Classical Hodgkin Lymphoma: Tumor Structure and Prognostic Value of the Immune Microenvironment

AA Gusak, KV Lepik, LV Fedorova, VV Markelov, VV Baykov

RM Gorbacheva Scientific Research Institute of Pediatric Oncology, Hematology and Transplantation; IP Pavlov First Saint Petersburg State Medical University, 6/8 L’va Tolstogo ul., Saint Petersburg, Russian Federation, 197022

For correspondence: Artem Aleksandrovich Gusak, 6/8 L’va Tolstogo ul., Saint Petersburg, Russian Federation, 197022; e-mail: artemgusak@yandex.ru

For citation: Gusak AA, Lepik KV, Fedorova LV, Markelov VV, Baykov VV. Classical Hodgkin Lymphoma: Tumor Structure and Prognostic Value of the Immune Microenvironment. Clinical oncohematology. 2023;16(3):242–62. (In Russ).

DOI: 10.21320/2500-2139-2023-16-3-242-262


ABSTRACT

Classical Hodgkin lymphoma (cHL) is a unique malignant lymphoid neoplasm characterized by tumor (Hodgkin and Reed-Sternberg) cells in the inflammatory and immunosuppressive microenvironment. The cHL microenvironment is a complex dynamic environment with immune cells, stromal elements, and extracellular matrix components, all of them interacting with each other and with tumor cells. This interaction basically underlies both disease progression and response to therapy. Currently, there is a growing interest in studying the structure and functions of cHL microenvironment, its prognostic value, and the potential of its components to be used as new therapeutic targets. During the last decade, the outcomes of refractory cHL treatment have considerably improved, in particular due to the administration of such PD-1 inhibitors as nivolumab and pembrolizumab. High cHL sensitivity to anti-PD-1 therapy can be accounted for by the PD-1/PD-L1-associated niche being formed in the tumor tissue as a result of intensive PD-L1 expression by tumor cells and macrophages as well as the expression of its PD-1 receptor by T-cells and M2-macrophages. More and more information becomes available about the possible mechanisms of antitumor response in anti-PD-1 treated cHL patients which seems to contradict the traditional understanding of CD8-mediated response in solid tumors. Cytotoxic effects of anti-PD-1 therapy in cHL tissues are likely to result from the interaction between tumor cells, macrophages, and CD4-positive Т-lymphocytes. This review discusses structural and regulatory relationships between tumor cells and microenvironment components, deals with new therapy approaches using various microenvironment components as targets, and summarizes currently available knowledge on prognosis based on the study of cHL microenvironment.

Keywords: classical Hodgkin lymphoma, microenvironment, immune checkpoints, macrophage polarization, immunosuppressive niche.

Received: April 12, 2023

Accepted: June 25, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Scott DW, Gascoyne RD. The tumour microenvironment in B cell lymphomas. Nat Rev Cancer. 2014;14(8):517–34. doi: 10.1038/nrc3774.
  2. Gallaher JA, Brown JS, Anderson ARA. The impact of proliferation-migration trade-offs on phenotypic evolution in cancer. Sci Rep. 2019;9(1):2425. doi: 10.1038/s41598-019-39636-x.
  3. Kuppers R, Engert A, Hansmann ML. Hodgkin lymphoma. J Clin Invest. 2012;122(10):3439–47. doi: 10.1172/JCI61245.
  4. Hertel CB, Zhou XG, Hamilton-Dutoit SJ, Junker S. Loss of B cell identity correlates with loss of B cell-specific transcription factors in Hodgkin/Reed-Sternberg cells of classical Hodgkin lymphoma. Oncogene. 2002;21(32):4908–20. doi: 10.1038/sj.onc.1205629.
  5. Kuppers R, Rajewsky K. The origin of Hodgkin and Reed/Sternberg cells in Hodgkin’s disease. Annu Rev Immunol. 1998;16:471–93. doi: 10.1146/annurev.immunol.16.1.471.
  6. Schwering I, Brauninger A, Klein U, et al. Loss of the B-lineage-specific gene expression program in Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood. 2003;101(4):1505–12. doi: 10.1182/blood-2002-03-0839.
  7. Weniger MA, Kuppers R. NF-κB deregulation in Hodgkin lymphoma. Semin Cancer Biol. 2016;39:32–9. doi: 10.1016/j.semcancer.2016.05.001.
  8. Tiacci E, Ladewig E, Schiavoni G, et al. Pervasive mutations of JAK-STAT pathway genes in classical Hodgkin lymphoma. Blood. 2018;131(22):2454–65. doi: 10.1182/blood-2017-11-814913.
  9. Garces de Los Fayos Alonso I, Liang HC, Turner SD, et al. The Role of Activator Protein-1 (AP-1) Family Members in CD30-Positive Lymphomas. Cancers (Basel). 2018;10(4):93. doi: 10.3390/cancers10040093.
  10. Jundt F, Anagnostopoulos I, Forster R, et al. Activated Notch1 signaling promotes tumor cell proliferation and survival in Hodgkin and anaplastic large cell lymphoma. Blood. 2002;99(9):3398–403. doi: 10.1182/blood.v99.9.3398.
  11. Zheng B, Fiumara P, Li YV, et al. MEK/ERK pathway is aberrantly active in Hodgkin disease: a signaling pathway shared by CD30, CD40, and RANK that regulates cell proliferation and survival. Blood. 2003;102(3):1019–27. doi: 10.1182/blood-2002-11-3507.
  12. Aravinth SP, Rajendran S, Li Y, et al. Epstein-Barr virus-encoded LMP1 induces ectopic CD137 expression on Hodgkin and Reed-Sternberg cells via the PI3K-AKT-mTOR pathway. Leuk Lymphoma. 2019;60(11):2697–704. doi: 10.1080/10428194.2019.1607330.
  13. Tiacci E, Doring C, Brune V, et al. Analyzing primary Hodgkin and Reed-Sternberg cells to capture the molecular and cellular pathogenesis of classical Hodgkin lymphoma. Blood. 2012;120(23):4609–20. doi: 10.1182/blood-2012-05-428896.
  14. Gruss HJ, Hirschstein D, Wright B, et al. Expression and function of CD40 on Hodgkin and Reed-Sternberg cells and the possible relevance for Hodgkin’s disease. Blood. 1994;84(7):2305–14.
  15. Gruss HJ, Duyster J, Herrmann F. Structural and biological features of the TNF receptor and TNF ligand superfamilies: interactive signals in the pathobiology of Hodgkin’s disease. Ann Oncol. 1996;7(Suppl 4):19–26. doi: 10.1093/annonc/7.suppl_4.s19.
  16. Yurchenko M, Sidorenko SP. Hodgkin’s lymphoma: the role of cell surface receptors in regulation of tumor cell fate. Exp Oncol. 2010;32(4):214–23.
  17. Chiu A, Xu W, He B, et al. Hodgkin lymphoma cells express TACI and BCMA receptors and generate survival and proliferation signals in response to BAFF and APRIL. Blood. 2007;109(2):729–39. doi: 10.1182/blood-2006-04-015958.
  18. Brune MM, Juskevicius D, Haslbauer J, et al. Genomic Landscape of Hodgkin Lymphoma. Cancers (Basel). 2021;13(4):682. doi: 10.3390/cancers13040682.
  19. Steidl C, Telenius A, Shah SP, et al. Genome-wide copy number analysis of Hodgkin Reed-Sternberg cells identifies recurrent imbalances with correlations to treatment outcome. Blood. 2010;116(3):418–27. doi: 10.1182/blood-2009-12-257345.
  20. Thomas RK, Kallenborn A, Wickenhauser C, et al. expression of c-FLIP in Hodgkin and Reed-Sternberg cells. Am J Pathol. 2002;160(4):1521–8. doi: 10.1016/S0002-9440(10)62578-3.
  21. Zhao X, Qiu W, Kung J, et al. Bortezomib induces caspase-dependent apoptosis in Hodgkin lymphoma cell lines and is associated with reduced c-FLIP expression: a gene expression profiling study with implications for potential combination therapies. Leuk Res. 2008;32(2):275–85. doi: 10.1016/j.leukres.2007.05.024.
  22. Maggio EM, Van Den Berg A, de Jong D, et al. Low frequency of FAS mutations in Reed-Sternberg cells of Hodgkin’s lymphoma. Am J Pathol. 2003;162(1):29–35. doi: 10.1016/S0002-9440(10)63795-9.
  23. Metkar SS, Naresh KN, Redkar AA, et al. Expression of Fas and Fas ligand in Hodgkin’s disease. Leuk Lymphoma. 1999;33(5–6):521–30. doi: 10.3109/10428199909058456.
  24. Verbeke CS, Wenthe U, Grobholz R, Zentgraf H. Fas ligand expression in Hodgkin lymphoma. Am J Surg Pathol. 2001;25(3):388–94. doi: 10.1097/00000478-200103000-00014.
  25. Mathas S, Lietz A, Anagnostopoulos I, et al. c-FLIP mediates resistance of Hodgkin/Reed-Sternberg cells to death receptor-induced apoptosis. J Exp Med. 2004;199(8):1041–52. doi: 10.1084/jem.20031080.
  26. Weniger MA, Kuppers R. Molecular biology of Hodgkin lymphoma. Leukemia. 2021;35(4):968–81. doi: 10.1038/s41375-021-01204-6.
  27. Roemer MG, Advani RH, Ligon AH, et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J Clin Oncol. 2016;34(23):2690–7. doi: 10.1200/JCO.2016.66.4482.
  28. Ai L, Xu A, Xu J. Roles of PD-1/PD-L1 Pathway: Signaling, Cancer, and Beyond. Adv Exp Med Biol. 2020;1248:33–59. doi: 10.1007/978-981-15-3266-5_3.
  29. Green MR, Monti S, Rodig SJ, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010;116(17):3268–77. doi: 10.1182/blood-2010-05-282780.
  30. Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. 2012;18(6):1611–8. doi: 10.1158/1078-0432.CCR-11-1942.
  31. Steidl C, Shah SP, Woolcock BW, et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature. 2011;471(7338):377–81. doi: 10.1038/nature09754.
  32. Reichel J, Chadburn A, Rubinstein PG, et al. Flow sorting and exome sequencing reveal the oncogenome of primary Hodgkin and Reed-Sternberg cells. Blood. 2015;125(7):1061–72. doi: 10.1182/blood-2014-11-610436.
  33. Dhatchinamoorthy K, Colbert JD, Rock KL. Cancer Immune Evasion Through Loss of MHC Class I Antigen Presentation. Front Immunol. 2021;12:636568. doi: 10.3389/fimmu.2021.636568.
  34. Thibodeau J, Bourgeois-Daigneault MC, Lapointe R. Targeting the MHC Class II antigen presentation pathway in cancer immunotherapy. Oncoimmunology. 2012;1(6):908–16. doi: 10.4161/onci.21205.
  35. Roemer MG, Advani RH, Redd RA, et al. Classical Hodgkin Lymphoma with Reduced β2M/MHC Class I Expression Is Associated with Inferior Outcome Independent of 9p24.1 Status. Cancer Immunol Res. 2016;4(11):910–6. doi: 10.1158/2326-6066.CIR-16-0201.
  36. Roemer MGM, Redd RA, Cader FZ, et al. Major Histocompatibility Complex Class II and Programmed Death Ligand 1 Expression Predict Outcome After Programmed Death 1 Blockade in Classic Hodgkin Lymphoma. J Clin Oncol. 2018;36(10):942–50. doi: 10.1200/JCO.2017.77.3994.
  37. Murray P, Bell A. Contribution of the Epstein-Barr Virus to the Pathogenesis of Hodgkin Lymphoma. Curr Top Microbiol Immunol. 2015;390(Pt 1):287–313. doi: 10.1007/978-3-319-22822-8_12.
  38. Xu M, Zhang WL, Zhu Q, et al. Genome-wide profiling of Epstein-Barr virus integration by targeted sequencing in Epstein-Barr virus associated malignancies. Theranostics. 2019;9(4):1115–24. doi: 10.7150/thno.29622.
  39. Massini G, Siemer D, Hohaus S. EBV in Hodgkin Lymphoma. Mediterr J Hematol Infect Dis. 2009;1(2):e2009013. doi: 10.4084/MJHID.2009.013.
  40. Murray PG, Young LS. An etiological role for the Epstein-Barr virus in the pathogenesis of classical Hodgkin lymphoma. Blood. 2019;134(7):591–6. doi: 10.1182/blood.2019000568.
  41. Carbone A, Gloghini A. Epstein Barr Virus-Associated Hodgkin Lymphoma. Cancers (Basel). 2018;10(6):163. doi: 10.3390/cancers10060163.
  42. Santisteban-Espejo A, Perez-Requena J, Atienza-Cuevas L, et al. Prognostic Role of the Expression of Latent-Membrane Protein 1 of Epstein-Barr Virus in Classical Hodgkin Lymphoma. Viruses. 2021;13(12):2523. doi: 10.3390/v13122523.
  43. Kuppers R. The biology of Hodgkin’s lymphoma. Nat Rev Cancer. 2009;9(1):15–27. doi: 10.1038/nrc2542.
  44. Rengstl B, Newrzela S, Heinrich T, et al. Incomplete cytokinesis and re-fusion of small mononucleated Hodgkin cells lead to giant multinucleated Reed-Sternberg cells. Proc Natl Acad Sci USA. 2013;110(51):20729–34. doi: 10.1073/pnas.1312509110.
  45. Aldinucci D, Gloghini A, Pinto A, et al. The classical Hodgkin’s lymphoma microenvironment and its role in promoting tumour growth and immune escape. J Pathol. 2010;221(3):248–63. doi: 10.1002/path.2711.
  46. Alaggio R, Amador C, Anagnostopoulos I, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia. 2022;36(7):1720–48. doi: 10.1038/s41375-022-01620-2.
  47. Bennett MH, Tu A, Hudson GV. Analysis of grade 1 Hodgkin’s disease (Report no 6). Clin Radiol. 1981;32(5):491–8. doi: 10.1016/s0009-9260(81)80174-2.
  48. Bennett MH, MacLennan KA, Easterling MJ, et al. The prognostic significance of cellular subtypes in nodular sclerosing Hodgkin’s disease: an analysis of 271 non-laparotomised cases (BNLI report no. 22). Clin Radiol. 1983;34(5):497–501. doi: 10.1016/s0009-9260(83)80148-2.
  49. MacLennan KA, Bennett MH, Tu A, et al. Relationship of histopathologic features to survival and relapse in nodular sclerosing Hodgkin’s disease. A study of 1659 patients. Cancer. 1989;64(8):1686–93. doi: 10.1002/1097-0142(19891015)64:8<1686::aid-cncr2820640822>3.0.co;2-i.
  50. Van Spronsen DJ, Vrints LW, Hofstra G, et al. Disappearance of prognostic significance of histopathological grading of nodular sclerosing Hodgkin’s disease for unselected patients, 1972–92. Br J Haematol. 1997;96(2):322–7. doi: 10.1046/j.1365-2141.1997.d01-2010.x.
  51. Pileri SA, Ascani S, Leoncini L, et al. Hodgkin’s lymphoma: the pathologist’s viewpoint. J Clin Pathol. 2002;55(3):162–76. doi: 10.1136/jcp.55.3.162.
  52. Lorenzen J, Thiele J, Fischer R. The mummified Hodgkin cell: cell death in Hodgkin’s disease. J Pathol. 1997;182(3):288–98. doi: 10.1002/(SICI)1096-9896(199707)182:3<288::AID-PATH859>3.0.CO;2-3.
  53. Eberle FC, Mani H, Jaffe ES. Histopathology of Hodgkin’s lymphoma. Cancer J. 2009;15(2):129–37. doi: 10.1097/PPO.0b013e31819e31cf.
  54. Wang HW, Balakrishna JP, Pittaluga S, Jaffe ES. Diagnosis of Hodgkin lymphoma in the modern era. Br J Haematol. 2019;184(1):45–59. doi: 10.1111/bjh.15614.
  55. Tzankov A, Bourgau C, Kaiser A, et al. Rare expression of T-cell markers in classical Hodgkin’s lymphoma. Mod Pathol. 2005;18(12):1542–9. doi: 10.1038/modpathol.3800473.
  56. Venkataraman G, Song JY, Tzankov A, et al. Aberrant T-cell antigen expression in classical Hodgkin lymphoma is associated with decreased event-free survival and overall survival. Blood. 2013;121(10):1795–804. doi: 10.1182/blood-2012-06-439455.
  57. Wang M, Zhao J, Zhang L, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8(5):761–73. doi: 10.7150/jca.17648.
  58. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer. 2018;118(1):9–16. doi: 10.1038/bjc.2017.434.
  59. Andre MPE, Carde P, Viviani S, et al. Long-term overall survival and toxicities of ABVD vs BEACOPP in advanced Hodgkin lymphoma: A pooled analysis of four randomized trials. Cancer Med. 2020;9(18):6565–75. doi: 10.1002/cam4.3298.
  60. Engert A, Diehl V, Franklin J, et al. Escalated-Dose BEACOPP in the treatment of patients with advanced-stage Hodgkin’s lymphoma: 10 years of follow-up of the GHSG HD9 study. J Clin Oncol. 2009;27(27):4548–54. doi: 10.1200/JCO.2008.19.8820.
  61. Schmitz N, Pfistner B, Sextro M, et al. Aggressive conventional chemotherapy compared with high-dose chemotherapy with autologous haemopoietic stem-cell transplantation for relapsed chemosensitive Hodgkin’s disease: A randomised trial. Lancet. 2002;359(9323):2065–71. doi: 10.1016/S0140-6736(02)08938-9.
  62. Arai S, Fanale M, Devos S, et al. Defining a Hodgkin lymphoma population for novel therapeutics after relapse from autologous hematopoietic cell transplant. Leuk Lymphoma. 2013;54(11):2531–3. doi: 10.3109/10428194.2013.798868.
  63. Chen R, Gopal A, Smith SE, et al. Five-Year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2016;128(12):1562–6. doi: 10.1182/blood-2016-02-699850.
  64. Armand P, Engert A, Younes A, et al. Nivolumab for relapsed/refractory classic Hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: Extended follow-up of the multicohort single-arm phase II checkmate 205 trial. J Clin Oncol. 2018;36(14):1428–39. doi: 10.1200/JCO.2017.76.0793.
  65. Lepik KV, Mikhailova NB, Moiseev IS, et al. Nivolumab for the treatment of relapsed and refractory classical Hodgkin lymphoma after ASCT and in ASCT-naive patients. Leuk Lymphoma. 2019;60(9):2316–9. doi: 10.1080/10428194.2019.1573368.
  66. Flores MBA, Corvinos MSa, Elez MM, et al. A new approach to the study of Hodgkin lymphoma by flow cytometry. Pathology. 2023;55(1):86–93. doi: 10.1016/j.pathol.2022.07.005.
  67. Cader FZ, Schackmann RCJ, Hu X, et al. Mass cytometry of Hodgkin lymphoma reveals a CD4+ regulatory T-cell-rich and exhausted T-effector microenvironment. Blood. 2018;132(8):825–36. doi: 10.1182/blood-2018-04-843714.
  68. Scott DW, Steidl C. The classical Hodgkin lymphoma tumor microenvironment: macrophages and gene expression-based modeling. Hematology Am Soc Hematol Educ Program. 2014;2014(1):144–50. doi: 10.1182/asheducation-2014.1.144.
  69. Carey CD, Gusenleitner D, Lipschitz M, et al. Topological analysis reveals a PD-L1-associated microenvironmental niche for Reed-Sternberg cells in Hodgkin lymphoma. Blood. 2017;130(22):2420–30. doi: 10.1182/blood-2017-03-770719.
  70. Elaldi R, Hemon P, Petti L, et al. High Dimensional Imaging Mass Cytometry Panel to Visualize the Tumor Immune Microenvironment Contexture. Front Immunol. 2021;12:666233. doi: 10.3389/fimmu.2021.666233.
  71. Ptacek J, Locke D, Finck R, et al. Multiplexed ion beam imaging (MIBI) for characterization of the tumor microenvironment across tumor types. Lab Invest. 2020;100(8):1111–23. doi: 10.1038/s41374-020-0417-4.
  72. Aldinucci D, Celegato M, Casagrande N. Microenvironmental interactions in classical Hodgkin lymphoma and their role in promoting tumor growth, immune escape and drug resistance. Cancer Lett. 2016;380(1):243–52. doi: 10.1016/j.canlet.2015.10.007.
  73. Aldinucci D, Lorenzon D, Cattaruzza L, et al. Expression of CCR5 receptors on Reed-Sternberg cells and Hodgkin lymphoma cell lines: involvement of CCL5/Rantes in tumor cell growth and microenvironmental interactions. Int J Cancer. 2008;122(4):769–76. doi: 10.1002/ijc.23119.
  74. Van den Berg A, Visser L, Poppema S. High expression of the CC chemokine TARC in Reed-Sternberg cells. A possible explanation for the characteristic T-cell infiltrate in Hodgkin’s lymphoma. Am J Pathol. 1999;154(6):1685–91. doi: 10.1016/S0002-9440(10)65424-7.
  75. Hnatkova M, Mocikova H, Trneny M, Zivny J. The biological environment of Hodgkin’s lymphoma and the role of the chemokine CCL17/TARC. Prague Med Rep. 2009;110(1):35–41.
  76. Niens M, Visser L, Nolte IM, et al. Serum chemokine levels in Hodgkin lymphoma patients: highly increased levels of CCL17 and CCL22. Br J Haematol. 2008;140(5):527–36. doi: 10.1111/j.1365-2141.2007.06964.x.
  77. Cattaruzza L, Gloghini A, Olivo K, et al. Functional coexpression of interleukin (IL)-7 and its receptor (IL-7R) on Hodgkin and Reed-Sternberg cells: Involvement of IL-7 in tumor cell growth and microenvironmental interactions of Hodgkin’s lymphoma. Int J Cancer. 2009;125(5):1092–101. doi: 10.1002/ijc.24389.
  78. Vera-Lozada G, Minnicelli C, Segges P, et al. Interleukin 10 (IL-10) proximal promoter polymorphisms beyond clinical response in classical Hodgkin lymphoma: Exploring the basis for the genetic control of the tumor microenvironment. Oncoimmunology. 2018;7(5):e1389821. doi: 10.1080/2162402X.2017.1389821.
  79. Hsu SM, Lin J, Xie SS, et al. Abundant expression of transforming growth factor-beta 1 and -beta 2 by Hodgkin’s Reed-Sternberg cells and by reactive T lymphocytes in Hodgkin’s disease. Hum Pathol. 1993;24(3):249–55. doi: 10.1016/0046-8177(93)90034-e.
  80. Kadin ME, Agnarsson BA, Ellingsworth LR, Newcom SR. Immunohistochemical evidence of a role for transforming growth factor beta in the pathogenesis of nodular sclerosing Hodgkin’s disease. Am J Pathol. 1990;136(6):1209–14.
  81. Maggio E, van den Berg A, Diepstra A, et al. Chemokines, cytokines and their receptors in Hodgkin’s lymphoma cell lines and tissues. Ann Oncol. 2002;13(Suppl 1):52–6. doi: 10.1093/annonc/13.s1.52.
  82. Fischer M, Juremalm M, Olsson N, et al. Expression of CCL5/RANTES by Hodgkin and Reed-Sternberg cells and its possible role in the recruitment of mast cells into lymphomatous tissue. Int J Cancer. 2003;107(2):197–201. doi: 10.1002/ijc.11370.
  83. Liu Y, Sattarzadeh A, Diepstra A, et al. The microenvironment in classical Hodgkin lymphoma: an actively shaped and essential tumor component. Semin Cancer Biol. 2014;24:15–22. doi: 10.1016/j.semcancer.2013.07.002.
  84. Skinnider BF, Mak TW. The role of cytokines in classical Hodgkin lymphoma. Blood. 2002;99(12):4283–97. doi: 10.1182/blood-2002-01-0099.
  85. Machado L, Jarrett R, Morgan S, et al. Expression and function of T cell homing molecules in Hodgkin’s lymphoma. Cancer Immunol Immunother. 2009;58(1):85–94. doi: 10.1007/s00262-008-0528-z.
  86. Opinto G, Agostinelli C, Ciavarella S, et al. Hodgkin Lymphoma: A Special Microenvironment. J Clin Med. 2021;10(20):4665. doi: 10.3390/jcm10204665.
  87. Zijtregtop EAM, Tromp I, Dandis R, et al. The Prognostic Value of Eight Immunohistochemical Markers Expressed in the Tumor Microenvironment and on Hodgkin Reed-Sternberg Cells in Pediatric Patients With Classical Hodgkin Lymphoma. Pathol Oncol Res. 2022;28:1610482. doi: 10.3389/pore.2022.1610482.
  88. Baumforth KR, Birgersdotter A, Reynolds GM, et al. Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin’s lymphoma cells mediates Up-regulation of CCL20 and the migration of regulatory T cells. Am J Pathol. 2008;173(1):195–204. doi: 10.2353/ajpath.2008.070845.
  89. Nagpal P, Descalzi-Montoya DB, Lodhi N. The circuitry of the tumor microenvironment in adult and pediatric Hodgkin lymphoma: cellular composition, cytokine profile, EBV, and exosomes. Cancer Rep (Hoboken). 2021;4(2):e1311. doi: 10.1002/cnr2.1311.
  90. Massini G, Siemer D, Hohaus S. EBV in Hodgkin Lymphoma. Mediterr J Hematol Infect Dis. 2009;1(2):e2009013. doi: 10.4084/MJHID.2009.013.
  91. Baumforth KR, Birgersdotter A, Reynolds GM, et al. Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin’s lymphoma cells mediates up-regulation of CCL20 and the migration of regulatory T cells. Am J Pathol. 2008;173(1):195–204. doi: 10.2353/ajpath.2008.070845.
  92. Pavlovic A, Glavina Durdov M, Capkun V, et al. Classical Hodgkin Lymphoma with Positive Epstein-Barr Virus Status is Associated with More FOXP3 Regulatory T Cells. Med Sci Monit. 2016;22:2340–6. doi: 10.12659/msm.896629.
  93. Steidl C, Connors JM, Gascoyne RD. Molecular pathogenesis of Hodgkin’s lymphoma: increasing evidence of the importance of the microenvironment. J Clin Oncol. 2011;29(14):1812–26. doi: 10.1200/JCO.2010.32.8401.
  94. Peh SC, Kim LH, Poppema S. TARC, a CC chemokine, is frequently expressed in classic Hodgkin’s lymphoma but not in NLP Hodgkin’s lymphoma, T-cell-rich B-cell lymphoma, and most cases of anaplastic large cell lymphoma. Am J Surg Pathol. 2001;25(7):925–9. doi: 10.1097/00000478-200107000-00011.
  95. Driessen J, Kersten MJ, Visser L, et al. Prognostic value of TARC and quantitative PET parameters in relapsed or refractory Hodgkin lymphoma patients treated with brentuximab vedotin and DHAP. Leukemia. 2022;36(12):2853–62. doi: 10.1038/s41375-022-01717-8.
  96. Kopinska A, Koclega A, Francuz T, et al. Serum thymus and activation-regulated chemokine (TARC) levels in newly diagnosed patients with Hodgkin lymphoma: a new promising and predictive tool? Preliminary report. J Hematopathol. 2021;14(4):277–81. doi: 10.1007/s12308-021-00470-8.
  97. Romano I, Puccini B, Signori L, et al. Serum TARC Concentration Kinetic in Classical Hodgkin Lymphoma during First-Line Treatment. Blood. 2021;138(Suppl 1):4500. doi: 10.1182/blood-2021-148137.
  98. Zijtregtop EAM, Tromp I, Dandis R, et al. The Prognostic Value of Eight Immunohistochemical Markers Expressed in the Tumor Microenvironment and on Hodgkin Reed-Sternberg Cells in Pediatric Patients With Classical Hodgkin Lymphoma. Pathol Oncol Res. 2022;28:1610482. doi: 10.3389/pore.2022.1610482.
  99. Vassilakopoulos TP, Nadali G, Angelopoulou MK, et al. Serum interleukin-10 levels are an independent prognostic factor for patients with Hodgkin’s lymphoma. Haematologica. 2001;86(3):274–81.
  100. Aldinucci D, Borghese C, Casagrande N. Formation of the Immunosuppressive Microenvironment of Classic Hodgkin Lymphoma and Therapeutic Approaches to Counter It. Int J Mol Sci. 2019;20(10):2416. doi: 10.3390/ijms20102416.
  101. Menendez V, Solorzano JL, Fernandez S, et al. The Hodgkin Lymphoma Immune Microenvironment: Turning Bad News into Good. Cancers (Basel). 2022;14(5):1360. doi: 10.3390/cancers14051360.
  102. Ferrarini I, Rigo A, Visco C, et al. The Evolving Knowledge on T and NK Cells in Classic Hodgkin Lymphoma: Insights into Novel Subsets Populating the Immune Microenvironment. Cancers (Basel). 2020;12(12):3757. doi: 10.3390/cancers12123757.
  103. Vardhana S, Younes A. The immune microenvironment in Hodgkin lymphoma: T cells, B cells, and immune checkpoints. Haematologica. 2016;101(7):794–802. doi: 10.3324/haematol.2015.132761.
  104. Liu WR, Shipp MA. Signaling pathways and immune evasion mechanisms in classical Hodgkin lymphoma. Blood. 2017;130(21):2265–70. doi: 10.1182/blood-2017-06-781989.
  105. Joller N, Kuchroo VK. Tim-3, Lag-3, and TIGIT. Curr Top Microbiol Immunol. 2017;410:127–56. doi: 10.1007/82_2017_62.
  106. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity. 2016;44(5):989–1004. doi: 10.1016/j.immuni.2016.05.001.
  107. Sakuishi K, Apetoh L, Sullivan JM, et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207(10):2187–94. doi: 10.1084/jem.20100643.
  108. Butte MJ, Keir ME, Phamduy TB, et al. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27(1):111–22. doi: 10.1016/j.immuni.2007.05.016.
  109. Patel SS, Weirather JL, Lipschitz M, et al. The microenvironmental niche in classic Hodgkin lymphoma is enriched for CTLA-4-positive T cells that are PD-1-negative. Blood. 2019;134(23):2059–69. doi: 10.1182/blood.2019002206.
  110. Aoki T, Chong LC, Takata K, et al. Single-Cell transcriptome analysis reveals disease-defining T-cell subsets in the tumor microenvironment of classic Hodgkin lymphoma. Cancer Discov. 2019;10(3):406–21. doi: 10.1158/2159-8290.CD-19-0680.
  111. Gusak A, Fedorova L, Lepik K, et al. Immunosuppressive Microenvironment and Efficacy of PD-1 Inhibitors in Relapsed/Refractory Classic Hodgkin Lymphoma: Checkpoint Molecules Landscape and Macrophage Populations. Cancers. 2021;13(22):5676. doi: 10.3390/cancers13225676.
  112. Greaves P, Clear A, Owen A, et al. Defining characteristics of classical Hodgkin lymphoma microenvironment T-helper cells. Blood. 2013;122(16):2856–63. doi: 10.1182/blood-2013-06-508044.
  113. Taylor JG, Truelove E, Clear A, et al. PDL1 shapes the classical Hodgkin lymphoma microenvironment without inducing T-cell exhaustion. Haematologica. 2023;108(4):1068–82. doi: 10.3324/haematol.2022.280014.
  114. Diefenbach CS, Hong F, Ambinder RF, et al. Ipilimumab, nivolumab, and brentuximab vedotin combination therapies in patients with relapsed or refractory Hodgkin lymphoma: phase 1 results of an open-label, multicentre, phase 1/2 trial. Lancet Haematol. 2020;7(9):e660–e670. doi: 10.1016/S2352-3026(20)30221-0.
  115. Wein F, Kuppers R. The role of T cells in the microenvironment of Hodgkin lymphoma. J Leukoc Biol. 2016;99(1):45–50. doi: 10.1189/jlb.3MR0315-136R.
  116. Aldinucci D, Gloghini A, Pinto A, et al. The role of CD40/CD40L and interferon regulatory factor 4 in Hodgkin lymphoma microenvironment. Leuk Lymphoma. 2012;53(2):195–201. doi: 10.3109/10428194.2011.605190.
  117. Veldman J, Visser L, Huberts-Kregel M, et al. Rosetting T cells in Hodgkin lymphoma are activated by immunological synapse components HLA class II and CD58. Blood. 2020;136(21):2437–41. doi: 10.1182/blood.2020005546.
  118. Abdul Razak FR, Diepstra A, Visser L, van den Berg A. CD58 mutations are common in Hodgkin lymphoma cell lines and loss of CD58 expression in tumor cells occurs in Hodgkin lymphoma patients who relapse. Genes Immun. 2016;17(6):363–6. doi: 10.1038/gene.2016.30.
  119. Schneider M, Schneider S, Zuhlke-Jenisch R, et al. Alterations of the CD58 gene in classical Hodgkin lymphoma. Genes Chromosomes Cancer. 2015;54(10):638–45. doi: 10.1002/gcc.22276.
  120. Mulder TA, Andersson ML, Pena-Perez L, et al. Immune Biomarkers in the Peripheral Blood and Tumor Microenvironment of Classical Hodgkin Lymphoma Patients in Relation to Tumor Burden and Response to Treatment. Hemasphere. 2022;6(11):e794. doi: 10.1097/HS9.0000000000000794.
  121. Koenecke C, Ukena SN, Ganser A, Franzke A. Regulatory T cells as therapeutic target in Hodgkin’s lymphoma. Expert Opin Ther Targets. 2008;12(6):769–82. doi: 10.1517/14728222.12.6.769.
  122. Tanijiri T, Shimizu T, Uehira K, et al. Hodgkin’s Reed-Sternberg cell line (KM-H2) promotes a bidirectional differentiation of CD4+CD25+Foxp3+ T cells and CD4+ cytotoxic T lymphocytes from CD4+ naive T cells. J Leukoc Biol. 2007;82(3):576–84. doi: 10.1189/jlb.0906565.
  123. Littringer K, Moresi C, Rakebrandt N, et al. Common Features of Regulatory T Cell Specialization During Th1 Responses. Front Immunol. 2018;9:1344. doi: 10.3389/fimmu.2018.01344.
  124. Marshall NA, Christie LE, Munro LR, et al. Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma. Blood. 2004;103(5):1755–62. doi: 10.1182/blood-2003-07-2594.
  125. Pan Y, Yu Y, Wang X, Zhang T. Tumor-Associated Macrophages in Tumor Immunity. Front Immunol. 2020;11:583084. doi: 10.3389/fimmu.2020.583084.
  126. Steidl C, Farinha P, Gascoyne RD. Macrophages predict treatment outcome in Hodgkin’s lymphoma. Haematologica. 2011;96(2):186–9. doi: 10.3324/haematol.2010.033316.
  127. Karihtala K, Leivonen SK, Bruck O, et al. Prognostic Impact of Tumor-Associated Macrophages on Survival Is Checkpoint Dependent in Classical Hodgkin Lymphoma. Cancers (Basel). 2020;12(4):877. doi: 10.3390/cancers12040877.
  128. Yao Y, Xu XH, Jin L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front Immunol. 2019;10:792. doi: 10.3389/fimmu.2019.00792.
  129. Shapouri-Moghaddam A, Mohammadian S, Vazini H, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233(9):6425–40. doi: 10.1002/jcp.26429.
  130. Mantovani A, Sozzani S, Locati M, et al. Macrophage polarization: Tumor-Associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23(11):549–55. doi: 10.1016/s1471-4906(02)02302-5.
  131. Barros MHM, Segges P, Vera-Lozada G, et al. Macrophage polarization reflects T cell composition of tumor microenvironment in pediatric classical Hodgkin lymphoma and has impact on survival. PLoS ONE 2015;10(5):e0124531. doi: 10.1371/journal.pone.0124531.
  132. Najafi M, Goradel NH, Farhood B, et al. Macrophage polarity in cancer: A review. J Cell Biochem. 2018;120(3):2756–65. doi: 10.1002/jcb.27646.
  133. Jiang Z, Sun H, Yu J, et al. Targeting CD47 for cancer immunotherapy. J Hematol Oncol. 2021;14(1):180. doi: 10.1186/s13045-021-01197-w.
  134. Li Z, Li Y, Gao J, et al. The role of CD47-SIRPα immune checkpoint in tumor immune evasion and innate immunotherapy. Life Sci. 2021;273:119150. doi: 10.1016/j.lfs.2021.119150.
  135. Oronsky B, Carter C, Reid T, et al. Just eat it: A review of CD47 and SIRP-α Semin Oncol. 2020;47(2–3):117–24. doi: 10.1053/j.seminoncol.2020.05.009.
  136. Gholiha AR, Hollander P, Lof L, et al. Checkpoint CD47 expression in classical Hodgkin lymphoma. Br J Haematol. 2022;197(5):580–9. doi: 10.1111/bjh.18137.
  137. Russ A, Hua AB, Montfort WR, et al. Blocking “don’t eat me” signal of CD47-SIRPα in hematological malignancies, an in-depth review. Blood Rev. 2018;32(6):480–9. doi: 10.1016/j.blre.2018.04.005.
  138. Hayat SMG, Bianconi V, Pirro M, et al. CD47: role in the immune system and application to cancer therapy. Cell Oncol (Dordr). 2020;43(1):19–30. doi: 10.1007/s13402-019-00469-5.
  139. Lu D, Ni Z, Liu X, et al. Beyond T Cells: Understanding the Role of PD-1/PD-L1 in Tumor-Associated Macrophages. J Immunol Res. 2019;2019:1919082. doi: 10.1155/2019/1919082.
  140. Vari F, Arpon D, Keane C, et al. Immune evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. Blood. 2018;131(16):1809–19. doi: 10.1182/blood-2017-07-796342.
  141. Gordon SR, Maute RL, Dulken BW, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017;545(7655):495–9. doi: 10.1038/nature22396.
  142. Li W, Wu F, Zhao S, et al. Correlation between PD-1/PD-L1 expression and polarization in tumor-associated macrophages: A key player in tumor immunotherapy. Cytokine Growth Factor Rev. 2022;67:49–57. doi: 10.1016/j.cytogfr.2022.07.004.
  143. De la Cruz-Merino L, Lejeune M, Nogales Fernandez E, et al. Role of immune escape mechanisms in Hodgkin’s lymphoma development and progression: a whole new world with therapeutic implications. Clin Dev Immunol. 2012;2012:756353. doi: 10.1155/2012/756353.
  144. Calabretta E, d’Amore F, Carlo-Stella C. Immune and Inflammatory Cells of the Tumor Microenvironment Represent Novel Therapeutic Targets in Classical Hodgkin Lymphoma. Int J Mol Sci. 2019;20(21):5503. doi: 10.3390/ijms20215503.
  145. Yamamoto R, Nishikori M, Kitawaki T, et al. PD-1-PD-1 ligand interaction contributes to immunosuppressive microenvironment of Hodgkin lymphoma. Blood. 2008;111(6):3220–4. doi: 10.1182/blood-2007-05-085159.
  146. Xiong H, Mittman S, Rodriguez R, et al. Anti-PD-L1 Treatment Results in Functional Remodeling of the Macrophage Compartment. Cancer Res. 2019;79(7):1493–506. doi: 10.1158/0008-5472.CAN-18-3208.
  147. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71. doi: 10.1038/nature13954.
  148. Willenbrock K, Roers A, Blohbaum B, et al. CD8(+) T cells in Hodgkin’s disease tumor tissue are a polyclonal population with limited clonal expansion but little evidence of selection by antigen. Am J Pathol. 2000;157(1):171–5. doi: 10.1016/S0002-9440(10)64528-2.
  149. Neefjes J, Jongsma MLM, Paul P, et al. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 2011;11(12):823–36. doi: 10.1038/nri3084.
  150. Nagasaki J, Togashi Y, Sugawara T, et al. The critical role of CD4+ T cells in PD-1 blockade against MHC-II-expressing tumors such as classic Hodgkin lymphoma. Blood Adv. 2020;4(17):4069–82. doi: 10.1182/bloodadvances.2020002098.
  151. Cader FZ, Hu X, Goh WL, et al. A peripheral immune signature of responsiveness to PD-1 blockade in patients with classical Hodgkin lymphoma. Nat Med. 2020;26(9):1468–79. doi: 10.1038/s41591-020-1006-1.
  152. Reinke S, Brockelmann PJ, Iaccarino I, et al. Tumor and microenvironment response but no cytotoxic T-cell activation in classic Hodgkin lymphoma treated with anti-PD1. Blood. 2020;136(25):2851–63. doi: 10.1182/blood.2020008553.
  153. Brockelmann PJ, Goergen H, Keller U, et al. Efficacy of Nivolumab and AVD in Early-Stage Unfavorable Classic Hodgkin Lymphoma: The Randomized Phase 2 German Hodgkin Study Group NIVAHL Trial. JAMA Oncol. 2020;6(6):872–80. doi: 10.1001/jamaoncol.2020.0750.
  154. Chen R, Zinzani PL, Lee HJ, et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood. 2019;134(14):1144–53. doi: 10.1182/blood.2019000324.
  155. Lepik KV, Fedorova LV, Kondakova EV, et al. A Phase 2 Study of Nivolumab Using a Fixed Dose of 40 mg (Nivo40) in Patients With Relapsed/Refractory Hodgkin Lymphoma. Hemasphere. 2020;4(5):e480. doi: 10.1097/HS9.0000000000000480.
  156. Herrera AF, Chen R, Palmer J, et al. PET-adapted nivolumab or nivolumab plus ICE as first salvage therapy in relapsed or refractory Hodgkin lymphoma. Blood. 2019;134(Suppl 1):239. doi: 10.1182/blood-2019-123162.
  157. Allen PB, Savas H, Evens AM, et al. Pembrolizumab followed by AVD in untreated early unfavorable and advanced-stage classical Hodgkin lymphoma. Blood. 2021;137(10):1318–26. doi: 10.1182/blood.2020007400.
  158. Herrera AF, Burton C, Radford J, et al. Avelumab in relapsed/refractory classical Hodgkin lymphoma: phase 1b results from the JAVELIN Hodgkins trial. Blood Adv. 2021;5(17):3387–96. doi: 10.1182/bloodadvances.2021004511.
  159. Timmerman J, Lavie D, Johnson NA, et al. Favezelimab (anti–LAG-3) plus pembrolizumab in patients with relapsed or refractory (R/R) classical Hodgkin lymphoma (cHL) after anti–PD-1 treatment: An open-label phase 1/2 study. J Clin Oncol. 2022;40(Suppl 16):7545. doi: 10.1200/JCO.2022.40.16_suppl.7545.
  160. Diefenbach CS, Hong F, Ambinder RF, et al. Ipilimumab, nivolumab, and brentuximab vedotin combination therapies in patients with relapsed or refractory Hodgkin lymphoma: phase 1 results of an open-label, multicentre, phase 1/2 trial. Lancet Haematol. 2020;7(9):e660–e670. doi: 10.1016/S2352-3026(20)30221-0.
  161. Fares CM, van Allen EM, Drake CG, et al. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ Book. 2019;39:147–64. doi: 10.1200/EDBK_240837.
  162. Smaglo BG, Aldeghaither D, Weiner LM. The development of immunoconjugates for targeted cancer therapy. Nat Rev Clin Oncol. 2014;11(11):637–48. doi: 10.1038/nrclinonc.2014.159.
  163. Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol. 2012;30(18):2183–9. doi: 10.1200/JCO.2011.38.0410.
  164. Hamadani M, Collins GP, Samaniego F, et al. Phase 1 study of ADCT-301 (Camidanlumab Tesirine), a novel pyrrolobenzodiazepine-based antibody drug conjugate, in relapsed/refractory classical Hodgkin Lymphoma. Blood. 2018;132(Suppl 1):928. doi: 10.1182/blood-2018-99-118198.
  165. Flynn MJ, Hartley JA. The emerging role of anti-CD25 directed therapies as both immune modulators and targeted agents in cancer. Br J Haematol. 2017;179(1):20–35. doi: 10.1111/bjh.14770.
  166. Zelenay S, Lopes-Carvalho T, Caramalho I, et al. Foxp3+ CD25- CD4 T cells constitute a reservoir of committed regulatory cells that regain CD25 expression upon homeostatic expansion. Proc Natl Acad Sci USA. 2005;102(11):4091–6. doi: 10.1073/pnas.0408679102.
  167. Zinzani PL, Carlo-Stella C, Hamadani M, et al. Camidanlumab tesirine efficacy and safety in an open-label, multicenter, phase 2 study of patients (pts) with relapsed or refractory classical Hodgkin lymphoma (r/r cHL). Hematol Oncol. 2021;39(S2):125–7. doi: 10.1002/hon.75_2879.
  168. Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66(2):605–12. doi: 10.1158/0008-5472.CAN-05-4005.
  169. Smith CC, Perl AE, Lasater E, et al. PLX3397 Is An Investigational Selective FLT3 Inhibitor That Retains Activity Against the Clinically-Relevant FLT3-ITD/F691L “Gatekeeper” Mutation in Vitro. Blood. 2011;118(21):764. doi: 10.1182/blood.V118.21.764.764.
  170. Moskowitz CH, Younes A, de Vos S, et al. CSF1R Inhibition by PLX3397 in Patients with Relapsed or Refractory Hodgkin Lymphoma: Results From a Phase 2 Single Agent Clinical Trial. Blood. 2012;120(21):1638. doi: 10.1182/blood.V120.21.1638.1638.
  171. Fujiwara T, Yakoub MA, Chandler A, et al. CSF1/CSF1R Signaling Inhibitor Pexidartinib (PLX3397) Reprograms Tumor-Associated Macrophages and Stimulates T-cell Infiltration in the Sarcoma Microenvironment. Mol Cancer Ther. 2021;20(8):1388–99. doi: 10.1158/1535-7163.MCT-20-0591.
  172. Dickinson MJ, Carlo-Stella C, Morschhauser F, et al. Glofitamab for Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med. 2022;387(24):2220–31. doi: 10.1056/NEJMoa2206913.
  173. Rothe A, Sasse S, Topp MS, et al. A phase 1 study of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2015;125(26):4024–31. doi: 10.1182/blood-2014-12-614636.
  174. Kerbauy LN, Marin ND, Kaplan M, et al. Combining AFM13, a Bispecific CD30/CD16 Antibody, with Cytokine-Activated Blood and Cord Blood-Derived NK Cells Facilitates CAR-like Responses Against CD30+ Malignancies. Clin Cancer Res. 2021;27(13):3744–56. doi: 10.1158/1078-0432.CCR-21-0164.
  175. Rajendran S, Li Y, Ngoh E, et al. Development of a Bispecific Antibody Targeting CD30 and CD137 on Hodgkin and Reed-Sternberg Cells. Front Oncol. 2019;9:945. doi: 10.3389/fonc.2019.00945.
  176. Piccione EC, Juarez S, Liu J, et al. A bispecific antibody targeting CD47 and CD20 selectively binds and eliminates dual antigen expressing lymphoma cells. MAbs. 2015;7(5):946–56. doi: 10.1080/19420862.2015.1062192.
  177. Wang Y, Ni H, Zhou S, et al. Tumor-selective blockade of CD47 signaling with a CD47/PD-L1 bispecific antibody for enhanced anti-tumor activity and limited toxicity. Cancer Immunol Immunother. 2021;70(2):365–76. doi: 10.1007/s00262-020-02679-5.
  178. Kamdar M, Solomon SR, Arnason J, et al. Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet. 2022;399(10343):2294–308. doi: 10.1016/S0140-6736(22)00662-6.
  179. Ramos CA, Grover CA, Beaven AW, et al. Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin lymphoma. J Clin Oncol. 2020;38(32):3794–804. doi: 10.1200/JCO.20.01342.
  180. Ruella M, Klichinsky M, Kenderian SS, et al. Overcoming the Immunosuppressive Tumor Microenvironment of Hodgkin Lymphoma Using Chimeric Antigen Receptor T Cells. Cancer Discov. 2017;7(10):1154–67. doi: 10.1158/2159-8290.CD-16-0850.
  181. Shah SR, Tran TM. Lenalidomide in myelodysplastic syndrome and multiple myeloma. Drugs. 2007;67(13):1869–81. doi: 10.2165/00003495-200767130-00005.
  182. Wang M, Fowler N, Wagner-Bartak N, et al. Oral lenalidomide with rituximab in relapsed or refractory diffuse large cell, follicular and transformed lymphoma: a phase II clinical trial. Leukemia. 2013;27(9):1902–9. doi: 10.1038/leu.2013.95.
  183. Fehniger TA, Larson S, Trinkaus K, et al. A phase 2 multicenter study of lenalidomide in relapsed or refractory classical Hodgkin lymphoma. Blood. 2011;118(19):5119–25. doi: 10.1182/blood-2011-07-362475.
  184. Kuruvilla J, Taylor D, Wang L, et al. Phase II trial of lenalidomide in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2008;112(11):3052. doi: 10.1182/blood.V112.11.3052.3052.
  185. Alonso-Alvarez S, Vidriales MB, Caballero MD, et al. The number of tumor infiltrating T-cell subsets in lymph nodes from patients with Hodgkin lymphoma is associated with the outcome after first line ABVD therapy. Leuk Lymphoma. 2017;58(5):1144–52. doi: 10.1080/10428194.2016.1239263.
  186. Zawati I, Adouni O, Manai M, et al. FOXP3+/CD68+ ratio within the tumor microenvironment may serve as a potential prognostic factor in classical Hodgkin lymphoma. Hum Immunol. 2022;83(12):843–56. doi: 10.1016/j.humimm.2022.08.013.
  187. Oudejans JJ, Jiwa NM, Kummer JA, et al. Activated cytotoxic T cells as prognostic marker in Hodgkin’s disease. Blood. 1997;89(4):1376–82.
  188. Alvaro T, Lejeune M, Salvado MT, et al. Outcome in Hodgkin’s lymphoma can be predicted from the presence of accompanying cytotoxic and regulatory T cells. Clin Cancer Res. 2005;11(4):1467–73. doi: 10.1158/1078-0432.CCR-04-1869.
  189. Kelley TW, Pohlman B, Elson P, Hsi ED. The ratio of FOXP3+ regulatory T cells to granzyme B+ cytotoxic T/NK cells predicts prognosis in classical Hodgkin lymphoma and is independent of bcl-2 and MAL expression. Am J Clin Pathol. 2007;128(6):958–65. doi: 10.1309/NB3947K383DJ0LQ2.
  190. Schreck S, Friebel D, Buettner M, et al. Prognostic impact of tumour-infiltrating Th2 and regulatory T cells in classical Hodgkin lymphoma. Hematol Oncol. 2009;27(1):31–9. doi: 10.1002/hon.878.
  191. Agostinelli C, Gallamini A, Stracqualursi L, et al. The combined role of biomarkers and interim PET scan in prediction of treatment outcome in classical Hodgkin’s lymphoma: a retrospective, European, multicentre cohort study. Lancet Haematol. 2016;3(10):e467–e479. doi: 10.1016/S2352-3026(16)30108-9.
  192. Muenst S, Hoeller S, Dirnhofer S, Tzankov A. Increased programmed death-1+ tumor-infiltrating lymphocytes in classical Hodgkin lymphoma substantiate reduced overall survival. Hum Pathol. 2009;40(12):1715–22. doi: 10.1016/j.humpath.2009.03.025.
  193. Nguyen TT, Frater JL, Klein J, et al. Expression of TIA1 and PAX5 in Classical Hodgkin Lymphoma at Initial Diagnosis May Predict Clinical Outcome. Appl Immunohistochem Mol Morphol. 2016;24(6):383–91. doi: 10.1097/PAI.0000000000000200.
  194. Chetaille B, Bertucci F, Finetti P, et al. Molecular profiling of classical Hodgkin lymphoma tissues uncovers variations in the tumor microenvironment and correlations with EBV infection and outcome. Blood. 2009;113(12):2765–3775. doi: 10.1182/blood-2008-07-168096.
  195. Greaves P, Clear A, Coutinho R, et al. Expression of FOXP3, CD68, and CD20 at diagnosis in the microenvironment of classical Hodgkin lymphoma is predictive of outcome. J Clin Oncol. 2013;31(2):256–62. doi: 10.1200/JCO.2011.39.9881.
  196. Wang C, Xia B, Wang T, et al. PD-1, FOXP3, and CSF-1R expression in patients with Hodgkin lymphoma and their prognostic value. Int J Clin Exp Pathol. 2018;11(4):1923–34.
  197. Tzankov A, Meier C, Hirschmann P, et al. Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin’s lymphoma. Haematologica. 2008;93(2):193–200. doi: 10.3324/haematol.11702.
  198. Lacet DFR, Oliveira CC. The role of immunohistochemistry in the assessment of classical Hodgkin lymphoma microenvironment. Int J Clin Exp Pathol. 2022;15(10):412–24.
  199. Moerdler S, Ewart M, Friedman DL, et al. LAG-3 is expressed on a majority of tumor infiltrating lymphocytes in pediatric Hodgkin lymphoma. Leuk Lymphoma. 2021;62(3):606–13. doi: 10.1080/10428194.2020.1839651.
  200. Annibali O, Bianchi A, Grifoni A, et al. A novel scoring system for TIGIT expression in classic Hodgkin lymphoma. Sci Rep. 2021;11(1):7059. doi: 10.1038/s41598-021-86655-8.
  201. Karihtala K, Leivonen SK, Karjalainen-Lindsberg ML, et al. Checkpoint protein expression in the tumor microenvironment defines the outcome of classical Hodgkin lymphoma patients. Blood Adv. 2022;6(6):1919–31. doi: 10.1182/bloodadvances.2021006189.
  202. Guo B, Cen H, Tan X, Ke Q. Meta-analysis of the prognostic and clinical value of tumor-associated macrophages in adult classical Hodgkin lymphoma. BMC Med. 2016;14(1):159. doi: 10.1186/s12916-016-0711-6.
  203. Klein JL, Nguyen TT, Bien-Willner GA, et al. CD163 Immunohistochemistry is superior to CD68 in predicting outcome in classical Hodgkin lymphoma. Am J Clin Pathol. 2014;141(3):381–7. doi: 10.1309/AJCP61TLMXLSLJYS.
  204. Barros MHM, Hauck F, Dreyer JH, et al. Macrophage polarisation: An immunohistochemical approach for identifying M1 and M2 macrophages. PLoS One. 2013;8(11):e80908. doi: 10.1371/journal.pone.0080908.
  205. Barros MH, Segges P, Vera-Lozada G, et al. Macrophage polarization reflects T cell composition of tumor microenvironment in pediatric classical Hodgkin lymphoma and has impact on survival. PLoS One. 2015;10(5):e0124531. doi: 10.1371/journal.pone.0124531.
  206. Steidl C, Lee T, Shah SP, et al. Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med. 2010;362(10):875–85. doi: 10.1056/NEJMoa0905680.
  207. Tudor CS, Distel LV, Eckhardt J, et al. B cells in classical Hodgkin lymphoma are important actors rather than bystanders in the local immune reaction. Hum Pathol. 2013;44(11):2475–86. doi: 10.1016/j.humpath.2013.06.006.
  208. Mizuno H, Nakayama T, Miyata Y, et al. Mast cells promote the growth of Hodgkin’s lymphoma cell tumor by modifying the tumor microenvironment that can be perturbed by bortezomib. Leukemia. 2012;26(10):2269–76. doi: 10.1038/leu.2012.81.
  209. Glimelius I, Edstrom A, Fischer M, et al. Angiogenesis and mast cells in Hodgkin lymphoma. Leukemia. 2005;19(12):2360–2. doi: 10.1038/sj.leu.2403992.
  210. Ribatti D, Tamma R, Annese T, et al. Inflammatory microenvironment in classical Hodgkin’s lymphoma with special stress on mast cells. Front Oncol. 2022;12:964573. doi: 10.3389/fonc.2022.964573.
  211. Komi DEA, Redegeld FA. Role of Mast Cells in Shaping the Tumor Microenvironment. Clin Rev Allergy Immunol. 2020;58(3):313–25. doi: 10.1007/s12016-019-08753-w.
  212. Nakayama S, Yokote T, Hiraoka N, et al. Role of mast cells in fibrosis of classical Hodgkin lymphoma. Int J Immunopathol Pharmacol. 2016;29(4):603–11. doi: 10.1177/0394632016644447.
  213. Molin D, Fischer M, Xiang Z, et al. Mast cells express functional CD30 ligand and are the predominant CD30L-positive cells in Hodgkin’s disease. Br J Haematol. 2001;114(3):616–23. doi: 10.1046/j.1365-2141.2001.02977.x.
  214. Molin D, Edstrom A, Glimelius I, et al. Mast cell infiltration correlates with poor prognosis in Hodgkin’s lymphoma. Br J Haematol. 2002;119(1):122–4. doi: 10.1046/j.1365-2141.2002.03768.x.
  215. Keresztes K, Szollosi Z, Simon Z, et al. Retrospective analysis of the prognostic role of tissue eosinophil and mast cells in Hodgkin’s lymphoma. Pathol Oncol Res. 2007;13(3):237–42. doi: 10.1007/BF02893504.
  216. Andersen MD, Kamper P, Nielsen PS, et al. Tumour-associated mast cells in classical Hodgkin’s lymphoma: correlation with histological subtype, other tumour-infiltrating inflammatory cell subsets and outcome. Eur J Haematol. 2016;96(3):252–9. doi: 10.1111/ejh.12583.
  217. Swerdlow SH, Campo E, Harris NL, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Revised 4th edition. Lyon: IARC Press; 2017.
  218. Von Wasielewski S, Franklin J, Fischer R, et al. Nodular sclerosing Hodgkin disease: new grading predicts prognosis in intermediate and advanced stages. Blood. 2003;101(10):4063–9. doi: 10.1182/blood-2002-05-1548.
  219. Pinto A, Aldinucci D, Gloghini A, et al. The role of eosinophils in the pathobiology of Hodgkin’s disease. Ann Oncol. 1997;8(Suppl 2):89–96.
  220. Enblad G, Sundstrom C, Glimelius B. Infiltration of eosinophils in Hodgkin’s disease involved lymph nodes predicts prognosis. Hematol Oncol. 1993;11(4):187–93. doi: 10.1002/hon.2900110404.
  221. Von Wasielewski R, Seth S, Franklin J, et al. Tissue eosinophilia correlates strongly with poor prognosis in nodular sclerosing Hodgkin’s disease, allowing for known prognostic factors. Blood. 2000;95(4):1207–13.
  222. Konjevic G, Jurisic V, Jovic V, et al. Investigation of NK cell function and their modulation in different malignancies. Immunol Res. 2012;52(1–2):139–56. doi: 10.1007/s12026-012-8285-7.
  223. Chiu J, Ernst DM, Keating A. Acquired Natural Killer Cell Dysfunction in the Tumor Microenvironment of Classic Hodgkin Lymphoma. Front Immunol. 2018;9:267. doi: 10.3389/fimmu.2018.00267.
  224. Reiners KS, Kessler J, Sauer M, et al. Rescue of impaired NK cell activity in Hodgkin lymphoma with bispecific antibodies in vitro and in patients. Mol Ther. 2013;21(4):895–903. doi: 10.1038/mt.2013.14.
  225. Tursz T, Dokhelar MC, Lipinski M, Amiel JL. Low natural killer cell activity in patients with malignant lymphoma. Cancer. 1982;50(11):2333–5. doi: 10.1002/1097-0142(19821201)50:11<2333::aid-cncr2820501119>3.0.co;2-w.
  226. Alvaro-Naranjo T, Lejeune M, Salvado-Usach MT, et al. Tumor-infiltrating cells as a prognostic factor in Hodgkin’s lymphoma: a quantitative tissue microarray study in a large retrospective cohort of 267 patients. Leuk Lymphoma. 2005;46(11):1581–91. doi: 10.1080/10428190500220654.
  227. Zanna MY, Yasmin AR, Omar AR, et al. Review of Dendritic Cells, Their Role in Clinical Immunology, and Distribution in Various Animal Species. Int J Mol Sci. 2021;22(15):8044. doi: 10.3390/ijms22158044.
  228. Galati D, Zanotta S, Corazzelli G, et al. Circulating dendritic cells deficiencies as a new biomarker in classical Hodgkin lymphoma. Br J Haematol. 2019;184(4):594–604. doi: 10.1111/bjh.15676.
  229. Tudor CS, Bruns H, Daniel C, et al. Macrophages and dendritic cells as actors in the immune reaction of classical Hodgkin lymphoma. PLoS One. 2014;9(12):e114345. doi: 10.1371/journal.pone.0114345.
  230. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21(8):485–98. doi: 10.1038/s41577-020-00490-y.
  231. Romano A, Parrinello NL, Vetro C, et al. Circulating myeloid-derived suppressor cells correlate with clinical outcome in Hodgkin Lymphoma patients treated up-front with a risk-adapted strategy. Br J Haematol. 2015;168(5):689–700. doi: 10.1111/bjh.13198.
  232. Novosad O, Gorbach O, Skachkova O, et al. Role of Circulating Myeloid-Derived Suppressor Cell (MDSC) in Hodgkin Lymphoma (HL) Progression: Updated Prospective Study. Blood. 2020;136(Suppl 1):31. doi: 10.1182/blood-2020-141259.
  233. Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022;12(1):31–46. doi: 10.1158/2159-8290.CD-21-1059.
  234. Korkolopoulou P, Thymara I, Kavantzas N, et al. Angiogenesis in Hodgkin’s lymphoma: a morphometric approach in 286 patients with prognostic implications. Leukemia. 2005;19(6):894–900. doi: 10.1038/sj.leu.2403690.
  235. Karihtala K, Leivonen SK, Karjalainen-Lindsberg ML, et al. T026: Characterization of cancer-associated fibroblasts in classical Hodgkin lymphoma. Hemasphere. 2022;6(Suppl):13. doi: 10.1097/01.HS9.0000890672.81592.33.
  236. Bankov K, Doring C, Ustaszewski A, et al. Fibroblasts in Nodular Sclerosing Classical Hodgkin Lymphoma Are Defined by a Specific Phenotype and Protect Tumor Cells from Brentuximab-Vedotin Induced Injury. Cancers (Basel). 2019;11(11):1687. doi: 10.3390/cancers11111687.
  237. Kamper P, Bendix K, Hamilton-Dutoit S, et al. Tumor-infiltrating macrophages correlate with adverse prognosis and Epstein-Barr virus status in classical Hodgkin’s lymphoma. Haematologica. 2011;96(2):269–76. doi: 10.3324/haematol.2010.031542.
  238. Tan KL, Scott DW, Hong F, et al. Tumor-associated macrophages predict inferior outcomes in classic Hodgkin lymphoma: a correlative study from the E2496 Intergroup trial. Blood. 2012;120(16):3280–7. doi: 10.1182/blood-2012-04-421057.
  239. Azambuja D, Natkunam Y, Biasoli I, et al. Lack of association of tumor-associated macrophages with clinical outcome in patients with classical Hodgkin’s lymphoma. Ann Oncol. 2012;23(3):736–42. doi: 10.1093/annonc/mdr157.
  240. Werner L, Dreyer JH, Hartmann D, et al. Tumor-associated macrophages in classical Hodgkin lymphoma: hormetic relationship to outcome. Sci Rep. 2020;10(1):9410. doi: 10.1038/s41598-020-66010-z.
  241. Panico L, Ronconi F, Lepore M, et al. Prognostic role of tumor-associated macrophages and angiogenesis in classical Hodgkin lymphoma. Leuk Lymphoma. 2013;54(11):2418–25. doi: 10.3109/10428194.2013.778405.
  242. Ng WL, Ansell SM, Mondello P. Insights into the tumor microenvironment of B cell lymphoma. J Exp Clin Cancer Res. 2022;41(1):362. doi: 10.1186/s13046-022-02579-9.
  243. Chen X, Cho DB, Yang PC. Double staining immunohistochemistry. N Am J Med Sci. 2010;2(5):241–5. doi: 10.4297/najms.2010.2241.
  244. Parra ER, Uraoka N, Jiang M, et al. Validation of multiplex immunofluorescence panels using multispectral microscopy for immune-profiling of formalin-fixed and paraffin-embedded human tumor tissues. Sci Rep. 2017;7(1):13380. doi: 10.1038/s41598-017-13942-8.
  245. Ijsselsteijn ME, van der Breggen R, Farina Sarasqueta A, et al. A 40-Marker Panel for High Dimensional Characterization of Cancer Immune Microenvironments by Imaging Mass Cytometry. Front Immunol. 2019;10:2534. doi: 10.3389/fimmu.2019.02534.
  246. Marx V. Method of the Year: spatially resolved transcriptomics. Nat Methods. 2021;18:9–14. doi: 10.1038/s41592-020-01033-y.
  247. Lee MKI, Rabindranath M, Faust K, et al. Compound computer vision workflow for efficient and automated immunohistochemical analysis of whole slide images. J Clin Pathol. 2022:jclinpath-2021-208020. doi: 10.1136/jclinpath-2021-208020.
  248. Wilson CM, Ospina OE, Townsend MK, et al. Challenges and Opportunities in the Statistical Analysis of Multiplex Immunofluorescence Data. Cancers (Basel). 2021;13(12):3031. doi: 10.3390/cancers13123031.
  249. M’kacher R, Frenzel M, Al Jawhari M, et al. Establishment and Characterization of a Reliable Xenograft Model of Hodgkin Lymphoma Suitable for the Study of Tumor Origin and the Design of New Therapies. Cancers (Basel). 2018;10(11):414. doi: 10.3390/cancers10110414.

Perspectives for the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma

SV Semochkin1,2

1 PA Gertsen Moscow Oncology Research Institute, branch of the NMRC of Radiology, 3 2-i Botkinskii pr-d, Moscow, Russian Federation, 125284

2 NI Pirogov Russian National Research Medical University, 1 Ostrovityanova ul., Moscow, Russian Federation, 117997

For correspondence: Prof. Sergei Vyacheslavovich Semochkin, MD, PhD, 3 2-i Botkinskii pr-d, Moscow, Russian Federation, 125284; e-mail: semochkin_sv@rsmu.ru

For citation: Semochkin SV. Perspectives for the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma. Clinical oncohematology. 2023;16(3):229–41. (In Russ).

DOI: 10.21320/2500-2139-2023-16-3-229-241


ABSTRACT

In recent decades, the progress in multiple myeloma (MM) treatment has been linked to a clearer insight into the biology of this disease and practical application of new pharmaceutical classes, such as immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and monoclonal antibodies (MABs). Modern IMiDs (lenalidomide and pomalidomide) are thalidomide derivatives which despite the similarity of chemical structure show only a relative cross-resistance. Lenalidomide is a second-generation immunomodulator with high anti-tumor activity and a favorable safety profile. In 2006, the use of lenalidomide combined with dexamethasone (Rd regimen) was approved by FDA (USA) for the treatment of relapsed/refractory MM, and 9 years later, in 2015, for newly diagnosed MM. During 2015–2019, the treatment of relapsed MM applied the newly developed regimens involving Rd combined with bortezomib (VRd), carfilzomib (KRd), ixazomib (IRd), elotuzumab (ERd), and daratumumab (DRd), the so-called triplets. Pomalidomide is a third-generation drug used in lenalidomide-refractory patients. For patients with relapsed/refractory MM who received at least two therapy lines with lenalidomide and bortezomib, regimens with 3 drugs were introduced which include pomalidomide and dexamethasone combined with elotuzumab (EPd), isatuximab (Isa-Pd), and daratumumab (DPd). In 2010, the molecular target of IMiD action was discovered, that is protein cereblon (CRBN), a component of CRBN E3 ligase enzyme complex. The insight into this mechanism provided the basis for developing a new family of thalidomide derivatives which are now called CRBN E3 ligase modulators (CELMoDs). In phase I/II trials, two drugs belonging to this group (iberdomide and mezigdomide) showed promising activity in MM refractory to three classes of antitumor drugs (IMiDs, PIs, and anti-CD38 MABs). The present review is focused on prospective studies of IMiDs and CELMoDs at different stages of MM treatment.

Keywords: multiple myeloma, immunomodulatory drugs, cereblon E3 ligase modulators, lenalidomide, pomalidomide, iberdomide, mezigdomide.

Received: January 25, 2023

Accepted: May 28, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Менделеева Л.П., Вотякова О.М., Рехтина И.Г. и др. Множественная миелома. Современная онкология. 2020;22(4):6–28. doi: 10.26442/18151434.2020.4.200457.
    [Mendeleeva LP, Votjakova OM, Rehtina IG, et al. Multiple myeloma. Clinical recommendations. Journal of Modern Oncology. 2020;22(4):6–28. doi: 10.26442/18151434.2020.4.200457. (In Russ)]
  2. Usmani SZ, Hoering A, Cavo M, et al. Clinical predictors of long-term survival in newly diagnosed transplant eligible multiple myeloma—An IMWG Research Project. Blood Cancer J. 2018;8(12):123. doi: 10.1038/s41408-018-0155-7.
  3. Rajkumar SV. Multiple myeloma: Every year a new standard? Hematol Oncol. 2019;37(Suppl 1):62–5. doi: 10.1002/hon.2586.
  4. Семочкин С.В. Биологические основы применения иммуномодулирующих препаратов в лечении множественной миеломы. Онкогематология. 2010;(1):21–31. doi: 10.17650/1818-8346-2010-0-1-.
    [Semochkin SV. Biological basis of immunomodulatory preparations using in treatment of multiple myeloma. Oncohematology. 2010;(1):21–31. doi: 10.17650/1818-8346-2010-0-1-. (In Russ)]
  5. Lenz W, Knapp K. Thalidomide embryopathy. Dtsch Med Wochenschr. 1962;87(24):1232–42. doi: 10.1055/s-0028-1111892.
  6. Kelsey Thalidomide update: regulatory aspects. Teratology. 1988;38(3):221–6. doi: 10.1002/tera.1420380305.
  7. Barlogie B, Desikan R, Eddlemon P, et al. Extended survival in advanced and refractory multiple myeloma after single-agent thalidomide: Identification of prognostic factors in a phase 2 study of 169 patients. Blood. 2001;98(2):492–4. doi: 10.1182/blood.v98.2.492.
  8. Torre CD, Zambello R, Cacciavillani M, et al. Lenalidomide long-term neurotoxicity: Clinical and neurophysiologic prospective study. Neurology. 2016;87(11):1161–6. doi: 10.1212/WNL.0000000000003093.
  9. Fotiou D, Gavriatopoulou M, Terpos E, Dimopoulos MA. Pomalidomide- and dexamethasone-based regimens in the treatment of refractory/relapsed multiple myeloma. Ther Adv Hematol. 2022;13:20406207221090089. doi: 10.1177/20406207221090089.
  10. Charlinski G, Vesole DH, Jurczyszyn A. Rapid Progress in the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma. Cancers (Basel). 2021;13(18):4666. doi: 10.3390/cancers13184666.
  11. Barankiewicz J, Salomon-Perzynski A, Misiewicz-Krzeminska I, Lech-Maranda E. CRL4CRBN E3 Ligase Complex as a Therapeutic Target in Multiple Myeloma. Cancers (Basel). 2022;14(18):4492. doi: 10.3390/cancers14184492.
  12. Mori T, Ito T, Liu S, et al. Structural basis of thalidomide enantiomer binding to cereblon. Sci Rep. 2018;8(1):1294. doi: 10.1038/s41598-018-19202-7.
  13. Kronke J, Udeshi ND, Narla A, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 2014;343(6168):301–5. doi: 10.1126/science.1244851.
  14. Lu G, Middleton RE, Sun H, et al. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of Ikaros proteins. Science. 2014;343(6168):305–9. doi: 10.1126/science.1244917.
  15. Read KA, Jones DM, Freud AG, Oestreich KJ. Established and emergent roles for Ikaros transcription factors in lymphoid cell development and function. Immunol Rev. 2021;300(1):82–99. doi: 10.1111/imr.12936.
  16. Bjorklund CC, Lu L, Kang J, et al. Rate of CRL4(CRBN) substrate Ikaros and Aiolos degradation underlies differential activity of lenalidomide and pomalidomide in multiple myeloma cells by regulation of c-Myc and IRF4. Blood Cancer J. 2015;5(10):e354. doi: 10.1038/bcj.2015.66.
  17. Harada T, Ozaki S, Oda A, et al. Association of Th1 and Th2 cytokines with transient inflammatory reaction during lenalidomide plus dexamethasone therapy in multiple myeloma. Int J Hematol. 2013;97(6):743–8. doi: 10.1007/s12185-013-1321-0.
  18. Gandhi AK, Kang J, Havens CG, et al. Immunomodulatory agents lenalidomide and pomalidomide co-stimulate T cells by inducing degradation of T cell repressors Ikaros and Aiolos via modulation of the E3 ubiquitin ligase complex CRL4 (CRBN). Br J Haematol. 2014;164(6):811–21. doi: 10.1111/bjh.12708.
  19. Lagrue K, Carisey A, Morgan DJ, et al. Lenalidomide augments actin remodeling and lowers NK-cell activation thresholds. Blood. 2015;126(1):50–60. doi: 10.1182/blood-2015-01-625004.
  20. Davies FE, Raje N, Hideshima T, et al. Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood. 2001;98(1):210–6. doi: 10.1182/blood.V98.1.210.
  21. Fabro S, Schumacher H, Smith RL, et al. The metabolism of thalidomide: some biological effects of thalidomide and its metabolites. Br J Pharmacol Chemother. 1965;25(2):352–62. doi: 10.1111/j.1476-5381.1965.tb02055.x.
  22. Yamamoto J, Ito T, Yamaguchi Y, Handa H. Discovery of CRBN as a target of thalidomide: a breakthrough for progress in the development of protein degraders. Chem Soc Rev. 2022;51(15):6234–50. doi: 10.1039/d2cs00116k.
  23. Connarn JN, Hwang R, Gao Y, et al. Population Pharmacokinetics of Lenalidomide in Healthy Volunteers and Patients With Hematologic Malignancies. Clin Pharmacol Drug Dev. 2018;7(5):465–73. doi: 10.1002/cpdd.372.
  24. Kronke J, Fink EC, Hollenbach PW, et al. Lenalidomide induces ubiquitination and degradation of CK1α in del(5q) MDS. Nature. 2015;523(7559):183–8. doi: 10.1038/nature14610.
  25. Li Y, Wang X, O’Mara E, et al. Population pharmacokinetics of pomalidomide in patients with relapsed or refractory multiple myeloma with various degrees of impaired renal function. Clin Pharmacol. 2017;9:133–45. doi: 10.2147/CPAA.S144606.
  26. Shimizu N, Asatsuma-Okumura T, Yamamoto J, et al. PLZF and its fusion proteins are pomalidomide-dependent CRBN neosubstrates. Commun Biol. 2021;4(1):1277. doi: 10.1038/s42003-021-02801-y.
  27. Kasserra C, Assaf M, Hoffmann M, et al. Pomalidomide: evaluation of cytochrome P450 and transporter-mediated drug-drug interaction potential in vitro and in healthy subjects. J Clin Pharmacol. 2015;55(2):168–78. doi: 10.1002/jcph.384.
  28. Matyskiela ME, Zhang W, Man HW, et al. A cereblon modulator (CC-220) with improved degradation of Ikaros and Aiolos. J Med Chem. 2018;61(2):535–42. doi: 10.1021/acs.jmedchem.6b01921.
  29. Gooding S, Ansari-Pour N, Towfic F, et al. Multiple cereblon genetic changes are associated with acquired resistance to lenalidomide or pomalidomide in multiple myeloma. Blood. 2021;137(2):232–7. doi: 10.1182/blood.2020007081.
  30. Durie BGM, Hoering A, Sexton R, et al. Longer term follow-up of the randomized phase III trial SWOG S0777: bortezomib, lenalidomide and dexamethasone vs. lenalidomide and dexamethasone in patients (Pts) with previously untreated multiple myeloma without an intent for immediate autologous stem cell transplant (ASCT). Blood Cancer J. 2020;10(5):53. doi: 10.1038/s41408-020-0311-8.
  31. Perrot A, Lauwers-Cances V, Cazaubiel T, et al. Early versus late autologous stem cell transplant in newly diagnosed multiple myeloma: long-term follow-up analysis of the IFM 2009 trial. Blood. 2020;136(Suppl 1):39. doi: 10.1182/blood-2020-134538.
  32. Richardson PG, Jacobus SJ, Weller EA, et al. Lenalidomide, bortezomib, and dexamethasone (RVd) ± autologous stem cell transplantation (ASCT) and R maintenance to progression for newly diagnosed multiple myeloma (NDMM): The phase 3 DETERMINATION trial. J Clin Oncol. 2022;40(17_suppl):LBA4. doi: 10.1200/jco.2022.40.17_suppl.lba4.
  33. Kumar SK, Jacobus SJ, Cohen AD, et al. Carfilzomib or bortezomib in combination with lenalidomide and dexamethasone for patients with newly diagnosed multiple myeloma without intention for immediate autologous stem-cell transplantation (ENDURANCE): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. 2020;21(10):1317–30. doi: 10.1016/S1470-2045(20)30452-6.
  34. Tan C, Nemirovsky D, Derkach A, et al. Carfilzomib, Lenalidomide and Dexamethasone (KRd) Vs Bortezomib, Lenalidomide, and Dexamethasone (VRd) As Induction Therapy in Newly Diagnosed High-Risk Multiple Myeloma. Blood. 2022;140(Suppl 1):1817–9. doi: 10.1182/blood-2022-169161.
  35. Gay F, Musto P, Rota-Scalabrini D, et al. Carfilzomib with cyclophosphamide and dexamethasone or lenalidomide and dexamethasone plus autologous transplantation or carfilzomib plus lenalidomide and dexamethasone, followed by maintenance with carfilzomib plus lenalidomide or lenalidomide alone for patients with newly diagnosed multiple myeloma (FORTE): a randomised, open-label, phase 2 trial. Lancet Oncol. 2021;22(12):1705–20. doi: 10.1016/S1470-2045(21)00535-0.
  36. Семочкин С.В. Новые ингибиторы протеасомы в терапии множественной миеломы. Онкогематология. 2019;14(2):29–40. doi: 10.17650/1818-8346-2019-14-2-29-40.
    [Semochkin SV. New proteasome inhibitors in the management of multiple myeloma. Oncohematology. 2019;14(2):29–40. doi: 10.17650/1818-8346-2019-14-2-29-40. (In Russ)]
  37. Goldschmidt H, Mai EK, Bertschet U, et al. Elotuzumab in Combination with Lenalidomide, Bortezomib, Dexamethasone and Autologous Transplantation for Newly-Diagnosed Multiple Myeloma: Results from the Randomized Phase III GMMG-HD6 Trial. Blood. 2021;138(Suppl 1):486. doi: 10.1182/blood-2021-147323.
  38. Usmani SZ, Hoering A, Ailawadhi S, et al. Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): primary analysis of a randomised, phase 2 trial. Lancet Haematol. 2021;8(1):e45–e54. doi: 10.1016/S2352-3026(20)30354-9.
  39. Laubach JP, Kaufman JL, Sborov DW, et al. Daratumumab (DARA) plus lenalidomide, bortezomib, and dexamethasone (RVd) in patients (Pts) with transplant-eligible newly diagnosed multiple myeloma (NDMM): updated analysis of Griffin after 24 months of maintenance. Blood. 2021;138 (Suppl 1):79. doi: 10.1182/blood-2021-149024.
  40. Goldschmidt H, Mai EK, Bertsch U, et al. Addition of isatuximab to lenalidomide, bortezomib, and dexamethasone as induction therapy for newly diagnosed, transplantation-eligible patients with multiple myeloma (GMMG-HD7): part 1 of an open-label, multicentre, randomised, active-controlled, phase 3 trial. Lancet Haematol. 2022;9(11):e810–e821. doi: 10.1016/S2352-3026(22)00263-0.
  41. McCarthy PL, Holstein SA, Petrucci MT, et al. Lenalidomide Maintenance After Autologous Stem-Cell Transplantation in Newly Diagnosed Multiple Myeloma: A Meta-Analysis. J Clin Oncol. 2017;35(29):3279–89. doi: 10.1200/JCO.2017.72.6679.
  42. Jackson GH, Davies FE, Pawlyn C, et al. Lenalidomide maintenance versus observation for patients with newly diagnosed multiple myeloma (Myeloma XI): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2019;20(1):57–73. doi: 10.1016/S1470-2045(18)30687-9.
  43. Pawlyn C, Menzies T, Davies FE, et al. Defining the Optimal Duration of Lenalidomide Maintenance after Autologous Stem Cell Transplant – Data from the Myeloma XI Trial. Blood. 2022;140(Suppl 1):1371–2. doi: 10.1182/blood-2022-165376.
  44. Facon T, Dimopoulos MA, Dispenzieri A, et al. Final analysis of survival outcomes in the phase 3 FIRST trial of up-front treatment for multiple myeloma. Blood. 2018;131(3):301–10. doi: 10.1182/blood-2017-07-795047.
  45. Facon T, Venner CP, Bahlis NJ, et al. Oral ixazomib, lenalidomide, and dexamethasone for transplant-ineligible patients with newly diagnosed multiple myeloma. Blood. 2021;137(26):3616–28. doi: 10.1182/blood.2020008787.
  46. Kumar SK, Moreau P, Bahlis NJ, et al. Daratumumab Plus Lenalidomide and Dexamethasone (D-Rd) Versus Lenalidomide and Dexamethasone (Rd) Alone in Transplant-Ineligible Patients with Newly Diagnosed Multiple Myeloma (NDMM): Updated Analysis of the Phase 3 Maia Study. Blood. 2022;140(Suppl 1):10150–3. doi: 10.1182/blood-2022-163335.
  47. Moreau P, Kumar SK, Miguel JS, et al. Treatment of relapsed and refractory multiple myeloma: recommendations from the International Myeloma Working Group. Lancet Oncol. 2021;22(3):e105–e118. doi: 10.1016/S1470-2045(20)30756-7.
  48. Moreau P, Masszi T, Grzasko N, et al. Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med. 2016;374(17):1621–34. doi: 10.1056/NEJMoa1516282.
  49. Richardson PG, Kumar SK, Masszi T, et al. Final Overall Survival Analysis of the TOURMALINE-MM1 Phase III Trial of Ixazomib, Lenalidomide, and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma. J Clin Oncol. 2021;39(22):2430–42. doi: 10.1200/JCO.21.00972.
  50. Lonial S, Dimopoulos M, Palumbo A, et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N Engl J Med. 2015;373(7):621–31. doi: 10.1056/NEJMoa1505654.
  51. Dimopoulos MA, Lonial S, White D, et al. Elotuzumab, lenalidomide, and dexamethasone in RRMM: final overall survival results from the phase 3 randomized ELOQUENT-2 study. Blood Cancer J. 2020;10(9):91. doi: 10.1038/s41408-020-00357-4.
  52. Stewart AK, Rajkumar SV, Dimopoulos MA, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med. 2015;372(2):142–52. doi: 10.1056/NEJMoa1411321.
  53. Siegel DS, Dimopoulos MA, Ludwig H, et al. Improvement in Overall Survival With Carfilzomib, Lenalidomide, and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma. J Clin Oncol 2018;36(8):728–34. doi: 10.1200/JCO.2017.76.5032.
  54. Bahlis NJ, Dimopoulos MA, White DJ, et al. Daratumumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a randomized, open-label, phase 3 study. 2020;34(7):1875–84. doi: 10.1038/s41375-020-0711-6.
  55. Dimopoulos MA, Oriol A, Nahi H, et al. Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with previously treated multiple myeloma: overall survival results from phase 3 POLLUX trial. Hemasphere. 2022;6(Suppl):13. doi: 10.1097/01.HS9.0000829592.26407.09.
  56. Manda S, Yimer HA, Noga SJ, et al. Feasibility of Long-term Proteasome Inhibition in Multiple Myeloma by in-class Transition From Bortezomib to Ixazomib. Сlin Lymphoma Myeloma Leuk. 2020;20(11):e910–e925. doi: 10.1016/j.clml.2020.06.024.
  57. Бессмельцев С.С. Режимы на основе помалидомида и дексаметазона при лечении рефрактерной/рецидивирующей множественной миеломы. Вестник гематологии. 2022;18(4):4–20.
    [Bessmeltsev SS. Pomalidomide- and dexamethasone-based regimens in the treatment of refractory/relapsed multiple myeloma. Vestnik gematologii. 2022;18(4):4–20. (In Russ)]
  58. Richardson PG, Oriol A, Beksac M, et al. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): a randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(6):781–94. doi: 10.1016/S1470-2045(19)30152-4.
  59. Dimopoulos MA, Terpos E, Boccadoro M, et al. Daratumumab plus pomalidomide and dexamethasone versus pomalidomide and dexamethasone alone in previously treated multiple myeloma (APOLLO): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22(6):801–12. doi: 10.1016/S1470-2045(21)00128-5.
  60. Dimopoulos MA, Terpos E, Boccadoro M, et al. Subcutaneous daratumumab plus pomalidomide and dexamethasone (D-Pd) versus pomalidomide and dexamethasone (Pd) alone in patients with relapsed or refractory multiple myeloma (RRMM): overall survival results from the phase 3 APOLLO study. Blood. 2022;140(Suppl 1):7272–4. doi 10.1182/blood-2022–163483.
  61. Attal M, Richardson PG, Rajkumar SV, et al. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet. 2019;394(10214):2096–107. doi: 10.1016/S0140-6736(19)32556-5.
  62. Richardson PG, Perrot A, San-Miguel J, et al. Isatuximab Plus Pomalidomide/Low-Dose Dexamethasone Versus Pomalidomide/Low-Dose Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma (ICARIA-MM): Characterization of Subsequent Antimyeloma Therapies. Blood. 2022;140(Suppl 1):608–10. doi: 10.1182/blood-2022-159710.
  63. Dimopoulos MA, Dytfeld D, Grosicki S, et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. N Engl J Med. 2018;379(19):1811–22. doi: 10.1056/NEJMoa1805762.
  64. Dimopoulos MA, Dytfeld D, Grosicki S, et al. Elotuzumab Plus Pomalidomide and Dexamethasone for Relapsed/Refractory Multiple Myeloma: Final Overall Survival Analysis From the Randomized Phase II ELOQUENT-3 Trial. J Clin Oncol. 2023;41(3):568–78. doi: 10.1200/JCO.21.02815.
  65. Pasvolsky O, Yeshurun M, Fraser R, et al. Maintenance therapy after second autologous hematopoietic cell transplantation for multiple myeloma. A CIBMTR analysis. Bone Marrow Transplant. 2022;57(1):31–7. doi: 10.1038/s41409-021-01455-y.
  66. Garderet L, Kuhnowski F, Berge В, et al. Phase II Study of the Combination of Pomalidomide with Dexamethasone As Maintenance Therapy after First Relapse Treatment with PCD Followed or Not By Autologous Stem Cell Transplant in Multiple Myeloma Patients. Blood. 2021;138(Suppl 1):2753. doi: 10.1182/blood-2021-152136.
  67. Thakurta A, Pierceall WE, Amatangelo MD, et al. Developing next generation immunomodulatory drugs and their combinations in multiple myeloma. Oncotarget. 2021;12(15):1555–63. doi: 10.18632/oncotarget.27973.
  68. Ye Y, Gaudy A, Schafer P, et al. First-in-Human, Single- and Multiple-Ascending-Dose Studies in Healthy Subjects to Assess Pharmacokinetics, Pharmacodynamics, and Safety/Tolerability of Iberdomide, a Novel Cereblon E3 Ligase Modulator. Clin Pharmacol Drug Dev. 2021;10(5):471–85. doi: 10.1002/cpdd.869.
  69. You W, Pang J. Pharmacokinetics, bioavailability and metabolism of CC-92480 in rat by liquid chromatography combined with electrospray ionization tandem mass spectrometry. Biomed Chromatogr. 2021;35(9):e5139. doi: 10.1002/bmc.5139.
  70. Van de Donk NWC, Popat R, Hulin C, et al. Results from CC-220-MM-001 Dose-expansion Phase of Iberdomide plus Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma. Hemasphere. 2022;6:14–5. doi: 10.1097/01.HS9.0000829600.37424.88.
  71. Lonial S, Abdallah A, Anwer F, et al. Iberdomide (IBER) in Combination with Dexamethasone (DEX) in Relapsed/Refractory Multiple Myeloma (RRMM): Results from the Anti-B-Cell Maturation Antigen (BCMA)-Exposed Cohort of the CC-220-MM-001 Trial. Blood. 2022;140(Suppl 1):4398–400. doi: 10.1182/blood-2022-158180.
  72. Richardson PG, Trudel S, Quach H, et al. Mezigdomide (CC-92480), a Potent, Novel Cereblon E3 Ligase Modulator (CELMoD), Combined with Dexamethasone (DEX) in Patients (pts) with Relapsed/Refractory Multiple Myeloma (RRMM): Preliminary Results from the Dose-Expansion Phase of the CC-92480-MM-001 Trial. Blood. 2022;140(Suppl 1):1366–8. doi: 10.1182/blood-2022-157945.
  73. Семочкин С.В. Механизмы действия противоопухолевых иммуномодуляторов — от тератогенности к терапии множественной миеломы. Гематология и трансфузиология. 2022;67(2):240–60. doi: 10.35754/0234-5730-2022-67-2-240-260.
    [Semochkin SV. Mechanisms of action of immunomodulatory drugs — from teratogenicity to treatment of multiple myeloma. Russian journal of hematology and transfusiology. 2022;67(2):240–60. doi: 10.35754/0234-5730-2022-67-2-240-260. (In Russ)]

The Prognostic Role of Genetic Aberrations in Mantle Cell Lymphoma: A Literature Review and Clinical Experience

EV Kleina1, SV Voloshin1,2, YuS Vokueva1, OD Petukhova1, EV Motyko1, MP Bakai1, DV Kustova1, AN Kirienko1, SYu Linnikov1, EV Karyagina3, OS Uspenskaya4, IS Zyuzgin5, SV Sidorkevich1, IS Martynkevich1

1 Russian Research Institute of Hematology and Transfusiology, 16 2-ya Sovetskaya ul., Saint Petersburg, Russian Federation, 191024

2 SM Kirov Military Medical Academy, 6 Akademika Lebedeva ul., Saint Petersburg, Russian Federation, 194044

3 Municipal Hospital No. 15, 4 Avangardnaya ul., Saint Petersburg, Russian Federation, 198205

4 Leningrad Regional Clinical Hospital, 45 korp. 2A Lunacharskogo pr-t, Saint Petersburg, Russian Federation, 194291

5 NN Petrov National Medical Cancer Research Center, 68 Leningradskaya ul., Pesochnyi pos., Saint Petersburg, Russian Federation, 197758

For correspondence: Elizaveta Vyacheslavovna Kleina, 16 2-ya Sovetskaya ul., Saint Petersburg, Russian Federation, 191024; e-mail: elizabeth.kleina@gmail.com

For citation: Kleina EV, Voloshin SV, Vokueva YuS, et al. The Prognostic Role of Genetic Aberrations in Mantle Cell Lymphoma: A Literature Review and Own Experience. Clinical oncohematology. 2023;16(2):213–26. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-213-226


ABSTRACT

Mantle cell lymphoma (MCL) is a type of peripheral B-cell non-Hodgkin’s lymphoma characterized by constitutive cyclin D1 overexpression leading to cell-cycle dysregulation and disruption of DNA damage repair. Apart from the typical translocation t(11;14)(q13;q32) and more rare variants, such as t(2;11)(p11;q13) and t(11;22)(q13;q11), a considerable number of patients quite often show secondary molecular and chromosomal aberrations underlying heterogeneity of the clinical course of MCL. Among a wide range of molecular genetic abnormalities, particular attention during the last years has been concentrated on studying the so-called double-hit MCL within a subgroup of patients with translocations involving CCND1 and MYC genes. Double-hit MCL is distinguished with rapid progression and tumor generalization at the time of diagnosis. Poor prognosis and low survival rates in most MCL patients call for the fastest possible diagnosis. Morphological and immunohistochemical as well as genetic methods (standard cytogenetic technique and fluorescence in situ hybridization) contribute to improving the quality of evidence-based diagnosis. The results of comprehensive diagnostic studies optimize prognosis assessment and treatment decision making in clinic.

Keywords: mantle cell lymphoma, double-hit MCL, karyotype, genetic aberrations, prognostic factors.

Received: August 16, 2022

Accepted: February 27, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Kallen ME, Rao NP, Kulkarni SK, et al. B-lymphoblastic transformation of mantle cell lymphoma/leukemia with “double hit” changes. J Hematopathol. 2015;8(1):31–6. doi: 10.1007/s12308-014-0229-9.
  2. Geling Li, Zhou Yi, Sindhu Ch, et al. An unusual case of co-existing classic mantle cell lymphoma and transformed lymphoma with Burkitt-like features with leukemic presentation. J Hematopathol. 2016;9(2):91–9. doi: 10.1007/s12308-016-0274-7.
  3. Delas A, Sophie D, Brousset P, Laurent C. Unusual concomitant rearrangements of Cyclin D1 and MYC genes in blastoid variant of mantle cell lymphoma: Case report and review of literature. Pathol Res Pract. 2013;209(2):115–9. doi: 10.1016/j.prp.2012.12.001.
  4. Sarkozy C, Terre C, Jardin F, et al. Complex karyotype in mantle cell lymphoma is a strong prognostic factor for the time to treatment and overall survival, independent of the MCL international prognostic index. Genes Chromosomes Cancer. 2014;53(1):106–16. doi: 10.1002/gcc.22123.
  5. Oka K, Ohno T, Kita K, et al. PRAD1 gene over-expression in mantle-cell lymphoma but not in other low-grade B-cell lymphomas, including extranodal lymphoma. Br J Haematol. 1994;86(4):786–91. doi: 10.1111/j.1365-2141.1994.tb04830.x.
  6. Rule S. The modern approach to mantle cell lymphoma. Hematol Oncol. 2019;37(1):66–9. doi: 10.1002/hon.2596.
  7. Geisler CH, Kolstad A, Laurell A, et al. The mantle cell lymphoma international prognostic index (MIPI) is superior to the international prognostic index (IPI) in predicting survival following intensive first-line immunochemotherapy and autologous stem cell transplantation (ASCT). Blood. 2010;115(8):1530–3. doi: 10.1182/blood-2009-08-236570.
  8. Vose JM. Mantle cell lymphoma: 2017 update on diagnosis, risk-stratification, and clinical management. Am J Hematol. 2017;92(8):806–13. doi: 10.1002/ajh.24797.
  9. Ladha A, Zhao J, Epner EM, Pu JJ. Mantle cell lymphoma and its management: where are we now? Exp Hematol Oncol. 2019;8:2. doi: 10.1186/s40164-019-0126-0.
  10. Sander B, Quintanilla-Martinez L, Ott G, et al. Mantle cell lymphoma – a spectrum from indolent to aggressive disease. Virchows Arch. 2016;468(3):245–57. doi: 10.1007/s00428-015-1840-6.
  11. Maddocks K. Update on mantle cell lymphoma. Blood. 2018;132(16):1647–56. doi: 10.1182/blood-2018-03-791392.
  12. Hoster E, Dreyling M, Klapper W, et al. German Low Grade Lymphoma Study Group (GLSG); European Mantle Cell Lymphoma Network. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood. 2008;111(2):558–65. doi: 10.1182/blood-2007-06-095331.
  13. Berard CW, Dorfman RF. Histopathology of malignant lymphomas. Clin Haematol. 1974;3:39–45.
  14. Miao Y, Lin P, Wang W, et al. CCND1-IGH Fusion-Amplification and MYC Copy Number Gain in a Case of Pleomorphic Variant Mantle Cell Lymphoma. Am J Clin Pathol. 2016;146(6):747–52. doi: 10.1093/ajcp/aqw194.
  15. Hanel W, Epperla N. Emerging therapies in mantle cell lymphoma. J Hematol Oncol. 2020;13(1):79. doi: 10.1186/s13045-020-00914-1.
  16. Reya T, O’Riordan M, Okamura R, et al. Wnt signaling regulates B lymphocyte proliferation through a LEF-1 dependent mechanism. Immunity. 2000;13(1):15–24. doi: 10.1016/s1074-7613(00)00004-2.
  17. Tandon B, Peterson L, Gao J, et al. Nuclear overexpression of lymphoid-enhancer-binding factor 1 identifies chronic lymphocytic leukemia/small lymphocytic lymphoma in small B-cell lymphomas. Mod Pathol. 2011;24(11):1433–43. doi: 10.1038/modpathol.2011.103.
  18. Menter T, Dirnhofer S, Tzankov A. LEF1: a highly specific marker for the diagnosis of chronic lymphocytic B cell leukaemia/small lymphocytic B cell lymphoma. J Clin Pathol. 2015;68(6):473–8. doi: 10.1136/jclinpath-2015-202862.
  19. Walther N, Ulrich A, Vockerodt M, et al. Aberrant lymphocyte enhancer-binding factor 1 expression is characteristic for sporadic Burkitt’s lymphoma. Am J Pathol. 2013;182(4):1092–8. doi: 10.1016/j.ajpath.2012.12.013.
  20. Mozos A, Royo C, Hartmann E. SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype. Haematologica. 2009;94(11):1555–62. doi: 10.3324/haematol.2009.010264.
  21. Lord M, Wasik AM, Christensson B, et al. The utility of mRNA analysis in defining SOX11 expression levels in mantle cell lymphoma and reactive lymph nodes. Haematologica. 2015;100(9):369–72. doi: 10.3324/haematol.2015.123885.
  22. Ek S, Dictor M, Jerkeman M, et al. Nuclear expression of the non B-cell lineage Sox11 transcription factor identifies mantle cell lymphoma. Blood. 2008;111(2):800–5. doi: 10.1182/blood-2007-06-093401.
  23. Wang X, Asplund AC, Porwit A. The subcellular Sox11 distribution pattern identifies subsets of mantle cell lymphoma: correlation to overall survival. Br J Haematol. 2008;143(2):248–52. doi: 10.1111/j.1365-2141.2008.07329.x.
  24. Royo C, Salaverria I, Hartmann EM, et al. The complex landscape of genetic alterations in mantle cell lymphoma. Semin Cancer Biol. 2011;21(5):322–34. doi: 10.1016/j.semcancer.2011.09.007.
  25. Klapper W, Hoster E, Determann O, et al. Ki-67 as a prognostic marker in mantle cell lymphoma-consensus guidelines of the pathology panel of the European MCL Network. J Hematop. 2009;2(2):103–11. doi: 10.1007/s12308-009-0036-x.
  26. Jares P, Colomer, D, Campo E, et al. Molecular pathogenesis of mantle cell lymphoma. J Clin Invest. 2012;122(10):3416–23. doi: 10.1172/JCI61272.
  27. Ishigaki T, Sasaki K, Watanabe K, et al. Amplification of IGH/CCND1 fusion gene in a primary plasma cell leukemia case. Cancer Genet Cytogenet. 2010;201(1):62–5. doi: 10.1016/j.cancergencyto.2010.05.006.
  28. Lovec H, Grzeschiczek A, Kowalski MB, et al. Cyclin D1/bcl-1 cooperates with myc genes in the generation of B-cell lymphoma in transgenic mice. EMBO J. 1994;13(15):3487–95. doi: 10.1002/j.1460-2075.1994.tb06655.x.
  29. Wang L, Tang G, Medeiros LJ, et al. MYC rearrangement but not extra MYC copies is an independent prognostic factor in patients with mantle cell lymphoma. Haematologica. 2021;106(5):1381–9. doi: 10.3324/haematol.2019.243071.
  30. Espinet B, Salaverria I, Bea S, et al. Incidence and prognostic impact of secondary cytogenetic aberrations in a series of 145 patients with mantle cell lymphoma. Genes Chromosomes Cancer. 2010;49(5):439–51. doi: 10.1002/gcc.20754.
  31. Nordstrom L, Sernbo S, Eden P, et al. SOX11 and TP53 add prognostic information to MIPI in a homogenously treated cohort of mantle cell lymphoma–a Nordic Lymphoma Group study. Br J Haematol. 2014;166(1):98–108. doi: 10.1111/bjh.12854.
  32. Aukema SM, Siebert R, Schuuring E, et al. Double-hit B-cell lymphomas. Blood. 2011;117(8):2319–31. doi: 10.1182/blood-2010-09-297879.
  33. Boerma EG, Siebert R, Kluin PM, et al. Translocations involving 8q24 in Burkitt lymphoma and other malignant lymphomas: a historical review of cytogenetics in the light of todays knowledge. Leukemia. 2009;23(2):225–34. doi: 10.1038/leu.2008.281.
  34. Gruszka-Westwood AM, Atkinson S, Summersgill BM, et al. Unusual case of leukemic mantle cell lymphoma with amplified CCND1/IGH fusion gene. Genes Chromosomes Cancer. 2002;33(2):206–12. doi: 10.1002/gcc.1216.
  35. Greenwell IB, Staton AD, Lee MJ, et al. Complex karyotype in patients with mantle cell lymphoma predicts inferior survival and poor response to intensive induction therapy. Cancer. 2018;124(11):2306–15. doi: 10.1002/cncr.31328.
  36. Halldorsdottir AM, Lundin A, Murray F, et al. Impact of TP53 mutation and 17p deletion in mantle cell lymphoma. Leukemia. 2011;25(12):1904–8. doi: 10.1038/leu.2011.162.
  37. Martin P, Chadburn A, Christos P, et al. Outcome of deferred initial therapy in mantle-cell lymphoma. J Clin Oncol. 2009;27(8):1209–13. doi: 10.1200/JCO.2008.19.6121.
  38. Abrisqueta P, Scott DW, Slack GW, et al. Observation as the initial management strategy in patients with mantle cell lymphoma. Ann Oncol. 2017;28(10):2489–95. doi: 10.1093/annonc/mdx333.
  39. Adam P, Schiefer AI, Prill S, et al. Incidence of preclinical manifestations of mantle cell lymphoma and mantle cell lymphoma in situ in reactive lymphoid tissues. Mod Pathol. 2012;25(12):1629–36. doi: 10.1038/modpathol.2012.117.
  40. Espinet B, Ferrer A, Bellosillo B, et al. Distinction between asymptomatic monoclonal B-cell lymphocytosis with cyclin D1 overexpression and mantle cell lymphoma: from molecular profiling to flow cytometry. Clin Cancer Res. 2014;20(4):1007–19. doi: 10.1158/1078-0432.CCR-13-1077.
  41. Lecluse Y, Lebailly P, Roulland S, et al. t(11;14)-positive clones can persist over a long period of time in the peripheral blood of healthy individuals. Leukemia. 2009;23(6):1190–3. doi: 10.1038/leu.2009.31.
  42. Karube K, Scarfo L, Campo E, Ghia P. Monoclonal B cell lymphocytosis and “in situ” lymphoma. Semin Cancer Biol. 2014;24:3–14. doi: 10.1016/j.semcancer.2013.08.003.
  43. Racke F, Simpson S, Christian B, et al. Evidence of Long Latency Periods Prior to Development of Mantle Cell Lymphoma. Blood. 2010;116(21):323. doi: 10.1182/blood.V116.21.323.323.
  44. Edlefsen KL, Greisman HA, Yi HS, et al. Early lymph node involvement by mantle cell lymphoma limited to the germinal center: report of a case with a novel “follicular in situ” growth pattern. Am J Clin Pathol. 2011;136(2):276–81. doi: 10.1309/AJCP6KFFGTC8PLVR.
  45. Chapman-Fredricks J, Sandoval-Sus J, Vega F, et al. Progressive leukemic non-nodal mantle cell lymphoma associated with deletions of TP53, ATM, and/or 13q14. Ann Diagn Pathol. 2014;18(4):214–9. doi: 10.1016/j.anndiagpath.2014.03.006.

Ibrutinib Efficacy in First-Line Therapy of High-Risk Patients Vs. Second- and Third-Line Therapies of Resistant Chronic Lymphocytic Leukemia

NV Kurkina1,2, EA Repina1, PV Volkova1, AA Repin1

1 NP Ogarev National Research Mordovia State University, 68 Bolshevistskaya ul., Saransk, Russian Federation, 430005

2 Republican Clinical Hospital No. 4, 32 Ul’yanova ul., Saransk, Russian Federation, 430032

For correspondence: Nadezhda Viktorovna Kurkina, MD, PhD, 32 Ul’yanova ul., Saransk, Russian Federation, 430032; Tel.: +7(927)172-48-63; e-mail: nadya.kurckina@yandex.ru

For citation: Kurkina NV, Repina EA, Volkova PV, Repin AA. Ibrutinib Efficacy in First-Line Therapy of High-Risk Patients Vs. Second- and Third-Line Therapies of Resistant Chronic Lymphocytic Leukemia. Clinical oncohematology. 2023;16(2):209–12. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-209-212


ABSTRACT

Risk stratification appears to be the most valid criterion in decision-making regarding optimal specific therapy in chronic lymphocytic leukemia (CLL). The CLL International Prognostic Index takes account of unfavorable cytogenetic abnormalities (del(17p)/del(11q) and/or TP53 gene mutations) as well as the mutation status of immunoglobulin heavy chain variable (IGHV) region genes. Unmutated V(H)-status is commonly associated with such prognostically unfavorable genetic markers as del(17p)/del(11q), trisomy 12, and TP53 mutation. The combination of unmutated V(H)-status with unfavorable karyotype abnormalities (del(17p)/del(11q)) negatively affects the prognosis and overall survival rate. Besides, in high-risk CLL, the efficacy of therapy is rather low, and the development of refractoriness is possible. Targeted therapy (Bruton tyrosine kinase inhibitors) both in first line and in resistant CLL considerably increases the probability of achieving long-term remission. The present paper provides the comparative analysis of clinical and hematological efficacy and tolerability of ibrutinib in first-line CLL therapy of high-risk patients as well as second- and third-line therapies of resistant CLL. Ibrutinib shows high efficacy and low toxicity. First-line ibrutinib treatment results in a faster response and effectively reduces the probability of CLL progression. Second- and third-line ibrutinib treatment allows to overcome CLL resistance without impairing patients’ quality of life.

Keywords: chronic lymphocytic leukemia, high-risk group, deletion del(17p), TP53 gene mutation, ibrutinib, efficacy, toxicity.

Received: October 9, 2022

Accepted: February 28, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Wierda WG, Byrd JC, Abramson JS, et al. Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma, Version 4.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2020;18(2):185–217. doi: 10.6004/jnccn.2020.0006.
  2. Hallek MJ, Cheson BD, Catovsky D, Caligaris-Cappio F. 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. Российские клинические рекомендации по диагностике и лечению лимфопролиферативных заболеваний. Под ред. И.В. Поддубной, В.Г. Савченко. М.: Буки Веди, 2018. С. 179–200.
    [Poddubnaya IV, Savchenko VG, eds. Rossiiskie klinicheskie rekomendatsii po diagnostike i lecheniyu limfoproliferativnykh zabolevanii. (Russian clinical guidelines on diagnosis and treatment of lymphoproliferative disorders.) Moscow: Buki Vedi Publ.; 2018. рp. 179–200. (In Russ)]
  4. The International CLL-IPI working group. An international prognostic index for patients with chronic lymphocytic leukaemia (CLL-IPI): a meta-analysis of individual patient data. Lancet Oncol. 2016;17(6):779–90. doi: 10.1016/S1470-2045(16)30029-8.
  5. Pflug N, Bahlo J, Shanafelt T, Eichhorst B. Development of a comprehensive prognostic index for patients with chronic lymphocytic leukemia. 2014;12(4):49–62. doi: 10.1182/blood-2014-02-556399.
  6. Zenz T, Eichhorst B, Busch R, et al. TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol. 2010;28(29):4473–9. doi: 10.1200/JCO.2009.27.8762.
  7. Rossi D, Khiabanian H, Spina V, et al. Clinical impact of small TP53 mutated subclones in chronic lymphocytic leukemia. Blood. 2014;123(14):2139–47. doi: 10.1182/blood-2013-11-539726.
  8. Никитин Е.А., Судариков А.Б. Хронический лимфолейкоз высокого риска: история, определение, диагностика и лечение. Клиническая онкогематология. 2013;6(1):59–67.
    [Nikitin EA, Sudarikov AB. High­risk chronic lymphocytic leukemia: history, definition, diagnosis, and management. Klinicheskaya onkogematologiya. 2013;6(1):59–67. (In Russ)]
  9. Strati P, Shanafelt TD. Monoclonal B-cell lymphocytosis and early-stage chronic lymphocytic leukemia: Diagnosis, natural history, and risk stratification. Blood. 2015;126(4):454–62. doi: 10.1182/blood-2015-02-585059.
  10. Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373(25):2425–37. doi: 10.1056/NEJMoa1509388.
  11. Woyach JA, Ruppert AS, Heerema NA, et al. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med. 2018;379(26):2517–28. doi: 10.1056/NEJMoa1812836.

Plasma Cell-Free DNA in Patients with Diffuse Large B-Cell and B-Cell High-Grade (Double Hit/Triple Hit) Lymphomas

SYu Smirnova, EE Nikulina, NG Gabeeva, DA Koroleva, SA Tatarnikova, AK Smol’yaninova, EG Gemdzhian, EE Zvonkov, AB Sudarikov

National Research Center for Hematology, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Svetlana Yurevna Smirnova, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(926)879-65-94; e-mail: smirnova-s-ju@yandex.ru

For citation: Smirnova SYu, Nikulina EE, Gabeeva NG, et al. Plasma Cell-Free DNA in Patients with Diffuse Large B-Cell and B-Cell High-Grade (Double Hit/Triple Hit) Lymphomas. Clinical oncohematology. 2023;16(2):200–8. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-200-208


ABSTRACT

Aim. To study plasma cell-free DNA (pcfDNA) concentration and B-cell clonality in patients with diffuse large B-cell (DLBCL) and B-cell high-grade lymphomas prior to and at different stages of chemotherapy as well as the correlation between the data obtained and clinical and laboratory parameters.

Materials & Methods. The study enrolled 23 DLBCL patients and 7 healthy donors (HD). Plasma was prepared from whole blood by centrifugation, pcfDNA was isolated with the commercial kit Qiagen (Germany). The concentration of pcfDNA was determined using fluorometer Qubit (USA). В-cell clonality was estimated by immunoglobulin gene analysis (BIOMED-2 protocol) in the tumor tissue and bone marrow core biopsy specimens obtained on diagnosis date as well as in the pcfDNA at 5 end points: prior to chemotherapy and after cycles 1, 2, 3, and 4.

Results. Prior to therapy, all DLBCL patients showed significantly higher pcfDNA concentration than HD. Immunochemotherapy cycle 1 resulted in considerable increase in pcfDNA concentration. After cycle 2 and subsequent cycles, pcfDNA concentration gradually decreased. After cycle 4, the mean pcfDNA concentration was comparable with that of HD. In 95 % of patients В-cell clonality in pcfDNA corresponded to that identified in the tumor specimen. After immunochemotherapy cycle 1, В-cell clonality was detected in 50 % of patients, after cycle 2 it was shown by 15 %. Only 1 female patient retained В-cell clonality after therapy cycles 3 and 4. In HD, no В-cell clonality in pcfDNA was identified. Prior to therapy, the analysis revealed no correlation of either pcfDNA concentration or В-cell clonality in pcfDNA with age, sex, tumor spread, presence or absence of extranodal lesions, proliferation index Ki-67, and lactate dehydrogenase concentration.

Conclusion. In patients with malignant hematological tumors, pcfDNA seems to be an interesting, easily accessible biological material deserving further investigation. Any studies of pcfDNA require long-term dynamical analysis and standardized methods of collection, storage and processing of the data obtained. In the long run, with more and more information, pcfDNA can become an important diagnostic marker of tumor heterogeneity and a reliable relapse predictor.

Keywords: cell-free DNA, plasma, diffuse large B-cell lymphoma, B-cell high-grade lymphoma, liquid biopsy.

Received: October 5, 2022

Accepted: March 3, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Swerdlow SH, Steven H, Campo E, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Revised 4th edition. Lyon: IARC Press; 2017.
  2. Pfreundschuh M, Kuhnt E, Trumper L, et al. CHOP-like chemotherapy with or without rituximab in young patients with good-prognosis diffuse large-B-cell lymphoma: 6-year results of an open-label randomised study of the MabThera International Trial (MInT) Group. Lancet Oncol. 2011;12(11):1013–22. doi: 10.1016/S1470-2045(11)70235-2.
  3. Reddy A, Zhang J, Davis NS, et al. S. Genetic and Functional Drivers of Diffuse Large B Cell Lymphoma. Cell. 2017;171(2):481–494.e15. doi: 10.1016/j.cell.2017.09.027.
  4. Chapuy B, Stewart C, Dunford AJ, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and Nat Med. 2018;24(5):679–90. doi: 10.1038/s41591-018-0016-8.
  5. Schmitz R, Wright GW, Huang DW, et al. Genetics and pathogenesis of diffuse large B-cell lymphom N Engl J Med. 2018;378(15):1396–407. doi: 10.1056/NEJMoa1801445.
  6. Eichenauer DA, Aleman BMP, Andre M, et al. Hodgkin lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29(Suppl 4):iv19– doi: 10.1093/annonc/mdy080.
  7. Cwynarski K, Marzolini MAV, Barrington SF, et al. The management of primary mediastinal B-cell lymphoma: a British Society for Haematology Good Practice Paper. Br J Haematol. 2019;185(3):402– doi: 10.1111/bjh.15731.
  8. Brink I, Reinhardt MJ, Hoegerle S, et al. Increased metabolic activity in the thymus gland studied with 18F-FDG PET: age dependency and frequency after chemotherapy. J Nucl Med. 2001;42(4):591–5.
  9. Cohen JB, Behera M, Thompson CA, Flowers CR. Evaluating surveillance imaging for diffuse large B-cell lymphoma and Hodgkin lymphoma. Blood. 2017;129(5):561– doi: 10.1182/blood-2016-08-685073.
  10. Chien SH, Liu CJ, Hu YW, et al. Frequency of surveillance computed tomography in non-Hodgkin lymphoma and the risk of secondary primary malignancies: A nationwide population-based study. Int J Cancer. 2015;137(3):658–65. doi: 10.1002/ijc.29433.
  11. Thompson CA, Charlson ME, Schenkein E, et al. Surveillance CT scans are a source of anxiety and fear of recurrence in long-term lymphoma survivors. Ann Oncol. 2010;21(11):2262– doi: 10.1093/annonc/mdq215.
  12. Roschewski M, Dunleavy K, Pittaluga S, et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16(5):541–9. doi: 10.1016/S1470-2045(15)70106-3.
  13. Kurtz DM, Scherer F, Jin MC, et al. Circulating Tumor DNA Measurements As Early Outcome Predictors in Diffuse Large B-Cell Lymphoma. J Clin Oncol. 2018;36(28):2845– doi: 10.1200/JCO.2018.78.5246.
  14. Гаврилина О.А., Звонков Е.Е., Судариков А.Б. и др. Детекция В-клеточной клональности в костном мозге при диффузной В-крупноклеточной лимфоме. Гематология и трансфузиология. 2015;60(2):26–31.
    [Gavrilina OA, Zvonkov EE, Sudarikov AB, et al. Detection of bone marrow B-cell clonality in diffuse large B-cell lymphoma. Gematologiya i transfuziologiya. 2015;60(2):26–31. (In Russ)]
  15. Hohaus S, Giachelia M, Massini G, et al. Cell-free circulating DNA in Hodgkin’s and non-Hodgkin’s lymphomas. Ann Oncol. 2009;20(8):1408–13. doi: 10.1093/annonc/mdp006.
  16. Kristensen LS, Hansen JW, Kristensen SS, et al. Aberrant methylation of cell-free circulating DNA in plasma predicts poor outcome in diffuse large B cell lymphoma. Clin Epigenet. 2016;8(1):95. doi: 10.1186/s13148-016-0261-y.
  17. Li M, Jia Y, Xu J, et al. Assessment of the circulating cell-free DNA marker association with diagnosis and prognostic prediction in patients with lymphoma: a single-center experience. Ann Hematol. 2017;96(8):1343– doi: 10.1007/s00277-017-3043-5.
  18. Li M, Xu C. Circulating Cell-free DNA Utility for the Surveillance of Patients with Treated Diffuse Large B-cell Lymphoma. Clin Oncol (R Coll Radiol). 2017;29(9):637– doi: 10.1016/j.clon.2017.03.008.
  19. Armand P, Oki Y, Neuberg DS, et al. Detection of circulating tumour DNA in patients with aggressive B-cell non-Hodgkin lymphoma. Br J Haematol. 2013;163(1):123–6. doi: 10.1111/bjh.12439.
  20. Bo J, Sun L, Wang W, et al. Novel diagnostic biomarker for patients with Non-Hodgkin’s Lymphoma by IgH gene rearrangement. J Cancer Res Ther. 2016;12(2):903–8. doi: 10.4103/0973-1482.157345.
  21. He J, Wu J, Jiao Y, et al. IgH gene rearrangements as plasma biomarkers in Non-Hodgkin’s lymphoma patients. Oncotarget. 2011;2(3):178–85. doi: 10.18632/oncotarget.235.
  22. Herrera AF, Kim HT, Kong KA, et al. Next-generation sequencing-based detection of circulating tumour DNA After allogeneic stem cell transplantation for lymphoma. Br J Haematol. 2016;175(5):841– doi: 10.1111/bjh.14311.
  23. Hossain NM, Dahiya S, Le R, et al. Circulating tumor DNA assessment in patients with diffuse large B-cell lymphoma following CAR T-cell therapy. Leuk Lymphoma. 2019;60(2):503– doi: 10.1080/10428194.2018.1474463.
  24. Kurtz DM, Green MR, Bratman SV, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679–87. doi: 10.1182/blood-2015-03-635169.
  25. Scherer F, Kurtz DM, Newman AM, et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8(364):364ra155. doi: 10.1126/scitranslmed.aai8545.
  26. Zhong L, Huang WF. Better detection of Ig heavy chain and TCRγ gene rearrangement in plasma cell-free DNA from patients with non-Hodgkin Lymphoma. Neoplasma. 2010;57(6):507–11. doi: 10.4149/neo_2010_06_507.
  27. Alcaide M, Yu S, Bushell K, et al. Multiplex Droplet Digital PCR Quantification of Recurrent Somatic Mutations in Diffuse Large B-Cell and Follicular Lymphoma. Clin Chem. 2016;62(9):1238–47. doi: 10.1373/clinchem.2016.255315.
  28. Assouline SE, Nielsen TH, Yu S, et al. Phase 2 study of panobinostat with or without rituximab in relapsed diffuse large B-cell lymphoma. Blood. 2016;128(2):185–94. doi: 10.1182/blood-2016-02-699520.
  29. Bohers E, Viailly PJ, Becker S, et al. Non-invasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort. Blood Cancer J. 2018;8(8):74. doi: 10.1038/s41408-018-0111-6.
  30. Bohers E, Viailly PJ, Dubois S, et al. Somatic mutations of cell-free circulating DNA detected by next-generation sequencing reflect the genetic changes in both germinal center B-cell-like and activated B-cell-like diffuse large B-cell lymphomas at the time of diagnosis. Haematologica. 2015;100(7):e280–е28 doi: 10.3324/haematol.2015.123612.
  31. Camus V, Sarafan-Vasseur N, Bohers E, et al. Digital PCR for quantification of recurrent and potentially actionable somatic mutations in circulating free DNA from patients with diffuse large B-cell lymphoma. Leuk Lymphoma. 2016;57(9):2171–9. doi: 10.3109/10428194.2016.1139703.
  32. Rossi D, Diop F, Spaccarotella E, et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood. 2017;129(14):1947– doi: 10.1182/blood-2016-05-719641.
  33. Bessi L, Viailly PJ, Bohers E, et al. Somatic mutations of cell-free circulating DNA detected by targeted next-generation sequencing and digital droplet PCR in classical Hodgkin lymphoma. Leuk Lymphoma. 2019;60(2):498–502. doi: 10.1080/10428194.2018.1492123.
  34. Kurtz DM. Prognostication with circulating tumor DNA: is it ready for prime time? Hematology Am Soc Hematol Educ Program. 2019;2019(1):47–52. doi: 10.1182/hematology.2019000013.
  35. Rossi D, Kurtz DM, Roschewski M, et al. The development of liquid biopsy for research and clinical practice in lymphomas: Report of the 15-ICML workshop on ctDNA. Hematol Oncol. 2020;38(1):34– doi: 10.1002/hon.2704.
  36. Sriram D, Lakhotia R, Fenske TS. Measurement of circulating tumor DNA to guide management of patients with lymphoma. Clin Adv Hematol Oncol. 2019;17(9):509–
  37. Wu FT, Lu L, Xu W, Li JY. Circulating tumor DNA: clinical roles in diffuse large B cell lymphoma. Ann Hematol. 2019;98(2):255– doi: 10.1007/s00277-018-3529-9.
  38. Arzuaga-Mendez J, Prieto-Fernandez E, Lopez-Lopez E, et al. Cell-free DNA as a biomarker in diffuse large B-cell lymphoma: A systematic review. Crit Rev Oncol Hematol. 2019;139:7–15. doi: 10.1016/j.critrevonc.2019.04.013.
  39. Deeren D, Van Der Linden M, Dedeurwaerdere F, et al. Circulating cell-free DNA for response evaluation of intravascular lymphoma. Ann Hematol. 2019;98(8):2021– doi: 10.1007/s00277-019-03677-z.
  40. Mandel P, Meyais P. Les acides nucleiques du plasma sanguin chez l’homme [Nuclear Acids In Human Blood Plasma]. C R Seances Soc Biol Fil. 1948;142(3–4):241–3.
  41. Stroun M, Anker P, Maurice P, et al. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology. 1989;46(5):318–22. doi: 10.1159/000226740.
  42. Siravegna G, Mussolin B, Venesio T, et al. How liquid biopsies can change clinical practice in oncology. Ann Oncol. 2019;30(10):1580– doi: 10.1093/annonc/mdz227.
  43. Maco M, Kupcova K, Herman V, et al. Circulating tumor DNA in Hodgkin lymphoma. Ann Hematol. 2022;101(11):2393–403. doi: 10.1007/s00277-022-04949-x.
  44. Van Dongen JJ, Langerak AW, Bruggemann M, et al. Design and standardization of PCR primers and protocols for detection of clonal immunoglobulin and T-cell receptor gene recombinations in suspect lymphoproliferations: report of the BIOMED-2 Concerted Action BMH4-CT98-3936. Leukemia. 2003;17(12):2257–317. doi: 10.1038/sj.leu.2403202.
  45. Langerak AW, Groenen PJ, van Krieken JHJm, van Dongen J Immunoglobulin/T-cell receptor clonality diagnostics. Expert Opin Med Diagn. 2007;1(4):451–61. doi: 10.1517/17530059.1.4.451.

Long-Term Results of Classical Hodgkin’s Lymphoma Treatment in Real-World Clinical Practice: Experience of Novosibirsk Hematological Unit

MS Voitko1,2, TI Pospelova1,2, IN Nechunaeva2, YaYu Shebunyaeva1

1 Novosibirsk State Medical University, 52 Krasnyi pr-t, Novosibirsk, Russian Federation 630091

2 Municipal Clinical Hospital No. 2, 21 Polzunova ul., Novosibirsk, Russian Federation 630051

For correspondence: Mariya Sergeevna Voitko, MD, PhD, 21 Polzunova ul., Novosibirsk, Russian Federation, 630051; e-mail: voytko.marie@yandex.ru

For citation: Voitko MS, Pospelova TI, Nechunaeva IN, Shebunyaeva YaYu. Long-Term Results of Classical Hodgkin’s Lymphoma Treatment in Real-World Clinical Practice: Experience of Novosibirsk Hematological Unit. Clinical oncohematology. 2023;16(2):192–9. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-192-199


ABSTRACT

Aim. To assess the long-term results of classical Hodgkin’s lymphoma (cHL) treatment in Novosibirsk in real-world clinical practice.

Materials & Methods. The study enrolled 408 cHL patients treated and followed-up at the hematological unit of the Novosibirsk Municipal Clinical Hospital No. 2 from January 2008 to December 2021. The median age of patients was 33 years (range 26–44 years). Among them 223 (54.7 %) female and 185 (45.3 %) male patients. There were more patients with cHL stages III (n = 103; 25.2 %) and IV (n = 120; 29.4 %) than with stage II, which was identified in 185 (45.4 %) patients. ABVD regimen was administered to 132 (32.3 %) patients, 47 (11.5 %) patients received ABVD escalated to BEACOPP. BEACOPP therapy was performed in 229 (56.2 %) patients. Subsequent radiotherapy was assigned to 202 (49.5 %) patients. Second-line therapy was required by 89 (21.8 %) patients with relapsed and resistant cHL.

Results. The 10-year overall survival (OS) was 81 %, and the 5-year OS was 91 %. Similar progression-free survival (PFS) rates were 86 % and 77 %, respectively. The 10-year PFS in patients with stage II was 87 %, while in patients with stages II (mediastinal bulky mass), III and IV, it was only 69 % (= 0.002). The 10-year OS in patients with localized stages was 91 %, and in patients with generalized stages it was 79 % (= 0.0006). The 10-year OS in patients less than 45 years of age was 88 %, and in patients more than 45 years of age it was 69 %. The 10-year PFS in patients less than 45 years of age was 84 %, and in the older age group it was 60 % (= 0.001).

Conclusion. The study results demonstrate high rates of long-term survival of cHL patients and are well comparable with the data of other study groups. Nevertheless, scientific research should be continued to develop optimal risk-adapted programs of cHL chemotherapy and to define further prospects for improving the treatment outcomes of this malignant tumor.

Keywords: classical Hodgkin’s lymphoma, overall survival, progression-free survival, immediate and long-term treatment results.

Received: October 28, 2022

Accepted: March 2, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Демина Е.А. Руководство по лечению лимфомы Ходжкина. М.: ГРУППА РЕМЕДИУМ, 2021.
    [Demina EA. Rukovodstvo po lecheniyu limfomy Khodzhkina. (Guidelines for Hodgkin’s lymphoma treatment.) Moscow: GRUPPA REMEDIUM Publ.; 2021. (In Russ)]
  2. Shanbhag S, Ambinder RF. Hodgkin lymphoma: A review and update on recent progress. CA Cancer J Clin. 2018;68(2):116–32. doi: 10.3322/caac.21438.
  3. Kaseb H, Babiker H. Hodgkin Lymphoma. Treasure Island (FL): StatPearls Publishing LLC; 2020.
  4. Salati M, Cesaretti M, Macchia M, et al. Epidemiological overview of Hodgkin lymphoma across the Mediterranean basin. Mediterr J Hematol Infect Dis. 2014;6(1):e2014048. doi: 10.4084/MJHID.2014.048.
  5. Jetley S, Khetrapal S, Pujani M, et al. Unusual histology in Hodgkin’s Lymphoma: report of an interesting case. Indian J Surg Oncol. 2017;8(2):181–4. doi: 10.1007/s13193-016-0602-5.
  6. MacLennan KA, Bennett MH, Tu A, et al. Relationship of histopathologic features to survival and relapse in nodular sclerosing Hodgkin’s disease. A study of 1659 patients. Cancer. 1989;64(8):1686–93. doi: 10.1002/1097-0142(19891015)64:8<1686::aid-cncr2820640822>3.0.co;2-i.
  7. Szychot E, Shankar A, Haider А, Ramsay A. Variant histology nodular lymphocyte predominant Hodgkin lymphoma – a route to transformation? Br J Haematol. 2018;181(3):403–6. doi: 10.1111/bjh.14607.
  8. Dominguez RA, Alfaro LJ, de la Cruz Merino L, et al. SEOM clinical guidelines for the treatment of Hodgkin’s lymphoma. Clin Transl Oncol. 2015;17(12):1005–13. doi: 10.1007/s12094-015-1429-1.
  9. Российские клинические рекомендации по диагностике и лечению лимфопролиферативных заболеваний. Под ред. И.В. Поддубной, В.Г. Савченко. М.: Буки Веди, 2018. 324 с.
    [Poddubnaya IV, Savchenko VG, eds. Rossiiskie klinicheskie rekomendatsii po diagnostike i lecheniyu limfoproliferativnykh zabolevanii. (Russian clinical guidelines on diagnosis and treatment of lymphoproliferative disorders.) Moscow: Buki Vedi Publ.; 2018. 324 р. (In Russ)]
  10. Brice P, de Kerviler E, Friedberg JW. Classical Hodgkin lymphoma. Lancet. 2021;398(10310):1518–27. doi: 10.1016/S0140-6736(20)32207-8.
  11. Shamoon RP, Ali MD, Shabila NP. Overview and outcome of Hodgkin’s Lymphoma: Experience of a single developing country’s oncology centre. PLoS One. 2018;13(4):e0195629. doi: 10.1371/journal.pone.0195629.
  12. Edgren G, Liang L, Adami HO, Chang ET. Enigmatic sex disparities in cancer incidence. Eur J Epidemiol 2012;27(3):187–96. doi: 10.1007/s10654-011-9647-5.
  13. Никитин Е.А., Шаркунов Н.Н., Маркарян В.Г. и др. Диагностика и лечение лимфомы Ходжкина в практическом здравоохранении: анализ госпитального регистра городской клинической больницы им. С.П. Боткина. Онкогематология. 2016;11(3):8–19. doi: 10.17650/1818-8346-2016-11-3-8-19.
    [Nikitin ЕА, Sharkunov NN, Markaryan VG, et al. Treatment patterns, outcomes and long-term toxicity among patients with Hodgkin’s lymphoma in real world: results of a hospital based registry. Oncohematology. 2016;11(3):8–19. doi: 10.17650/1818-8346-2016-11-3-8-19. (In Russ)]
  14. Капланов К.Д., Волков Н.П., Клиточенко Т.Ю. и др. Результаты анализа регионального регистра пациентов с диффузной В-крупноклеточной лимфомой: факторы риска и проблемы иммунохимиотерапии. Клиническая онкогематология. 2019;12(2):154–64. doi: 10.21320/2500-2139-2019-12-2-154-164.
    [Kaplanov KD, Volkov NP, Klitochenko TYu, et al. Analysis Results of the Regional Registry of Patients with Diffuse Large B-cell Lymphoma: Risk Factors and Chemo-Immunotherapy Issues. Clinical oncohematology. 2019;12(2):154–64. doi: 10.21320/2500-2139-2019-12-2-154-164. (In Russ)]
  15. Мочкин Н.Е., Саржевский В.О., Дубинина Ю.Н. и др. Результаты лечения классической лимфомы Ходжкина, включающего высокодозную химиотерапию с трансплантацией аутологичных гемопоэтических стволовых клеток, в НМХЦ им. Н.И. Пирогова. Клиническая онкогематология. 2018;11(3):234–40. doi: 10.21320/2500-2139-2018-11-3-234-240.
    [Mochkin NE, Sarzhevskii VO, Dubinina YuN, et. al. Outcome of Classical Hodgkin’s Lymphoma Treatment Based on High-Dose Chemotherapy and Autologous Hematopoietic Stem Cell Transplantation: The Experience in the NI Pirogov Russian National Medical Center of Surgery. Clinical oncohematology. 2018;11(3):234–40. doi: 10.21320/2500-2139-2018-11-3-234-240. (In Russ)]
  16. Von Treschkow B, Kreissl S, Goergen H, et al. Intensive treatment strategies in advanced stage Hodgkin’s lymphoma (HD9 and HD12): analysis of long-term survival in two randomised trials. Lancet Haematol. 2018;5(10):e462–e473. doi: 10.1016/S2352-3026(18)30140-6.

Dynamics of SARS-CoV-2 RNA Detection in Patients and Employees of the National Research Center for Hematology During the First Two Years of the Novel COVID-19 Pandemic

OG Starkova, DS Tikhomirov, AYu Krylova, IO Snezhko, EN Ovchinnikova, OA Aleshina, TA Tupoleva, TV Gaponova

National Research Center for Hematology, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Oksana Gazimagomedovna Starkova, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(966)189-57-12; e-mail: oksanastar2006@rambler.ru

For citation: Starkova OG, Tikhomirov DS, Krylova AYu, et al. Dynamics of SARS-CoV-2 RNA Detection in Patients and Employees of the National Research Center for Hematology During the First Two Years of the Novel COVID-19 Pandemic. Clinical oncohematology. 2023;16(2):186–91. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-186-191


ABSTRACT

Background. COVID-19 required fundamental changes in healthcare management, also in medical care for oncological and hematological patients. Visits to healthcare organizations were minimized, 75 % of doctor appointments were converted to telemedicine consultations. The solutions aimed at preventing further spread of COVID-19 included establishing of observational units, distinguishing between patient and employee flows, regular SARS-CoV-2 RNA testing, reducing hospital stays and transferring patients with positive COVID-19 tests to the remodeled hospitals specializing in the novel coronavirus infection, as well as providing only emergency medical treatment and, as far as feasible, converting systemic chemotherapy to per os treatment, etc.

Aim. To assess SARS-CoV-2 RNA detection dynamics at the National Research Center for Hematology from April 2020 to January 2022 during the implementation of epidemic control measures.

Materials & Methods. The study was based on SARS-CoV-2 RNA testing of naso- and oropharyngeal samples obtained from patients and employees of the National Research Center for Hematology (hereafter referred to as Center). Besides, bronchoalveolar lavage fluid, lung tissue biopsies, and sputum were examined for SARS-CoV-2 RNA. The study was performed at the Center’s Virusology Department with the use of Sintol reagent kit “ПЦР-РВ-2019-nCov”.

Results. The study was based on 107,470 tests: 58,141 (54 %) of employees and 45,126 (46 %) of patients; 35,508 (33 %) of men and 71,962 (67 %) of women. In 1318 cases SARS-CoV-2 RNA was detected which accounted for 1.15 % of total test number. In the groups of employees/patients, virus detection rate was 1.42 %/1.09 % (< 0.001), and in male/female groups it was 1.3 %/1.2 %, respectively (= 0.154). The rate of infection in the groups of tumor and non-tumor hematological patients, as proved by SARS-CoV-2 RNA testing, was 1.24 % and 0.92 %, respectively (= 0.147). In employees and patients of the Center, a wave-like virus detection rate was observed. The largest number of infections was registered in April-June 2020 (79 patients and 170 employees), October-December 2020 (126 patients and 190 employees), and January 2022 (59 patients and 203 employees), which corresponded to the first, second, and fifth COVID-19 waves in Russia.

Conclusion. The analysis of data obtained at the National Research Center for Hematology demonstrated a wave-like SARS-CoV-2 RNA detection rate in employees and patients of the Center, which corresponded to the general trend in Russia. The SARS-CoV-2 RNA detection rate did not depend on sex of subjects under study and was not significantly different in the groups of tumor and non-tumor hematological patients. Although the patients in hematological hospital are more exposed to the risk of severe infectious complications, they showed laboratory markers for COVID-19 less frequently than the Center employees.

Keywords: COVID-19, SARS-CoV-2 RNA, novel coronavirus infection, hematological patients, tumor and non-tumor hematological diseases.

Received: September 5, 2022

Accepted: March 7, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Yang P, Wang X. COVID-19: a new challenge for human beings. Cell Mol Immunol. 2020;17(5):555–7. doi: 10.1038/s41423-020-0407-x.
  2. Bhagat S, Yadav N, Shah J, et al. Novel corona virus (COVID-19) pandemic: current status and possible strategies for detection and treatment of the disease. Expert Rev Anti Infect Ther. 2022;20(10):1275–98. doi: 10.1080/14787210.2021.1835469.
  3. Львов Д.К. Альховский С.В. Истоки пандемии COVID-19: экология и генетика коронавирусов (Betacoronavirus: Coronaviridae) SARS-COV, SARS-COV-2 (подрод Sarbecovirus), MERS-COV (подрод Merbecovirus). Вопросы вирусологии. 2020;65(2):62–70. doi: 10.36233/0507-4088-2020-65-2-62-70.
    [Lvov DK, Alkhovsky SV. Source of the COVID-19 pandemic: ecology and genetics of coronaviruses (Betacoronavirus: Coronaviridae) SARS-CoV, SARS-CoV-2 (subgenus Sarbecovirus), and MERS-CoV (subgenus Merbecovirus). Problems of Virology. 2020;65(2):62–70. doi: 10.36233/0507-4088-2020-65-2-62-70. (In Russ)]
  4. Drosten C, Gunther S, Preiser W, et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348(20):1967–76. doi: 10.1056/NEJMoa030747/
  5. Львов Д.К., Альховский С.В., Колобухина Л.В., Бурцева Е.И. Этиология эпидемической вспышки COVID-19 в г. Ухань (провинция Хубэй, Китайская Народная Республика), ассоциированной с вирусом 2019-nCoV (Nidovirales, Coronaviridae, Coronavirinae, Betacoronavirus, подрод Sarbecovirus): уроки эпидемии SARS-CoV. Вопросы вирусологии. 2020;65(1):6–16. doi: 10.36233/0507-4088-2020-65-1-6-15.
    [Lvov DK, Alkhovsky SV, Kolobukhina LV, Burtseva EI. Etiology of epidemic outbreaks COVID-19 in Wuhan, Hubei province, Chinese People Republic associated with 2019-nCoV (Nidovirales, Coronaviridae, Coronavirinae, Betacoronavirus, Subgenus Sarbecovirus): lessons of SARS-CoV outbreak. Problems of Virology. 2020;65(1):6–16. doi: 10.36233/0507-4088-2020-65-1-6-15. (In Russ)]
  6. Гарафутдинов Р.Р., Мавзютов А.Р., Никоноров Ю.М. и др. Бетакоронавирус SARS-CoV-2, его геном, разнообразие генотипов и молекулярно-биологические меры борьбы с ним. Биомика. 2020;12(2):242–71. doi: 10.31301/2221-6197.bmcs.2020-15.
    [Garafutdinov RR, Mavzyutov AR, Nikonorov YuM, et al. Betacoronavirus SARS-CoV-2, its genome, variety of genotypes and molecular-biological approaches to combat it. Biomics. 2020;12(2):242–71. doi: 10.31301/2221-6197.bmcs.2020-15. (In Russ)]
  7. Isidori A, de Level L, Gergis U, et al. Management of patients with hematologic malignancies during the COVID-19 pandemic: Practical considerations and lessons to be learned. Front Oncol. 2020;10:1439. doi: 10.3389/fonc.2020.01439.
  8. Salako O, Okunade K, Allsop M, et al. Upheaval in cancer care during the COVID-19 outbreak. Ecancermedicalscience. 2020;14:ed97. doi: 10.3332/ecancer.2020.ed9.
  9. Gupta M, Ahuja R, Gupta S, et al. Running of high patient volume radiation oncology department during COVID-19 crisis in India: Our institutional strategy. Radiat Oncol J. 2020;38(2):93–8. doi: 10.3857/roj.2020.0019
  10. Dalu D, Rota S, Cona MS, et al. A proposal of a “ready to use” COVID-19 control strategy in an Oncology ward: Utopia or reality? Crit Rev Oncol Hematol. 2021;157:103168. doi: 10.1016/j.critrevonc.2020.103168.
  11. Leung MST, Lin SG, Chow J, Harky A. COVID-19 and Oncology: Service transformation during pandemic. Cancer Med. 2020;9(19):7161–71. doi: 10.1002/cam4.3384.
  12. He Y, Lin Z, Tang D, et al. Strategic plan for management of COVID-19 in paediatric haematology and oncology departments. Lancet Haematol. 2020;7(5):e359–е doi: 10.1016/S2352-3026(20)30104-6.
  13. Гаврилина О.А., Васильева А.Н., Галстян Г.М. и др. Опыт работы обсервационного отделения для больных с патологией системы крови во время пандемии COVID-19. Гематология и трансфузиология. 2021;66(1):8–19. doi: 10.35754/0234-5730-2021-66-1-8-19.
    [Gavrilina OA, Vasileva AN, Galstyan GM, et al. Experience of haematological observatory ward during COVID-10 pandemic. Russian journal of hematology and transfusiology. 2021;66(1):8–19. doi: 10.35754/0234-5730-2021-66-1-8-19. (In Russ)]
  14. Годков М.А., Шустов В.В., Кашолкина Е.А. Динамика и гендерно-возрастные особенности эпидемического процесса COVID-19 в городе Москве (итоги скринингового обследования за 1,5 года). Лабораторная служба. 2021;10(4):30–7. doi: 10.17116/labs20211004130.
    [Godkov MA, Shustov VV, Kasholkina EA. Dynamics and gender and age features of the COVID-19 EPIDEMIC process in Moscow (results of screening survey for 1.5 years). Laboratornaya sluzhba. 2021;10(4):30–7. doi: 10.17116/labs20211004130. (In Russ)]
  15. Официальная статистика COVID-19 в России. [Интернет] Доступно по: https://coronavirusstat.ru/?ysclid=lc6d Ссылка активна на 27.12.2022. [Official COVID-19 morbidity statistics in Russia. (Internet) Available from: https://coronavirusstat.ru/?ysclid=lc6dwvg036948485677. Accessed 27.12.2022. (In Russ)]
  16. Liang W, Guan W, Chen R, et al. Cancer patients in SARS-CoV-2 infection: A nationwide analysis in China. Lancet Oncol. 2020;21(3):335–7. doi: 10.1016/S1470-2045(20)30096-6

The Prognostic Value of Somatic Mutations of Epigenetic Regulation Genes in Acute Myeloid Leukemias in Real-World Clinical Practice: Results of an Observational Non-Interventional Prospective Interregional Study

AA Shatilova1, IG Budaeva1, AV Petukhov1, SA Silonov1, AE Ershova1, TS Nikulina1, YuD Matvienko1, YuV Mirolyubova1, KV Bogdanov1, LV Anchukova2, YuS Neredko3, SYu Tyasko3, OE Ochirova4, AG Karpova4, ER Vasil’eva5, OD Serdyuk6, DA Yaskulskii6, DV Bukin7, YuA Alekseeva1, EG Lomaia1, LL Girshova1

1 VA Almazov National Medical Research Center, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341

2 Vologda Regional Clinical Hospital, 17 Lechebnaya ul., Vologda, Russian Federation, 160002

3 Stavropol Krai Clinical Oncology Dispensary, 182a Oktyabrskaya ul., Stavropol, Russian Federation, 355001

4 NA Semashko Republican Clinical Hospital, 12 Pavlova ul., Ulan-Ude, Russian Federation, 670031

5 Perm Krai Clinical Hospital, 85 Pushkina ul., Perm, Russian Federation, 614990

6 Clinical Oncology Dispensary No. 1, 146 Dimitrova ul., Krasnodar, Russian Federation, 350040

7 Regional Clinical Hospital, 105 Peterburgskoe sh., Tver, Russian Federation, 170036

For correspondence: Aleksina Alekseevna Shatilova, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341; Tel.: +7(911)476-35-58; e-mail: alexina-96@list.ru

For citation: Shatilova AA, Budaeva IG, Petukhov AV, et al. The Prognostic Value of Somatic Mutations of Epigenetic Regulation Genes in Acute Myeloid Leukemias in Real-World Clinical Practice: Results of an Observational Non-Interventional Prospective Interregional Study. Clinical oncohematology. 2023;16(2):174–85. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-174-185


ABSTRACT

Aim. To assess the rate of DNMT3A, IDH1, IDH2, and ASXL1 gene mutations and their effect on the prognosis both as isolated findings and in combination with well-known chromosomal aberrations and gene mutations in newly diagnosed acute myeloid leukemia (AML) patients from some regions of the Russian Federation.

Materials & Methods. The study enrolled 83 patients with newly diagnosed AML from 22 regions of the Russian Federation, who underwent molecular genetic examination for detecting IDH1 (R132), IDH2 (R140), ASXL1, and DNMT3A gene mutations with droplet digital PCR and Sanger sequencing methods.

Results. The mutation rate in DNMT3A was 16.7 %, in IDH1 (R132) it was 6 %, in IDH2 (R140) it was 9.6 %, and in ASXL1 it was 6 %. The R140 mutation in IDH2 correlated with the older age of patients. The mutations in IDH1 (R132), IDH2 (R140), and DNMT3A showed a significant association with mutated NPM1. The mutations in IDH1 (R132), IDH2 (R140) were reported to occur significantly more often in patients with normal karyotype. The IDH1 (R132) and IDH2 (R140) mutations appeared to have a favorable effect on AML prognosis, which is most likely to be associated with a high rate of their compatibility with NPM1 mutation. The mutated type of DNMT3A had a negative effect on overall survival of patients with NPM1 mutation. The mutation in ASXL1 also appeared to be an unfavorable prognostic factor for overall survival of patients with wild type NPM1.

Conclusion. A high rate of mutation occurrence in epigenetic regulation genes as well as the prognostic potential of these mutations in AML necessitate the need for determining the mutation status of DNMT3A, IDH1, IDH2, and ASXL1 in the context of primary diagnosis in real-world clinical practice.

Keywords: acute myeloid leukemias, IDH, DNMT3A, ASXL1, epigenetic regulation genes.

Received: September 25, 2022

Accepted: March 1, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. De Kouchkovsky I, Abdul-Hay M. Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J. 2016;6(7):e441. doi: 10.1038/bcj.2016.50.
  2. Cancer Genome Atlas Research Network; Ley TJ, Miller C, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74. doi: 10.1056/NEJMoa1301689.
  3. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424–47. doi: 10.1182/blood-2016-08-733196.
  4. Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood. 2002;100(13):4325–36. doi: 10.1182/blood-2002-03-0772.
  5. Острые миелоидные лейкозы. Клинические рекомендации. Под ред. А.Д. Каприна. М.: Ассоциация онкологов России, 2020.
    [AD Kaprin, ed. Ostrye mieloidnye leikozy. Klinicheskie rekomendatsii. (Acute myeloid leukemias. Clinical guidelines.) Moscow: Assotsiatsiya Onkologov Rossii Publ.; 2020. (In Russ)]
  6. Takahashi S. Current findings for recurring mutations in acute myeloid leukemia. J Hematol Oncol. 2011;4:36. doi: 10.1186/1756-8722-4-36.
  7. Kishtagari A, Levine RL. The Role of Somatic Mutations in Acute Myeloid Leukemia Pathogenesis. Cold Spring Harb Perspect Med. 2021;11(4):a034975. doi: 10.1101/cshperspect.a034975.
  8. Jiang Y, Dunbar A, Gondek LP, et al. Aberrant DNA methylation is a dominant mechanism in MDS progression to AML. Blood. 2009;113(6):1315–25. doi: 10.1182/blood-2008-06-163246.
  9. Зайкова Е.К., Белоцерковская Е.В., Зайцев Д.В. и др. Молекулярная диагностика мутаций гена FLT3 у пациентов с острыми миелоидными лейкозами. Клиническая онкогематология. 2020;13(2):150–60. doi: 10.21320/2500-2139-2020-13-2-150-160.
    [Zaikova EK, Belotserkovskaya EV, Zaytsev DV, et al. Molecular Diagnosis of FLT3 Mutations in Acute Myeloid Leukemia Patients. Clinical oncohematology. 2020;13(2):150–60. doi: 10.21320/2500-2139-2020-13-2-150-160. (In Russ)]
  10. Whitehall VL, Dumenil TD, McKeone DM, et al. Isocitrate dehydrogenase 1 R132C mutation occurs exclusively in microsatellite stable colorectal cancers with the CpG island methylator phenotype. Epigenetics. 2014;9(11):1454–60. doi: 10.4161/15592294.2014.971624.
  11. Berenstein R, Blau IW, Kar A, et al. Comparative examination of various PCR-based methods for DNMT3A and IDH1/2 mutations identification in acute myeloid leukemia. J Exp Clin Cancer Res. 2014;33(1):44. doi: 10.1186/1756-9966-33-4.
  12. Gelsi-Boyer V, Trouplin V, Adelaide J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800. doi: 10.1111/j.1365-2141.2009.07697.x.
  13. Shivarov V, Ivanova M, Naumova E. Rapid detection of DNMT3A R882 mutations in hematologic malignancies using a novel bead-based suspension assay with BNA(NC) probes. PLoS One. 2014;9(6):e99769. doi: 10.1371/journal.pone.0099769.
  14. Maiti A, Qiao W, Sasaki K, et al. Venetoclax with decitabine vs intensive chemotherapy in acute myeloid leukemia: A propensity score matched analysis stratified by risk of treatment-related mortality. Am J Hematol. 2021;96(3):282–91. doi: 10.1002/ajh.26061.
  15. Cherry EM, Abbott D, Amaya M, et al. Venetoclax and azacitidine compared with induction chemotherapy for newly diagnosed patients with acute myeloid leukemia. Blood Adv. 2021;5(24):5565–73. doi: 10.1182/bloodadvances.2021005538.
  16. Montalban-Bravo G, DiNardo CD. The role of IDH mutations in acute myeloid leukemia. Future Oncol. 2018;14(10):979–93. doi: 10.2217/fon-2017-0523.
  17. Kishtagari A, Levine RL. The Role of Somatic Mutations in Acute Myeloid Leukemia Pathogenesis. Cold Spring Harb Perspect Med. 2021;11(4):a034975. doi: 10.1101/cshperspect.a034975.
  18. DiNardo CD, Ravandi F, Agresta S, et al. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol. 2015;90(8):732–36. doi: 10.1002/ajh.24072.
  19. Zarnegar-Lumley S, Alonzo TA, Othus M, et al. Characteristics and prognostic effects of IDH mutations across the age spectrum in AML: a collaborative analysis from COG, SWOG, and ECOG. Blood. 2020;136(Suppl 1):31–2. doi: 10.1182/blood-2020-134211.
  20. Aref S, Kamel Areida el S, Abdel Aaal MF, et al. Prevalence and Clinical Effect of IDH1 and IDH2 Mutations Among Cytogenetically Normal Acute Myeloid Leukemia Patients. Clin Lymphoma Myeloma Leuk. 2015;15(9):550–5. doi: 10.1016/j.clml.2015.05.009.
  21. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28(14):2348–55. doi: 10.1200/JCO.2009.27.3730.
  22. Белоцерковская Е.В., Зайкова Е.К., Петухов А.В. и др. Выявление мутаций генов эпигенетической регуляции генома IDH1/2, DNMT3A, ASXL1 и их сочетания с мутациями FLT3, NPM1, RUNX1 у пациентов с острыми миелоидными лейкозами. Клиническая онкогематология. 2021;14(1):13–21. doi: 10.21320/2500-2139-2021-14-1-13-21.
    [Belotserkovskaya EV, Zaikova EK, Petukhov AV, et al. Identification of Mutations in IDH1/2, DNMT3A, ASXL1 Genes of Genome Epigenetic Regulation and Their Co-Occurrence with FLT3, NPM1, RUNX1 Mutations in Acute Myeloid Leukemia. Clinical oncohematology. 2021;14(1):13–21. doi: 10.21320/2500-2139-2021-14-1-13-21. (In Russ)]
  23. ElNahass YH, Badawy RH, ElRefaey FA, et al. IDH Mutations in AML Patients; A higher Association with Intermediate Risk Cytogenetics. Asian Pac J Cancer Prev. 2020;21(3):721–5. doi: 10.31557/APJCP.2020.21.3.721.
  24. Molenaar RJ, Thota S, Nagata Y, et al. Clinical and biological implications of ancestral and non-ancestral IDH1 and IDH2 mutations in myeloid neoplasms. Leukemia. 2015;29(11):2134–42. doi: 10.1038/leu.2015.91.
  25. Xu Q, Li Y, Lv N, et al. Correlation Between Isocitrate Dehydrogenase Gene Aberrations and Prognosis of Patients with Acute Myeloid Leukemia: A Systematic Review and Meta-Analysis. Clin Cancer Res. 2017;23(15):4511–22. doi: 10.1158/1078-0432.CCR-16-2628.
  26. Cai SF, Levine RL. Genetic and epigenetic determinants of AML pathogenesis. Semin Hematol. 2019;56(2):84–9. doi: 10.1053/j.seminhematol.2018.08.001.
  27. Park DJ, Kwon A, Cho BS, et al. Characteristics of DNMT3A mutations in acute myeloid leukemia. Blood Res. 2020;55(1):17–26. doi: 10.5045/br.2020.55.1.17.
  28. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363(25):2424–33. doi: 10.1056/NEJMoa1005143.
  29. Wang M, Yang C, Zhang L, Schaar DG. Molecular Mutations and Their Cooccurrences in Cytogenetically Normal Acute Myeloid Leukemia. Stem Cells Int. 2017;2017:6962379. doi: 10.1155/2017/6962379.
  30. Ostronoff F, Othus M, Ho PA, et al. Mutations in the DNMT3A exon 23 independently predict poor outcome in older patients with acute myeloid leukemia: a SWOG report. Leukemia. 2013;27(1):238–41. doi: 10.1038/leu.2012.168.
  31. Gaidzik VI, Schlenk RF, Paschka P, et al. Clinical impact of DNMT3A mutations in younger adult patients with acute myeloid leukemia: results of the AML Study Group (AMLSG). Blood. 2013;121(23):4769–77. doi: 10.1182/blood-2012-10-461624.
  32. Roller A, Grossmann V, Bacher U, et al. Landmark analysis of DNMT3A mutations in hematological malignancies. Leukemia. 2013;27(7):1573–8. doi: 10.1038/leu.2013.65.
  33. Kihara R, Nagata Y, Kiyoi H, et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia. 2014;28(8):1586–95. doi: 10.1038/leu.2014.55.
  34. Dunlap JB, Leonard J, Rosenberg M, et al. The combination of NPM1, DNMT3A, and IDH1/2 mutations leads to inferior overall survival in AML. Am J Hematol. 2019;94(8):913–20. doi: 10.1002/ajh.25517.
  35. Gelsi-Boyer V, Trouplin V, Adelaide J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145(6):788–800. doi: 10.1111/j.1365-2141.2009.07697.x.
  36. Asada S, Fujino T, Goyama S, Kitamura T. The role of ASXL1 in hematopoiesis and myeloid malignancies. Cell Mol Life Sci. 2019;76(13):2511–23. doi: 10.1007/s00018-019-03084-7.
  37. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med. 2016;374(23):2209–21. doi: 10.1056/NEJMoa1516192.
  38. Pratcorona M, Abbas S, Sanders MA, et al. Acquired mutations in ASXL1 in acute myeloid leukemia: prevalence and prognostic value. Haematologica. 2012;97(3):388–92. doi: 10.3324/haematol.2011.051532.
  39. Bohl SR, Bullinger L, Rucker FG. Epigenetic therapy: azacytidine and decitabine in acute myeloid leukemia. Expert Rev Hematol. 2018;11(5):361–71. doi: 10.1080/17474086.2018.1453802.
  40. NCCN Clinical Practice Guidelines in Oncology. Acute Myeloid Leukemia. Version 1.2022. (Internet) Available from: https://www.nccn.org/professionals/physician_gls/pdf/aml_blocks.pdf. (accessed 20.05.2022).
  41. Carter JL, Hege K, Yang J, et al. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther. 2020;5(1):288. doi: 10.1038/s41392-020-00361-x.

Efficacy and Toxicity of Induction Therapy in Patients with Newly Diagnosed Systemic AL Amyloidosis: Results of a Prospective Single-Center Clinical Study

IG Rekhtina, VA Khyshova, MV Solov’ev, LP Mendeleeva

National Research Center for Hematology, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Viktoriya Aleksandrovna Khyshova, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(983)413-02-00; e-mail: viktoria2102@icloud.com

For citation: Rekhtina IG, Khyshova VA, Solov’ev MV, Mendeleeva LP. Efficacy and Toxicity of Induction Therapy in Patients with Newly Diagnosed Systemic AL Amyloidosis: Results of a Prospective Single-Center Clinical Study. Clinical oncohematology. 2023;16(2):166–73. (In Russ).

DOI: 10.21320/2500-2139-2023-16-2-166-173


ABSTRACT

Aim. To assess the outcomes of induction therapy in patients with newly diagnosed systemic AL Amyloidosis (AL-А).

Materials & Methods. The prospective single-center clinical study enrolled 60 patients (32 women and 28 men) with newly diagnosed systemic AL-A stage I/IIIA. The median age was 59 years (range 34–74 years). In 57 patients, BorСyDex (bortezomib, cyclophosphamide, dexamethasone) was used as first-line therapy. RCd regimen (lenalidomide, cyclophosphamide, dexamethasone) was administered to 3 patients. Patients with the lack of efficacy or pronounced toxicity (n = 24) received second-line induction therapy with lenalidomide or melphalan combined with dexamethasone. High-dose chemotherapy with autologous hematopoietic stem cell transplantation (auto-HSCT) was administered to 11 (18 %) patients.

Results. Hematologic targeted response (complete remission [CR] and very good partial remission [VGPR]) to BorCyDex was achieved in 62 % of patients. As a result of all lines of induction therapy, including auto-HSCT, targeted response increased to 69 %, specifically in 7/51 (14 %) patients with stringent CR (sCR), 8/51 (16 %) patients with CR, and 20/51 (39 %) patients with VGPR. Renal response after BorCyDex was registered in 10/38 (26 %) patients, 6/31 (19 %) patients showed heart response, and in 4/5 (80 %) patients liver response was reported. All therapy lines with auto-HSCT led to organ response (in ≥ 1 organ) in 15/46 (32 %) patients. Clinical response was shown by all patients with achieved sCR, by 67 % of patients with CR, and 47 % with VGPR (= 0.04). With lower hematologic response rates, no clinical improvement was observed. With follow-up duration of 36 months, the median disease-free survival (without signs of hematologic and clinical progression) was not achieved. The 3-year overall survival was 80 %. Mortality during induction therapy was 10 % (6 patients died, including 2 patients with COVID-19). The planned 6 courses of BorCyDex could be completed only in 13 (23 %) out of 55 patients. During the induction therapy using BorCyDex, 4 patients died. The treatment was discontinued in 7/55 (12 %) patients due to its inefficacy and in 22/55 (39 %) patients because of severe peripheral and autonomic polyneuropathy. Nine (16 %) out of 55 patients with the achieved hematologic response showed excessive NT-proBNP elevation, which was accompanied by cardiovascular complications and provided ground for chemotherapy withdrawal.

Conclusion. Low organ recovery rate remains the most challenging issue for AL-A treatment. Hematologic response depth (achieved CR) is a critical factor in achieving clinical effect. The obtained data confirmed high toxicity of BorCyDex regimen in AL-A patients. Despite the advances in AL-А therapy which are associated with the use of proteasome inhibitors, treatment of this disease calls for new and more effective approaches.

Keywords: AL Amyloidosis, bortezomib, lenalidomide, efficacy, toxicity.

Received: October 31, 2022

Accepted: March 3, 2023

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Quock TP, Yan T, Chang E, et al. Epidemiology of AL amyloidosis: a real-world study using US claims data. Blood Adv. 2018;2(10):1046–53. doi: 10.1182/bloodadvances.2018016402.
  2. Hemminki K, Li Х, Forsti A, et al. Incidence and survival in non-hereditary amyloidosis in Sweden. BMC Public Health. 2012;12:974. doi: 10.1186/1471-2458-12-974.
  3. Sanchorawala V, Sun F, Quillen K, et al. Long-term outcome of patients with AL amyloidosis treated with high-dose melphalan and stem cell transplantation: 20-year experience. Blood. 2015;126(20):2345–7. doi: 10.1182/blood-2015-08-662726.
  4. Manwani R, Cohen O, Sharpley F, et al. A prospective observational study of 915 patients with systemic AL amyloidosis treated with upfront bortezomib. Blood. 2019;134(25):2271–80. doi: 10.1182/blood.2019000834.
  5. Gertz MA. How to manage primary amyloidosis. 2012;26(2):191–8. doi: 10.1038/leu.2011.219.
  6. Mikhael JR, Schuster SR, Jimenez-Zepeda VH, et al. Cyclophosphamide-bortezomib-dexamethasone (CyBorD) produces rapid and complete hematologic response in patients with AL amyloidosis. Blood. 2012;119(19):4391–4. doi: 10.1182/blood-2011-11-390930.
  7. Gertz MA. Immunoglobulin light chain amyloidosis: 2016 update on diagnosis, prognosis, and treatment. Am J Hematol. 2016;91(9):947–56. doi: 10.1002/ajh.24433.
  8. Palladini G, Sachchithanantham S, Milani P, et al. A European collaborative study of cyclophosphamide, bortezomib, and dexamethasone in upfront treatment of systemic AL amyloidosis. Blood. 2015;126(5):612–5. doi: 10.1182/blood-2015-01-620302.
  9. Shen KN, Feng J, Huang XF, et al. At least partial hematological response after first cycle of treatment predicts organ response and long-term survival for patients with AL amyloidosis receiving bortezomib-based treatment. Ann Hematol. 2017;96(12):2089–94. doi: 10.1007/s00277-017-3132-5.
  10. Dispenzieri A, Gertz MA, Kyle RA, et al. Serum cardiac troponins and N-terminal pro-brain natriuretic peptide: a staging system for primary systemic amyloidosis. J Clin Oncol. 2004;22(18):3751–7. doi: 10.1200/JCO.2004.03.029.
  11. Palladini G, Dispenzieri A, Gertz MA, et al. New criteria for response to treatment in immunoglobulin light chain amyloidosis based on free light chain measurement and cardiac biomarkers: impact on survival outcomes. J Clin Oncol. 2012;30(36):4541–9. doi: 10.1200/JCO.2011.37.7614.
  12. Palladini G, Hegenbart U, Milani P, et al. A staging system for renal outcome and early markers of renal response to chemotherapy in AL amyloidosis. Blood. 2014;124(15):2325–32. doi: 10.1182/blood-2014-04-570010.
  13. Dittrich T, Bochtler T, Kimmich C, et al. AL amyloidosis patients with low amyloidogenic free light chain levels at first diagnosis have an excellent prognosis. Blood. 2017;130(5):632–42. doi: 10.1182/blood-2017-02-767475.
  14. Milani P, Basset M, Russo F, et al. Patients with light-chain amyloidosis and low free light-chain burden have distinct clinical features and outcome. Blood. 2017;130(5):625–31. doi: 10.1182/blood-2017-02-767467.
  15. Kumar S, Dispenzieri A, Lacy MQ, et al. Revised prognostic staging system for light chain amyloidosis incorporating cardiac biomarkers and serum free light chain measurements. J Clin Oncol. 2012;30(9):989–95. doi: 10.1200/JCO.2011.38.5724.
  16. Comenzo RL, Reece D, Palladini G, et al. Consensus guidelines for the conduct and reporting of clinical trials in systemic light-chain amyloidosis. Leukemia. 2012;26(11):2317–25. doi: 10.1038/leu.2012.100.
  17. Palladini G, Kastritis E, Maurer MS, et al. Daratumumab plus CyBorD for patients with newly diagnosed AL amyloidosis: safety run-in results of ANDROMEDA. Blood. 2020;136(1):71–80. doi: 10.1182/blood.2019004460.
  18. Wechalekar A, Foard D, Rannagan L, et al. Characteristics and outcomes of 714 patients with systemic al amyloidosis – analysis of a prospective observational study (ALCHEMY study) [abstract]. Haematologica. 2014;99:221.
  19. Dispenzieri A, Dingli D, Kumar SK, et al. Discordance between serum cardiac biomarker and immunoglobulin-free light-chain response in patients with immunoglobulin light-chain amyloidosis treated with immune modulatory drugs. Am J Hematol. 2010;85(10):757–9. doi: 10.1002/ajh.21822.
  20. Dinner S, Witteles W, Afghahi A, et al. Lenalidomide, melphalan and dexamethasone in a population of patients with immunoglobulin light chain amyloidosis with high rates of advanced cardiac involvement. Haematologica. 2013;98(10):1593–9. doi: 10.3324/haematol.2013.084574.