcription factor networks. Mol Syst Biol. 2019;15(5):e8339. doi: 10.15252/msb.20188339.
  91. 91. Mansouri L, Wierzbinska JA, Plass C, Rosenquist R. Epigenetic deregulation in chronic lymphocytic leukemia: Clinical and biological impact. Semin Cancer Biol. 2018;51:1–11. doi: 10.1016/j.semcancer.2018.02.001.
  92. 92. Unoki M, Nakamura Y. EGR2 induces apoptosis in various cancer cell lines by direct transactivation of BNIP3L and BAK. Oncogene. 2003;22(14):2172–85. doi: 10.1038/sj.onc.1206222.
  93. 93. Prieto C, Kharas MG. RNA regulators in leukemia and lymphoma. Cold Spring Harb Perspect Med. 2020;10(5):a034967. doi: 10.1101/cshperspect.a034967.
  94. 94. Rice GI, Bond J, Asipu A, et al. Mutations involved in Aicardi-Goutieres syndrome implicate SAMHD1 as regulator of the innate immune response. Nat Genet. 2009;41(7):829–32. doi: 10.1038/ng.373.
  95. 95. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9(3):153–66. doi: 10.1038/nrc2602.
  96. 96. Wade M, Li YC, Wahl GM. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nat Rev Cancer. 2013;13(2):83–96. doi: 10.1038/nrc3430.
  97. 97. Harvey KF, Zhang X, Thomas DM. The Hippo pathway and human cancer. Nat Rev Cancer. 2013;13(4):246–57. doi: 10.1038/nrc3458.
  98. 98. Noorbakhsh N, Hayatmoghadam B, Jamali M, et al. The Hippo signaling pathway in leukemia: function, interaction, and carcinogenesis. Cancer Cell Int. 2021;21(1):705. doi: 10.1186/s12935-021-02408-7.
  99. 99. Bos JL. Ras Oncogenes in Human Cancer: A Review. Cancer Res. 1989;49(17):4682–9.
  100. 100. Jebaraj BMC, Kienle D, Buhler A, et al. BRAF mutations in chronic lymphocytic leukemia. Leuk Lymphoma. 2013;54(6):1177–82. doi: 10.3109/10428194.2012.742525.
  101. 101. Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003;3(6):459–65. doi: 10.1038/nrc1097.
  102. 102. Marechal Y, Queant S, Polizzi S, et al. Inositol 1,4,5-trisphosphate 3-kinase B controls survival and prevents anergy in B cells. Immunobiology. 2011;216(1–2):103–9. doi: 10.1016/j.imbio.2010.03.012.
  103. 103. Proud CG. mTORC1 regulates the efficiency and cellular capacity for protein synthesis. Biochem Soc Trans. 2013;41(4):923–6. doi: 10.1042/BST20130036.
  104. 104. Woyach JA, Furman RR, Liu TM, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370(24):2286–94. doi: 10.1056/NEJMoa1400029.
  105. 105. Severin F, Frezzato F, Martini V, et al. Three Different Jak2/Stat3-Related Pathways Favor the Survival of Chronic Lymphocytic Leukemia Neoplastic Clone. Blood. 2018;132(Suppl 1):4405. doi: 10.1182/blood-2018-99-114591.
  106. 106. Fasouli ES, Katsantoni E. JAK-STAT in early hematopoiesis and leukemia. Front Cell Dev Biol. 2021;9:669363. doi: 10.3389/fcell.2021.669363.
  107. 107. Groner B, von Manstein V. Jak Stat signaling and cancer: Opportunities, benefits and side effects of targeted inhibition. Mol Cell Endocrinol. 2017;451:1–14. doi: 10.1016/j.mce.2017.05.033.
  108. 108. Severin F, Frezzato F, Visentin A, et al. In chronic lymphocytic leukemia the JAK2/STAT3 pathway is constitutively activated and its inhibition leads to CLL cell death unaffected by the protective bone marrow microenvironment. Cancers (Basel). 2019;11(12):1939. doi: 10.3390/cancers11121939.
  109. 109. Massague J. TGFβ in cancer. Cell. 2008;134(2):215–30. doi: 10.1016/j.cell.2008.07.001.
  110. 110. Matveeva A, Kovalevska L, Kholodnyuk I, et al. The TGF-beta—SMAD pathway is inactivated in cronic lymphocytic leukemia cells. Exp Oncol. 2017;39(4):286–90.
  111. 111. Dong M, Blobe GC. Role of transforming growth factor-β in hematologic malignancies. Blood. 2006;107(12):4589–96. doi: 10.1182/blood-2005-10-4169.
  112. 112. Gimenez N, Schulz R, Higashi M, et al. Targeting IRAK4 disrupts inflammatory pathways and delays tumor development in chronic lymphocytic leukemia. Leukemia. 2020;34(1):100–14. doi: 10.1038/s41375-019-0507-8.
  113. 113. Rozovski U, Keating MJ, Estrov Z. Targeting inflammatory pathways in chronic lymphocytic leukemia. Crit Rev Oncol Hematol. 2013;88(3):655–66. doi: 10.1016/j.critrevonc.2013.07.011.
  114. 114. Михалева М.А., Мартынкевич И.С., Булдаков И.А. и др. Секвенирование нового поколения как метод познания природы хронического лимфолейкоза и перехода к персонифицированной терапии. Гематология. Трансфузиология. Восточная Европа. 2021;7(2):176–90. doi: 10.34883/PI.2021.7.2.006. [Mikhaleva M.A., Martynkevich I.S., Buldakov I.A., et al. Next generation sequencing as a method for discovering the nature of chronic lymphocytic leukemia and transition to personalized treatment. 2021;7(2):176–90. Hematology. Transfusiology. Eastern Europe. doi: 10.34883/PI.2021.7.2.006. (In Russ)]
  115. 115. Robbe P, Ridout KE, Vavoulis DV, et al. Whole-genome sequencing of chronic lymphocytic leukemia identifies subgroups with distinct biological and clinical features. Nat Genet. 2022;54(11):1675–89. doi: 10.1038/s41588-022-01211-y.
  116. 116. Herbst SA, Vesterlund M, Helmboldt AJ, et al. Proteogenomics refines the molecular classification of chronic lymphocytic leukemia. Nat Commun. 2022;13(1):6226. doi: 10.1038/s41467-022-33385-8.
  117. 117. Wang Z, Yan H, Boysen JC, et al. B cell receptor signaling drives APOBEC3 expression via direct enhancer regulation in chronic lymphocytic leukemia B cells. Blood Cancer J. 2022;12(7):99. doi: 10.1038/s41408-022-00690-w.
  118. 118. Maher N, Mouhssine S, Matti BF, et al. Treatment Refractoriness in Chronic Lymphocytic Leukemia: Old and New Molecular Biomarkers. Int J Mol Sci. 2023;24(12):10374. doi: 10.3390/ijms241210374.
  119. 119. Thijssen R, Tian L, Anderson MA, et al. Single-cell multiomics reveal the scale of multilayered adaptations enabling CLL relapse during venetoclax therapy. Blood. 2022;140(20):2127–41. doi: 10.1182/blood.2022016040.
Creative Commons License

This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License.

Copyright (c) 2025 Clinical Oncohematology