Bispecific Antibodies in Clinical Practice and Clinical Trials (Literature Review)

ON Solopova, VA Misyurin

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

For correspondence: Vsevolod Andreevich Misyurin, PhD in Biology, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(985)436-30-19; e-mail: vsevolod.misyurin@gmail.com

For citation: Solopova ON, Misyurin VA. Bispecific Antibodies in Clinical Practice and Clinical Trials (Literature Review). Clinical oncohematology. 2019;12(2):125–44.

DOI: 10.21320/2500-2139-2019-12-2-125-144


ABSTRACT

Therapeutic monoclonal antibodies have long been an effective tool deployed by physicians of different specialties, particularly by oncologists. Bispecific antibodies opened up new horizons in the treatment of cancer as they allow to involve a patient’s endogenous immunity in his or her fight against tumor. The present review covers all the formats and strategies used in engineering of bispecific antibodies which reached the stage of clinical trials and also focuses on the available results of these clinical trials.

Keywords: bispecific antibodies, blinatumomab, catumaxomab, emicizumab, clinical trial.

Received: August 13, 2018

Accepted: January 22, 2019

Read in PDF 


REFERENCES

  1. Monoclonal Antibodies Approved by the EMA and FDA for Therapeutic Use (status 2017) [Internet]. Available from: http://www.actip.org/products/monoclonal-antibodies-approved-by-the-ema-and-fda-for-therapeutic-use. (accessed 12.08.2018).

  2. Drugs@FDA: FDA Approved Drug Products [Internet]. Available from: https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm. (accessed 12.08.2018).

  3. Lindhofer H, Mocikat R, Steipe B, Thierfelder S. Preferential species-restricted heavy/light chain pairing in rat/mouse quadromas. Implications for a single-step purification of bispecific antibodies. J Immunol. 1995;155(1):219–25.

  4. Ridgway JB, Presta LG, Carter P. ‘Knobs-into-holes’ engineering of antibody CH3 domains for heavy chain heterodimerization. Protein Eng. 1996;9(7):617–21. doi: 10.1093/protein/9.7.617.

  5. Milstein C, Cuello AC. Hybrid hybridomas and their use in immunohistochemistry. Nature. 1983;305(5934):537–40. doi: 10.1038/305537a0.

  6. Scientific Trion Research [Internet]. Available from: http://www.trionresearch.com/index.php?id=88. (accessed 12.08.2018).

  7. Goere D, Glehen O, Mariette C, et al. Results of a phase II randomized study evaluating the potential benefit of a postoperative intraperitoneal immunotherapy after resection of peritoneal metastases from gastric carcinoma metastases (IIPOP-NCT01784900). J Clin Oncol. 2017;35(15_suppl):4064. doi: 10.1200/JCO.2017.35.15_suppl.4064.

  8. Borlak J, Langer F, Spanel R, et al. Immune-mediated liver injury of the cancer therapeutic antibody catumaxomab targeting EpCAM, CD3 and Fcγ Oncotarget. 2016;7(19):28059–74. doi: 10.18632/oncotarget.8574.

  9. Kurbacher CM, Horn O, Kurbacher JA, et al. Outpatient Intraperitoneal Catumaxomab Therapy for Malignant Ascites Related to Advanced Gynecologic Neoplasms. Oncologist. 2015;20(11):1333–41. doi: 10.1634/theoncologist.2015-0076.

  10. Study of the Trifunctional Antibody FBTA05 and Donor Lymphocyte Infusion in B-cell Lymphoma After Allogeneic Stem Cell Transplantation (STP-LYM-01). In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2010. Available from: https://clinicaltrials.gov/ct2/show/NCT01138579. (accessed 12.08.2018).

  11. Buhmann R, Michael S, Juergen H, et al. Immunotherapy with FBTA05 (Bi20), a trifunctional bispecific anti-CD3 х anti-CD20 antibody and donor lymphocyte infusion (DLI) in relapsed or refractory B-cell lymphoma after allogeneic stem cell transplantation: study protocol of an investigator-driven, open-label, non-randomized, uncontrolled, dose-escalating phase I/II-trial. J Transl Med. 2013;11(1):160. doi: 10.1186/1479-5876-11-160.

  12. Kiewe P, Hasmuller S, Kahlert S, et al. Phase I trial of the trifunctional anti-HER2 х anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clin Cancer Res. 2006;12(10):3085–91. doi: 10.1158/1078-0432.CCR-05-2436.

  13. Garber K. Bispecific antibodies rise again. Nat Rev Drug Discov. 2014;13(11):799–801. doi: 10.1038/nrd4478.

  14. Wu J, Fu J, Zhang M, Liu D. Blinatumomab: a bispecific T cell engager (BiTE) antibody against CD19/CD3 for refractory acute lymphoid leukemia. J Hematol Oncol. 2015;8(1):104. doi: 10.1186/s13045-015-0195-4.

  15. Zugmaier G, Klinger M, Schmidt M, Subklewe M. Clinical overview of anti-CD19 BiTE(®) and ex vivo data from anti-CD33 BiTE(®) as examples for retargeting T cells in hematologic malignancies. Mol Immunol. 2015;67(2):58–66. doi: 10.1016/j.molimm.2015.02.033.

  16. Kohnke T, Krupka C, Tischer J, et al. Increase of PD-L1 expressing B-precursor ALL cells in a patient resistant to the CD19/CD3-bispecific T cell engager antibody blinatumomab. J Hematol Oncol. 2015;8(1):111. doi: 10.1186/s13045-015-0213-6.

  17. Martinelli G, Boissel N, Chevallier P, et al. Complete hematologic and molecular response in adult patients with relapsed/refractory philadelphia chromosome-positive B-precursor acute lymphoblastic leukemia following treatment with blinatumomab: Results From a Phase II, Single-Arm, Multicenter Study. J Clin Oncol. 2017;35(16):1795–802. doi: 10.1200/JCO.2016.69.3531.

  18. Kantarjian H, Stein A, Gokbuget N, et al. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. N Engl J Med. 2017;376(9):836–47. doi: 10.1056/NEJMoa1609783.

  19. Munz M, Baeuerle PA, Gires O. The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res. 2009;69(14):5627–9. doi: 10.1158/0008-5472.CAN-09-0654.

  20. Pishvaian M, Morse MA, McDevitt J, et al. Phase 1 Dose Escalation Study of MEDI-565, a Bispecific T-Cell Engager that Targets Human Carcinoembryonic Antigen, in Patients With Advanced Gastrointestinal Adenocarcinomas. Clin Colorect Cancer. 2016;15(4):345–51. doi: 10.1016/j.clcc.2016.07.009.

  21. Friedrich M, Henn A, Raum T, et al. Preclinical characterization of AMG 330, a CD3/CD33-bispecific T-cell-engaging antibody with potential for treatment of acute myelogenous leukemia. Mol Cancer Ther. 2014;13(6):1549–57. doi: 10.1158/1535-7163.MCT-13-0956.

  22. Lee D, Kim D, Choi YB, et al. Simultaneous blockade of VEGF and DLL4 by HD105, a bispecific antibody, inhibits tumor progression and angiogenesis. MAbs. 2016;8(5):892–904. doi: 10.1080/19420862.2016.1171432.

  23. Gurney A, Hoey T. Anti-DLL4, a cancer therapeutic with multiple mechanisms of action. Vasc Cell. 2011;3(1):18. doi: 10.1186/2045-824X-3-18.

  24. Aliperta R, Cartellieri M, Feldmann A, et al. Bispecific antibody releasing-mesenchymal stromal cell machinery for retargeting T cells towards acute myeloid leukemia blasts. Blood Cancer J. 2015;5(9):e348. doi: 10.1038/bcj.2015.73.

  25. Grosse-Hovest L, Muller S, Minoia R, et al. Cloned transgenic farm animals produce a bispecific antibody for T cell-mediated tumor cell killing. Proc Natl Acad Sci USA. 2004;101(18):6858–63. doi: 10.1073/pnas.0308487101.

  26. Grosse-Hovest L, Hartlapp I, Marwan W, et al. A recombinant bispecific single-chain antibody induces targeted, supra-agonistic CD28-stimulation and tumor cell killing. Eur J Immunol. 2003;33(5):1334–40. doi: 10.1002/eji.200323322.

  27. Lenting PJ, Denis CV, Christophe OD. Emicizumab, a bispecific antibody recognizing coagulation factors IX and X: how does it actually compare to factor VIII? Blood. 2017;130(23):2463–8. doi: 10.1182/blood-2017-08-801662.

  28. Oldenburg J, Mahlangu JN, Kim B, et al. Emicizumab Prophylaxis in Hemophilia A with Inhibitors. N Engl J Med. 2017;377(9):809–18. doi: 10.1056/NEJMoa1703068.

  29. Sampei Z, Igawa T, Soeda T, et al. Identification and multidimensional optimization of an asymmetric bispecific IgG antibody mimicking the function of factor VIII cofactor activity. PLoS One. 2013;8(2):e57479. doi: 10.1371/journal.pone.0057479.

  30. Merchant AM, Zhu Z, Yuan JQ, et al. An efficient route to human bispecific IgG. Nat Biotechnol. 1998;16(7):677–81. doi: 10.1038/nbt0798-677.

  31. Ishiguro T, Sano Y, Komatsu SI, et al. An anti-glypican 3/CD3 bispecific T cell-redirecting antibody for treatment of solid tumors. Sci Transl Med. 2017;9(410):aaal4291. doi: 10.1126/scitranslmed.aal4291.

  32. Fury MG, Lipton A, Smith KM, et al. A phase-I trial of the epidermal growth factor receptor directed bispecific antibody MDX-447 without and with recombinant human granulocyte-colony stimulating factor in patients with advanced solid tumors. Cancer Immunol Immunother. 2007;57(2):155–63. doi: 10.1007/s00262-007-0357-5.

  33. Hearn A, York IA, Bishop C, Rock KL. Characterizing the specificity and cooperation of aminopeptidases in the cytosol and endoplasmic reticulum during MHC class I antigen presentation. J Immunol. 2010;184(9):4725–32. doi: 10.4049/jimmunol.0903125.

  34. Fury MG, Lipton A, Smith KM, et al. A phase-I trial of the epidermal growth factor receptor directed bispecific antibody MDX-447 without and with recombinant human granulocyte-colony stimulating factor in patients with advanced solid tumors. Cancer Immunol Immunother. 2007;57(2):155–63. doi: 10.1007/s00262-007-0357-5.

  35. James ND, Atherton PJ, Jones J, et al. A phase II study of the bispecific antibody MDX-H210 (anti-HER2 х CD64) with GM-CSF in HER2+ advanced prostate cancer. Br J Cancer. 2001;85(2):152–6. doi: 10.1054/bjoc.2001.1878.

  36. Atta-ur-Rahman. Frontiers in Clinical Drug Research. Hematology. 2014;1:378. doi: 10.2174/97816080585871140101.

  37. Barbara Ann Karmanos Cancer Institute; National Cancer Institute (NCI). Treated T Cells Followed by a Stem Cell Transplant in Treating Patients With Multiple Myeloma. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2009. Available from: https://clinicaltrials.gov/ct2/show/NCT00938626. (accessed 12.08.2018).

  38. Haoyang Yu, Zhengcheng LI, Jing Su. Bispecific antibody capable of being combined with immune cells to enhance tumor killing capability, and preparation method therefor and application thereof. US patent No. 20180021440A1. Available from: https://patents.google.com/patent/US20180021440A1/en. (accessed 12.08.2018).

  39. Introna M, Franceschetti M, Ciocca A, et al. Rapid and massive expansion of cord blood-derived cytokine-induced killer cells: an innovative proposal for the treatment of leukemia relapse after cord blood transplantation. Bone Marrow Transplant. 2006;38(9):621–7. doi: 10.1038/sj.bmt.1705503.

  40. Lee HK, Kim YG, Kim JS, et al. Cytokine-induced killer cells interact with tumor lysate-pulsed dendritic cells via CCR5 signaling. Cancer Lett. 2016;378(2):142–9. doi: 10.1016/j.canlet.2016.05.020.

  41. Linn YC, Lau LC, Hui KM. Generation of cytokine-induced killer cells from leukaemic samples with in vitro cytotoxicity against autologous and allogeneic leukaemic blasts. Br J Haematol. 2002;116(1):78–86. doi: 10.1046/j.1365-2141.2002.03247.x.

  42. Linn YC, Wang SM, Hui KM. Comparative gene expression profiling of cytokine-induced killer cells in response to acute myeloid leukemic and acute lymphoblastic leukemic stimulators using oligonucleotide arrays. Exp Hematol. 2005;33(6):671–81. doi: 10.1016/j.exphem.2005.03.005.

  43. Lopez RD, Waller EK, Lu PH, Negrin RS. CD58/LFA-3 and IL-12 provided by activated monocytes are critical in the in vitro expansion of CD56+ T cells. Cancer Immunol Immunother. 2001;49(12):629–40. doi: 10.1007/s002620000148.

  44. Anderson PM, Bach FH, Ochoa AC. Augmentation of cell number and LAK activity in peripheral blood mononuclear cells activated with anti-CD3 and interleukin-2. Cancer Immunol Immunother. 1988;27(1):82–8. doi: 10.1007/BF00205763.

  45. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer. 2004;4(1):45–60. doi: 10.1038/nrc1251.

  46. Treder M, Reusch U, Marschner J-P, et al. Combination of a cd30xcd16 antibody with a pd-1 antagonist for therapy. World patent No. 2016177846A1. Available from: https://patents.google.com/patent/WO2016177846A1/en. (accessed 12.08.2018).

  47. Reusch U, Duell J, Ellwanger K, et al. A tetravalent bispecific TandAb (CD19/CD3), AFM11, efficiently recruits T cells for the potent lysis of CD19(+) tumor cells. MAbs. 2015;7(3):584–604. doi: 10.1080/19420862.2015.1029216.

  48. Munoz L, Nomdedeu JF, Lopez O, et al. Interleukin-3 receptor alpha chain (CD123) is widely expressed in hematologic malignancies. Haematologica. 2001;86(12):1261–9.

  49. Testa U, Pelosi E, Frankel A. CD 123 is a membrane biomarker and a therapeutic target in hematologic malignancies. Biomark Res. 2014;2(1):4. doi: 10.1186/2050-7771-2-4.

  50. Heath JK, White SJ, Johnstone CN, et al. The human A33 antigen is a transmembrane glycoprotein and a novel member of the immunoglobulin superfamily. Proc Natl Acad Sci USA. 1997;94(2):469–74.

  51. Moore PA, Shah K, Yang Y, et al. Development of MGD007, a gpA33 х CD3-Bispecific DART Protein for T-Cell Immunotherapy of Metastatic Colorectal Cancer. Mol Cancer Ther. 2018;17(8):1761–72. doi: 10.1158/1535-7163.MCT-17-1086.

  52. Andrews LP, Marciscano AE, Drake CG, Vignali DA. LAG3 (CD223) as a cancer immunotherapy target. Immunol Rev. 2017;276(1):80–96. doi: 10.1111/imr.12519.

  53. Rockville MD. MacroGenics Presents Updated Data from Phase 1 Study of MGD010 at Annual European Congress of Rheumatology (EULAR 2017) [Internet]. Available from: http://ir.macrogenics.com/static-files/e6d9617a-90cb-4bb4–8520-fe59d8533ed6. (accessed 12.08.2018).

  54. Phase 1 Study Of PF-06863135, A BCMA-CD3 Bispecific Ab, In Relapse/Refractory Multiple Myeloma. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2017. Available from: https://clinicaltrials.gov/ct2/show/NCT03269136. (accessed 12.08.2018).

  55. Panowski SH, Kuo T, Chen A, et al. Preclinical Evaluation of a Potent Anti-Bcma CD3 Bispecific Molecule for the Treatment of Multiple Myeloma. Blood 2016;128:383.

  56. Regula JT, Lundh von Leithner P, Foxton R, et al. Targeting key angiogenic pathways with a bispecific CrossMAb optimized for neovascular eye diseases. EMBO Mol Med. 2016;8(11):1265–88. doi: 10.15252/emmm.201505889.

  57. Campochiaro PA, Hafiz G, Mir TA, et al. Pro-permeability Factors in Diabetic Macular Edema; the Diabetic Macular Edema Treated With Ozurdex Trial. Am J Ophthalmol. 2016;168:13–23. doi: 10.1016/j.ajo.2016.04.017.

  58. Pressman D. Radiolabeled antibodies. Ann NY Acad Sci. 1957;69(4):644–50. doi: 10.1111/j.1749-6632.1957.tb49702.x.

  59. Blythman HE, Casellas P, Gros O, et al. Immunotoxins: hybrid molecules of monoclonal antibodies and a toxin subunit specifically kill tumour cells. Nature. 1981;290(5802):145–6. doi: 10.1038/290145a0.

  60. Vallera DA, Todhunter DA, Kuroki DW, et al. A bispecific recombinant immunotoxin, DT2219, targeting human CD19 and CD22 receptors in a mouse xenograft model of B-cell leukemia/lymphoma. Clin Cancer Res. 2005;11(10):3879–88. doi: 10.1158/1078-0432.CCR-04-2290.

  61. Vallera DA, Chen H, Sicheneder AR, et al. Genetic alteration of a bispecific ligand-directed toxin targeting human CD19 and CD22 receptors resulting in improved efficacy against systemic B cell malignancy. Leuk Res. 2009;33(9):1233–42. doi: 10.1016/j.leukres.2009.02.006.

  62. Bachanova V, Frankel AE, Cao Q, et al. Phase I study of a bispecific ligand-directed toxin targeting CD22 and CD19 (DT2219) for refractory B-cell malignancies. Clin Cancer Res. 2015;21(6):1267–72. doi: 10.1158/1078-0432.CCR-14-2877.

  63. Vallera DA, Oh S, Chen H, et al. Bioengineering a unique deimmunized bispecific targeted toxin that simultaneously recognizes human CD22 and CD19 receptors in a mouse model of B-cell metastases. Mol Cancer Ther. 2010;9(6):1872–83. doi: 10.1158/1535-7163.MCT-10-0203.

  64. Taylor K, Howard CB, Jones ML, et al. Nanocell targeting using engineered bispecific antibodies. MAbs. 2015;7(1):53–65. doi: 10.4161/19420862.2014.985952.

  65. Anselmo AC, Mitragotri S. Nanoparticles in the clinic. Bioeng Transl Med. 2016;1(1):10–29. doi: 10.1002/btm2.10003.

  66. van Zandwijk N, Pavlakis N, Kao SC, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386–96. doi: 10.1016/S1470-2045(17)30621-6.

  67. Solomon BJ, Desai J, Rosenthal M, et al. A First-Time-In-Human Phase I Clinical Trial of Bispecific Antibody-Targeted, Paclitaxel-Packaged Bacterial Minicells. PLoS One. 2015;10(12):e0144559. doi: 10.1371/journal.pone.0144559.

  68. Labrijn AF, Meesters JI, Priem P, et al. Controlled Fab-arm exchange for the generation of stable bispecific IgG1. Nat Protoc. 2014;9(10):2450–63. doi: 10.1038/nprot.2014.169.

  69. Atamaniuk J, Gleiss A, Porpaczy E, et al. Overexpression of G protein-coupled receptor 5D in the bone marrow is associated with poor prognosis in patients with multiple myeloma. Eur J Clin Invest. 2012;42(9):953–60. doi: 10.1111/j.1365-2362.2012.02679.x.

  70. Janssen Research & Development, LLC. A Dose Escalation Study of JNJ-61186372 in Participants With Advanced Non-Small Cell Lung Cancer. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2018. Available from: https://clinicaltrials.gov/ct2/show/NCT02609776. (accessed 12.08.2018).

  71. Moretti P, Skegro D, Ollier R, et al. BEAT® the bispecific challenge: a novel and efficient platform for the expression of bispecific IgGs. BMC Proceed. 2013;7(Suppl 6):O9. doi: 10.1186/1753-6561-7-s6-o9.

  72. Skegro D, Stutz C, Ollier R, et al. Immunoglobulin domain interface exchange as a platform technology for the generation of Fc heterodimers and bispecific antibodies. J Biol Chem. 2017;292(23):9745759. doi: 10.1074/jbc.M117.782433.

  73. Bzymek KP, Puckett JW, Zer C, et al. Mechanically interlocked functionalization of monoclonal antibodies. Nat Commun. 2018;9(1):1580. doi: 10.1038/s41467-018-03976-5.

  74. Moore GL, Bautista C, Pong E, et al. A novel bispecific antibody format enables simultaneous bivalent and monovalent co-engagement of distinct target antigens. MAbs. 2011;3(6):546–57. doi: 10.4161/mabs.3.6.18123.

  75. Lazar GA, Dang W, Karki S, Engineered antibody Fc variants with enhanced effector function. Proc Natl Acad Sci USA. 2006;103(11):4005–10. doi: 10.1073/pnas.0508123103.

  76. Kiviniemi A, Gardberg M, Kivinen K, et al. Somatostatin receptor 2A in gliomas: Association with oligodendrogliomas and favourable outcome. Oncotarget. 2017;8(30):49123–32. doi: 10.18632/oncotarget.17097.

  77. Adams RL, Adams IP, Lindow SW, et al. Somatostatin receptors 2 and 5 are preferentially expressed in proliferating endothelium. Br J Cancer. 2005;92(8):1493–8. doi: 10.1038/sj.bjc.6602503.

  78. Effectiveness and Safety of SAR156597 in Treating Diffuse Systemic Sclerosis. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2018. Available from: https://clinicaltrials.gov/ct2/show/NCT02921971. (accessed 12.08.2018).

  79. Liu L. A Novel MET-EGFR Bispecific Antibody LY3164530 Shows Advantage Over Combining MET and EGFR Antibodies In Tumor Inhibition and Overcoming Resistance [Internet]. Available from: http://bispecific.com/wp-content/uploads/sites/90/2016/10/1630-Ling-Lui-YES.pdf. (accessed 12.08.2018).

  80. McDonagh CF, Huhalov A, Harms BD, et al. Antitumor activity of a novel bispecific antibody that targets the ErbB2/ErbB3 oncogenic unit and inhibits heregulin-induced activation of ErbB3. Mol Cancer Ther. 2012;11(3):582–93. doi: 10.1158/1535-7163.MCT-11-0820.

  81. Richards DA, Braiteh FS, Garcia AA, et al. A phase 1 study of MM-111, a bispecific HER2/HER3 antibody fusion protein, combined with multiple treatment regimens in patients with advanced HER2-positive solid tumors. J Clin Oncol. 2014;32(15s):651. doi: 10.1200/jco.2014.32.15_suppl.651.

  82. Fitzgerald JB, Johnson BW, Baum J, et al. MM-141, an IGF-IR- and ErbB3-directed bispecific antibody, overcomes network adaptations that limit activity of IGF-IR inhibitors. Mol Cancer Ther. 2014;13(2):410–25. doi: 10.1158/1535-7163.MCT-13-0255.

  83. Camblin AJ, Pace EA, Adams S, et al. Dual Inhibition of IGF-1R and ErbB3 Enhances the Activity of Gemcitabine and Nab-Paclitaxel in Preclinical Models of Pancreatic Cancer. Clin Cancer Res. 2018;24(12):2873–85. doi: 10.1158/1078-0432.CCR-17-2262.

  84. Tsai YC, Tsai TF. Anti-interleukin and interleukin therapies for psoriasis: current evidence and clinical usefulness. Ther Adv Musculoskelet Dis. 2017;9(11):277–94. doi: 10.1177/1759720X17735756.

  85. Dahlen E, Veitonmaki N, Norlen P. Bispecific antibodies in cancer immunotherapy. Ther Adv Vaccines Immunother. 2018;6(1):3–17. doi: 10.1177/2515135518763280.

  86. Els Conrath K, Lauwereys M, Wyns L, Muyldermans S. Camel single-domain antibodies as modular building units in bispecific and bivalent antibody constructs. J Biol Chem. 2001;276(10):7346–50. doi: 10.1074/jbc.M007734200.

  87. Elsadek B, Kratz F. Impact of albumin on drug delivery-new applications on the horizon. J Contr Release. 2012;157(1):4–28. doi: 10.1016/j.jconrel.2011.09.069.

  88. Moses E, Zeldin RK. Topline results from the Phase IIb monotherapy study of vobarilizumab, ALX-0061 (anti-IL-6R), in patients with moderate to severe RA. [Internet] Available from: http://www.ablynx.com/uploads/data/files/160706%20alx-0061%20c202%20webcast_final.pdf. (accessed 12.08.2018).

  89. Merck KGaA. Multiple Ascending Dose Trial of MSB0010841 (Anti-IL17A/F Nanobody) in Psoriasis Subjects. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US); 2017. Available from: https://clinicaltrials.gov/ct2/show/results/NCT02156466. (accessed 12.08.2018).

  90. Насонов Е.Л., Мазуров В.И., Усачева Ю.В. и др. Разработки отечественных оригинальных генно-инженерных биологических препаратов для лечения иммуновоспалительных ревматических заболеваний. Научно-практическая ревматология. 2017;55(2):201–10. doi: 10.14412/1995-4484-2017-201-210.

    [Nasonov EL, Mazurov VI, Usacheva YuV, et al. Developments of Russian original biological agents for the treatment of immunoinflammatory rheumatic diseases. Nauchno-prakticheskaya revmatologiya. 2017;55(2):201–10. doi: 10.14412/1995-4484-2017-201-210. (In Russ)]

  91. Костарева О.С. Исследование структурных основ взаимодействия провоспалительного цитокина с наноантителом для разработки высокоэффективных терапевтических препаратов [электронный документ]. Доступно по: http://рнф.рф/prjcard_int?17-74-10156. Ссылка активна на 12.08.2018.

    [Kostareva OS. The study of structural background of the interaction of a pro-inflammatory cytokine with a nanoantibody for the development of highly effective drugs [Internet]. Available from: http://рнф.рф/prjcard_int?17-74-10156. (accessed 12.08.2018) (In Russ)]

  92. Toft-Petersen M, Nederby L, Kjeldsen E, et al. Unravelling the relevance of CLEC12A as a cancer stem cell marker in myelodysplastic syndrome. Br J Haematol. 2016;175(3):393–401. doi: 10.1111/bjh.14270.

  93. Smith EJ, Olson K, Haber LJ, et al. A novel, native-format bispecific antibody triggering T-cell killing of B-cells is robustly active in mouse tumor models and cynomolgus monkeys. Sci Rep. 2015;5(1):17943. doi: 10.1038/srep17943.

  94. Murphy AJ, Macdonald LE, Stevens S, et al. Mice with megabase humanization of their immunoglobulin genes generate antibodies as efficiently as normal mice. Proc Natl Acad Sci USA. 2014;111(14):5153–8. doi: 10.1073/pnas.1324022111.

  95. Li Y, Hickson JA, Ambrosi DJ, et al. ABT-165, a Dual Variable Domain Immunoglobulin (DVD-Ig) Targeting DLL4 and VEGF, Demonstrates Superior Efficacy and Favorable Safety Profiles in Preclinical Models. Mol Cancer Ther. 2018;17(5):1039–50. doi: 10.1158/1535-7163.MCT-17-0800.

  96. Shutter JR, Scully S, Fan W, et al. DLL4, a novel Notch ligand expressed in arterial endothelium. Gen Dev. 2000;14(11):1313–8.

  97. Wozniak-Knopp G, Bartl S, Bauer A, et al. Introducing antigen-binding sites in structural loops of immunoglobulin constant domains: Fc fragments with engineered HER2/neu-binding sites and antibody properties. Protein Eng Des Sel. 2010;23(4):289–97. doi: 10.1093/protein/gzq005.

  98. Rucker F, Himmler G, Wozniak-Knopp G. Novel multivalent immunoglobulins. World patent No. 2008003103A2 [Internet]. Available from: https://patents.google.com/patent/WO2008003103A2. (accessed 12.08.2018).

  99. Oates J, Hassan NJ, Jakobsen BK. ImmTACs for targeted cancer therapy: Why, what, how, and which. Mol Immunol. 2015;67(2):67–74. doi: 10.1016/j.molimm.2015.01.024.

  100. Sacco JJ, Kalirai H, Kenyani J, et al. Recent breakthroughs in metastatic uveal melanoma: a cause for optimism? Fut Oncol. 2018;14(14):1335–8. doi: 10.2217/fon-2018-0116.

  101. Von Kreudenstein TS, Escobar-Carbrera E, Lario PI, et al. Improving biophysical properties of a bispecific antibody scaffold to aid developability: quality by molecular design. MAbs. 2013;5(5):646–54. doi: 10.4161/mabs.25632.

  102. Yao J, Caballero OL, Yung WK? et al. Tumor subtype-specific cancer-testis antigens as potential biomarkers and immunotherapeutic targets for cancers. Cancer Immunol Res. 2014;2(4):371–9. doi: 10.1158/2326-6066.CIR-13-0088.

  103. Лыжко Н.А., Ахлынина Т.В., Мисюрин А.В. и др. Повышение уровня экспрессия гена PRAME в опухолевых клетках сопровождается локализацией белка в клеточном ядре. Российский биотерапевтический журнал. 2015;14(4):19–30.

    [Lyzhko NA, Akhlynina TV, Misyurin AV, et al. The increased PRAME expression in cancer cells is associated with deposit of the protein in cell nucleus. Rossiiskii bioterapevticheskii zhurnal. 2015;14(4):19–30. (In Russ)]

  104. Pankov D, Sjostrom L, Kalidindi T, et al. In vivo immuno-targeting of an extracellular epitope of membrane bound preferentially expressed antigen in melanoma (PRAME). Oncotarget. 2017;8(39):65917–31. doi: 10.18632/oncotarget.19579.

  105. European medicine agency. Public statement. Removab: withdrawal of the marketing authorisation in the European Union [Internet]. Available from: https://www.ema.europa.eu/documents/public-statement/public-statement-removab-withdrawal-marketing-authorisation-european-union_en.pdf. (accessed 19.01.2019).

  106. BLINCYTO® (blinatumomab) Approved In Japan For The Treatment Of Relapsed Or Refractory B-cell Acute Lymphoblastic Leukemia [Internet]. Available from: https://www.amgen.com/media/news-releases/2018/09/blincyto-blinatumomab-approved-in-japan-for-the-treatment-of-relapsed-or-refractory-bcell-acute-lymphoblastic-leukemia/ (accessed 19.01.2019).

  107. Scott LJ, Kim ES. Emicizumab-kxwh: First Global Approval. Drugs. 2018;78(2):269–74. doi: 10.1007/s40265-018-0861-2.

Clinical Efficacy of Chelation Therapy in Patients with Low-Risk Myelodysplastic Syndrome

SV Gritsaev, II Kostroma, AA Zhernyakova

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

For correspondence: Sergei Vasil’evich Gritsaev, MD, PhD, 16 2-ya Sovetskaya str., Saint Petersburg, Russian Federation, 191024; Tel.: +7(812)717-54-68; e-mail: gritsaevsv@mail.ru

For citation: Gritsaev SV, Kostroma II, Zhernyakova AA. Clinical Efficacy of Chelation Therapy in Patients with Low-Risk Myelodysplastic Syndrome. Clinical oncohematology. 2019;12(2):120–4.

DOI: 10.21320/2500-2139-2019-12-2-120-124


ABSTRACT

The present literature review provides evidence that in patients with low-risk myelodysplastic syndrome and transfusion dependence blood parameters and survival rates can be improved by administration of iron chelators. Dose adequacy and therapy duration underlie clinical efficacy of chelators. Toxicity can be reduced by administrating a new formula of deferasirox that does not need to be dissolved in liquid before consuming.

Keywords: myelodysplastic syndrome, low risk, transfusion dependence, iron chelators, survival.

Received: August 20, 2018

Accepted: February 2, 2019

Read in PDF 


REFERENCES

  1. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89(6):2079–88.

  2. Bennett JM. Consensus statement on iron overload in myelodysplastic syndromes. Am J Hematol. 2008;83(11):858–61. doi: 10.1002/ajh.21269.

  3. Malcovati L, Porta MG, Pascutto C, et al. Prognostic factors and life expectancy in myelodysplastic syndromes classified according to WHO criteria: a basis for clinical decision making. J Clin Oncol. 2005;23(30):7594–603. doi: 1200/JCO.2005.01.7038.

  4. Steensma DP, Bennett JM. The myelodysplastic syndromes: Diagnosis and treatment. Mayo Clin Proc. 2006;81(1):104–30. doi: 10.4065/81.1.104.

  5. Mitchell M, Gore SD, Zeidan AM. Iron chelation therapy in myelodysplastic syndromes: where do we stand? Expert Rev Hematol. 2013;6(4):397–410. doi: 10.1586/17474086.2013.814456.

  6. Gattermann N, Jarisch A, Schlag R, et al. Deferasirox treatment of iron-overloaded chelation-naive and prechelated patients with myelodysplastic syndromes in medical practice: results from the observational studies eXtend and eXjange. Eur J Haematol. 2012;88(3):260–8. doi: 10.1111/j.1600-0609.2011.01726.x.

  7. List AF, Baer MR, Steensma DP, et al. Deferasirox reduces serum ferritin and labile plasma iron in RBC transfusion-dependent patients with myelodysplastic syndrome. J Clin Oncol. 2012;30(17):2134–9. doi: 10.1200/JCO.2010.34.1222.

  8. Greenberg PL, Koller CA, Cabantchik ZI, et al. Prospective assessment of effects on iron-overload parameters of deferasirox therapy in patients with myelodysplastic syndromes. Leuk Res. 2010;34(12):1560–5. doi: 10.1016/j.leukres.2010.06.013.

  9. Remacha AF, Arrizabalaga B, Del Canizo C, et al. Iron overload and chelation therapy in patients with low-risk myelodysplastic syndromes with transfusion requirements. Ann Hematol. 2010;89(2):147–54. doi: 10.1007/s00277-009-0794-7.

  10. Rose C, Brechignac S, Vassilief D, et al. Does iron chelation therapy improve survival in regularly transfused lower risk MDS patients? A multicenter study by the GFM (Groupe Francophone des Myelodysplasies). Leuk Res. 2010;34(7):864–70. doi: 10.1016/j.leukres.2009.12.004.

  11. Malcovati L. Impact of transfusion dependency and secondary iron overload on the survival of patients with myelodysplastic syndromes. Leuk Res. 2007;31(Suppl 3):S2–6. doi: 10.1016/S0145-2126(07)70459-9.

  12. Kohgo Y, Ikuta K, Ohtake T, et al. Body iron metabolism and pathophysiology of iron overload. Int J Hematol. 2008;88(1):7–15. doi: 10.1007/s12185-008-0120-5.

  13. Andrews NC. Closing the iron gate. N Engl J Med. 2012;366(4):376–7. doi: 10.1056/NEJMcibr1112780.

  14. Gardenghi S, Marongiu MF, Ramos P, et al. Ineffective erythropoiesis in beta-thalassemia is characterized by increased iron absorption mediated by down-regulation of hepcidin and up-regulation of ferroportin. 2007;109(11):5027–35. doi: 10.1182/blood-2006-09-048868.

  15. Andrews NC. Disorders of iron metabolism. N Engl J Med. 1999;341(26):1986–95. doi: 10.1056/NEJM199912233412607.

  16. Takatoku M, Uchiyama T, Okamoto S, et al. Retrospective nationwide survey of Japanese patients with transfusion-dependent MDS and aplastic anemia highlights the negative impact of iron overload on morbidity/mortality. Eur J Haematol. 2007;78(6):487–94. doi: 10.1111/j.1600-0609.2007.00842.x.

  17. Gattermann N. Iron overload in myelodysplastic syndromes. Int J Hematol. 2018;107(1):55–63. doi: 10.1007/s12185-017-2367-1.

  18. Lyons R, Marek B, Paleyc C, et al. Relation between chelation and clinical outcomes in lower-risk patients with myelodysplastic syndromes: Registry analysis at 5 years. Leuk Res. 2017;56:88–95. doi: 10.1016/j.leukres.2017.01.033.

  19. Leitch H, Parmar A, Wells R, et al. Overall survival in lower IPSS risk MDS by receipt of iron chelation therapy, adjusting for patient-related factors and measuring from time of first red blood cell transfusion dependence: an MDS-CAN analysis. Br J Haematol. 2017;179(1):83–97. doi: 10.1111/bjh.14825.

  20. Mainous A, Tanner R, Hulihan M, et al. The impact of chelation therapy on survival in transfusional iron overload: a meta-analysis of myelodysplastic syndrome. Br J Haematol. 2014;167(5):720–3. doi: 10.1111/bjh.13053.

  21. Abraham I, Yami M, Yun S et al. Survival outcomes in iron chelated and non-chelated patients with lower-risk myelodysplastic syndromes: Review and pooled analysis of observational studies. Leuk Res. 2017;57:104–8. doi: 10.1016/j.leukres.2017.03.007.

  22. Gattermann N, Finelli C, Della Porta M, et al. Hematologic responses to deferasirox therapy in transfusion-dependent patients with myelodysplastic syndromes. Haematologica. 2012;97(9):1364–71. doi: 10.3324/haematol.2011.048546.

  23. Pullarkat V. Objectives of iron chelation therapy in myelodysplastic syndromes: more than meets the eye? Blood. 2009;114(26):5251–5. doi: 10.1182/blood-2009-07-234062.

  24. Zeidan AM, Hendrick F, Friedmann E, et al. Deferasirox therapy is associated with reduced mortality risk in a medicare population with myelodysplastic syndromes. J Comp Eff Res. 2015;4(4):327–40. doi: 10.2217/cer.15.20.

  25. Improta S, Villa MR, Volpe A, et al. Transfusion-dependent low-risk myelodysplastic patients receiving deferasirox: Long-term follow-up. Oncol Lett. 2013;6(6):1774–8. doi: 10.3892/ol.2013.1617.

  26. Breccia M, Voso M, Spiriti M, et al. An increase in hemoglobin, platelets and white blood cells levels by iron chelation as single treatment in multitransfused patients with myelodysplastic syndromes: clinical evidences and possible biological mechanisms. Ann Hematol. 2015;94(5):771–7. doi: 10.1007/s00277-015-2341-z.

  27. Delforge M, Selleslag D, Beguin Y, et al. Adequate iron chelation therapy for at least six months improves survival in transfusion-dependent patients with lower risk myelodysplastic syndromes. Leuk Res. 2014;38(5):557–63. doi: 10.1016/j.leukres.2014.02.003.

  28. Cermak J, Jonasova A, Vondrakova J, et al. A comparative study of deferasirox and deferiprone in the treatment of iron overload in patients with myelodysplastic syndromes. Leuk Res. 2013;37(12):1612–5. doi: 10.1016/j.leukres.2013.07.021.

  29. Nolte F, Angelucci E, Breccia M, et al. Updated recommendations on the management of gastrointestinal disturbances during iron chelation therapy with deferasirox in transfusion dependent patients with myelodysplastic syndrome – emphasis on optimized dosing schedules and new formulations. Leuk Res. 2015;39(10):1028–33. doi: 10.1016/j.leukres.2015.06.008.

  30. Taher AT, Origa R, Perrotta S, et al. New film-coated tablet formulation of deferasirox is well tolerated in patients with thalassemia or lower-risk MDS: Results of the randomized, phase II ECLIPSE study. Am J Hematol. 2017;92(5):420–8. doi: 10.1002/ajh.24668.

  31. Джадену® (инструкция по медицинскому применению). Швейцария: Novartis pharma, AG. Доступно по: https://www.vidal.ru/drugs/dzhadenu. Ссылка активна на 30.11.2018. [Dzhadenu® (package insert). Switzerland: Novartis pharma, AG. Available from: https://www.vidal.ru/drugs/dzhadenu. (accessed 30.11.2018) (In Russ)]

Achievements and Challenges in Allogeneic Hematopoietic Stem Cell Transplantation in Cytogenetically Unfavorable Acute Leukemias (Literature Review)

NN Mamaev, TL Gindina, BV Afanas’ev

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

For correspondence: Nikolai Nikolaevich Mamaev, MD, PhD, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; e-mail: nikmamaev524@gmail.com

For citation: Mamaev NN, Gindina TL, Afanas’ev BV. Achievements and Challenges in Allogeneic Hematopoietic Stem Cell Transplantation in Cytogenetically Unfavorable Acute Leukemias (Literature Review). Clinical oncohematology. 2019;12(2):111–9.

DOI: 10.21320/2500-2139-2019-12-2-111-119


ABSTRACT

Literature review provides the analysis of treatment results of implementing allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with cytogenetically unfavorable acute myeloid and lymphoblastic leukemias including monosomal, complex, and hyperdiploid karyotypes, t(3;3)/inv(3), t(v;11)(v;q23), t(4;11)(q21;q23), t(9;22)(q34;q11) translocations, 17p abnormalities, and some other disorders. The major disadvantage of allo-HSCT seems to be linked to a strong chromosome-damaging effect of cytostatic drugs used in conditioning regimens which in turn is associated with additional chromosome abnormalities occurring in tumors, increasing genomic instability, and tumor progression. On the other hand, one of the advantages of allo-HSCT can consist in its specific “graft versus leukemia” (GVL) effect whose degree has not yet been adequately studied. To minimize the risks of allo-HSCT in above mentioned patients it appears appropriate to apply new treatment approaches based on de-escalation of chromosome- and whole-genome-damaging effects and also to introduce recent methods of active stimulation and qualitative assessment of GVL effect into clinical practice.

Keywords: acute leukemias, cytogenetically unfavorable variants, allo-HSCT, outcomes, additional chromosome abnormalities, “graft versus leukemia” effect.

Received: October 22, 2018

Accepted: February 2, 2019

Read in PDF 


REFERENCES

  1. Burnett AK, Hills RK. Who should be transplanted in first remission of acute myeloid leukemia? Curr Treatment Opt Oncol. 2011;12(4):329–40. doi: 10.1007/s11864-011-0169-x.

  2. Stelljes M, Beelen DW, Braess J, et al. Allogeneic transplantation as post-remission therapy for cytogenetically high-risk acute myeloid leukemia: landmark analysis from a single prospective multicenter trial. Haematologica. 2011;96(7):972–9. doi: 3324/haematol.2011.041004.

  3. Bejanyan N, Weisdorf DJ, Logan BR, et al. Survival of patients with acute myeloid leukemia relapsing after allogeneic hematopoietic cell transplantation: A Center for International Blood and Marrow Transplant Research Study. Biol Blood Marrow Transpl. 2015;21(3):454–9. doi: 10.1016/j.bbmt.2014.11.007.

  4. Dohner H, Estey EH, Grimwade D, et al. Diagnosis and management of acute myeloid leukemia in adults: 2017 ELN recommendation from an international expert panel. Blood. 2017;129(4):424–47. doi: 10.1182/blood-2016-08-733196.

  5. Vasu S, Kohlschmidt J, Mrozek K, et al. Ten-year outcome of patients with acute myeloid leukemia not treated with allogeneic transplantation in first complete remission. Blood Adv. 2018;2(13):1645–50. doi: 10.1182/bloodadvances.2017015222.

  6. de Lima M, Porter DL, Battiwalla M, et al. Proceedings from the National Cancer Institute’s Second International Workshop on the Biology, Prevention, and Treatment of Relapse after Hematopoietic Stem Cell Transplantation: Part III. Prevention and Treatment of Relapse after Allogeneic Transplantation. Biol Blood Marrow Transpl. 2014;20(1):4–13. doi: 1016/j.bbmt.2013.08.012.

  7. Christopeit М, Kroger N, Haferlach T, et al. Relapse assessment following allogeneic SCT in patients with MDS and AML. Ann Hematol. 2014;93(7):1097–110. doi: 1007/s00277-014-2046-8.

  8. Tsirigotis Р, Byrne M, Schmid C, et al. Relapse of AML after hematopoietic stem cell transplantation: methods of monitoring and preventive strategies. A review from the ALWP of the EBMT. Bone Marrow Transplant. 2016;51(11):1431–8. doi: 1038/bmt.2016.167.

  9. Cruz NM, Mencia-Trinchant N, Hassane DC, et al. Minimal residual disease in acute myelogenous leukemia. Int J Lab Hematol. 2017;39(Suppl 1):53–60. doi: 1111/ijlh.12670.

  10. Kroger N, Bishop M, Giralt S, et al. Third International workshop on the biology, prevention, and treatment of relapse after stem cell transplantation. Bone Marrow Transplant. 2018;53(1):1–2. doi: 10.1038/bmt.2017.218.

  11. Nahi H, Remberger M, Machaczka M, et al. Different impact of intermediate and unfavorable cytogenetics at the time of diagnosis of de novo AML after allo-SCT: a long-term retrospective analysis from a single institution. Med Oncol. 2012;29(4):2348–58. doi: 10.1007/s12032-011-0155-y.

  12. Cornelissen JJ, Blaise D. Hemopoietic stem cell transplantation for patients with AML in first complete remission. Blood. 2016;127(1):62–70. doi: 10.1182/blood-2015-07-604546.

  13. Gindina T, Mamaev N, Afanasyev B. Chromosome Abnormalities and Hematopoietic Stem Cell Transplantation. In: ML Larramendy, S Soloneski, eds. Chromosomal abnormalities – a hallmark manifestation of genomic instability. Croatia: INTECH; 2017. рр. 71–86. doi: 5772/67802.

  14. Hemmati PG, Schulze-Luchkov A, Terwey ThN, et al. Cytogenetic risk grouping by the monosomal karyotype classification is superior in predicting the outcome of acute myeloid leukemia undergoing allogeneic stem cell transplantation in complete remission. Eur J Hematol. 2013;92(2):102–10. doi: 10.1111/ejh.12216.

  15. Wang Y, Liu QF, Qin YZ, et al. Improved outcome of hematopoietic stem cell transplantation in a poor prognostic subgroup patients with mixed-lineage-leukemia-rearranged acute leukemia: results from a prospective, multicenter study. Am J Hematol. 2014;89(2):130–6. doi: 10.1002/ajh.23593.

  16. Parma M, Vigano C, Fumagalli M, et al. Good outcome for very high risk adult B cell acute lymphoblastic leukemia carring genetic abnormalities t(4;11)(q21;q23) or t(9;22)(q34;q11), if promtly submitted to allogeneic transplantation after obtaining a good molecular remission. Mediterr J Hematol Infect Dis. 2015;7(1):e2015041. doi: 10.4084/MJHID.2015.041.

  17. Fang M, Storer B, Estey E, et al. Outcome of patients with acute myeloid leukemia with monosomal karyotype who undergo hematopoietic stem cell transplantation. Blood. 2011;118(6):1490–4. doi: 10.1182/blood-2011-02-339721.

  18. Oran B, Dolan M, Cao Q, et al. Monosomal karyotype provides better prognostic prediction after allogeneic stem cell transplantation in patients with acute myelogenous leukemia. Biol Blood Marrow Transplant. 2011;17(3):356–64. doi: 10.1016/j.bbmt.2010.05.012.

  19. Cornelissen JJ, Breems D, van Putten WL, et al. Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories. J Clin Oncol. 2012;30(17):2140–6. doi: 10.1200/jco.2011.39.6499.

  20. Guo RJ, Atenafu FG, Craddock K, et al. Allogeneic hematopoietic cell transplantation may alleviate the negative prognostic impact of monosomal and complex karyotype on patients with acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20(5):690–5. doi: 10.1016/j.bbmt.2014.01.027.

  21. Pasquini M, Zhang M-J, Medeiros BC, et al. Hematopoietic cell transplantation outcomes in monosomal karyotype myeloid malignancies. Biol Blood Marrow Transplant. 2016;22(2):248–57. doi: 10.1016/j.bbmt.2015.08.024.

  22. Гиндина Т.Л., Мамаев Н.Н., Бондаренко С.Н. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при острых миелоидных лейкозах: прогностическое значение сложного кариотипа, включающего аномалии del(5q), –7, del(7q). Клиническая онкогематология. 2016;9(3):271–8. doi: 21320/2500-2139-2016-9-3-271-278.

    [Gindina TL, Mamaev NN, Bondarenko SN, et al. Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias: Prognostic Significance of Complex Karyotype Including del(5q), –7, del(7q) Abnormalities. Clinical oncohematology. 2016;9(3):271–8. 2016;9(3):271–8. doi: 10.21320/2500-2139-2016-9-3-271-278. (In Russ)]

  23. Koh K, Tomozawa D, Moriya Saito AM, et al. Early use of allogeneic hematopoietic stem cell transplantation for infants with MLL gene rearrangement-positive acute lymphoblastic leukemia. Leukemia. 2015;29(2):290–6. doi: 1038/leu.2014.172.

  24. Гиндина Т.Л., Мамаев Н.Н., Паина О.В.и др. Острый лимфобластный лейкоз c транслокацией t(4;11)(q21;q23)/KMT2A-AFF1: результаты аллогенной трансплантации гемопоэтических стволовых клеток у детей и взрослых. Клиническая онкогематология. 2017;10(3):342–50. doi: 21320/2500-2139-2017-10-3-342-350.

    [Gindina TL, Mamaev NN, Paina OV, et al. Acute Lymphoblastic Leukemia with t(4;11)(q21;q23)/KMT2A-AFF1 Translocation: The Results of Allogeneic Hematopoietic Stem Cells Transplantation in Children and Adults. Clinical oncohematology. 2017;10(3):342–50. doi: 10.21320/2500-2139-2017-10-3-342-350. (In Russ)]

  25. Poire X, Labopin M, Maertens J, et al. Allogeneic stem cell transplantation in adult patients with acute myeloid leukemia and 17p abnormalities in first complete remission: a study from the Acute Leukemia Working Party (ALWP) of the European Society for Blood and Marrow Transplantation (EBMT). J Hematol Oncol. 2017;10(1):20. doi: 10.1186/s13045-017-0393-3.

  26. Strickland SA, Sun Z, Ketterling RP, et al. Independent prognostic significance of monosomy 17 and impact of karyotype complexity in monosomal karyotype/complex karyotype acute myeloid leukemia: Results from FOUR ECOG-AGRIN prospective therapeutic trials. Leuk Res. 2017;59:55–64. doi: 10.1016/j.leukres.2017.05.010.

  27. Halaburda K, Labopin M, Houhou M, et al. AlloHSCT for inv(3)(q21;q26)t(3;3)(q21;q26) AML: a report from the acute leukemia working party of the European society for blood and marrow transplantation. Bone Marrow Transplant. 2018;53(6):683–91. doi: 10.1038/s41409-018-0165-x.

  28. Passerini V, Ozeri-Galai E, de Pagter MS, et al. The presence of extra chromosomes leads to genomic instability. Nat Commun. 2016;7(1):10754. doi: 10.1038/ncomms10754.

  29. Schmidt-Hieber M, Blau IW, Richter G, et al. Cytogenetic studies in acute leukemia patients relapsing after allogeneic stem cell transplantation. Cancer Gen Cytogenet. 2010;198(2):135–43. doi: 1016/j.cancergencyto.2010.01.005.

  30. Гиндина Т.Л., Мамаев Н.Н., БондаренкоС.Н. и др. Сложные хромосомные нарушения у больных с посттрансплантационными рецидивами острых лейкозов: клинические и теоретические аспекты. Клиническая онкогематология. 2015;8(1):69–77. doi: 10.21320/2500-2139-2015-8-1-69-77.

    [Gindina TL, Mamaev NN, Bondarenko SN, et al. Complex Chromosomal Aberrations in Patients with Post-Transplantation Relapses of Acute Leukemias: Clinical and Theoretical Aspects. Clinical oncohematology. 2015;8(1):69–77. doi: 10.21320/2500-2139-2015-8-1-69-77. (In Russ)]

  31. Breems DA, Van Putten WLL, De Greef GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol. 2008;26(29):4791–7. doi: 10.1200/jco.2008.16.0259.

  32. Kayzer S, Zucknick M, Dohner K, et al. Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies. Blood. 2011;119(2):551–8. doi: 10.1182/blood-2011-07-367508.

  33. Ciurea SM, Labopin G, Socie G, et al. Relapse and survival after transplantation for complex karyotype acute myeloid leukemia: a report from the acute leukemia working party of the European society for Blood and Marrow Transplantation and the University of Texas MD Anderson Cancer Center. 2018;124(10):2134–41. doi: 10.1002/cncr.31311.

  34. Schoch C, Haferlach T, Haase D, et al. Patients with the de novo acute myeloid leukemia and complex karyotype aberrations show a poor prognosis despite intensive treatment: a study of 90 patients. Br J Haematol. 2001;112(1):118–26. doi: 10.1046/j.1365-2141.2001.02511.x.

  35. Mrozek K. Cytogenetic, molecular genetics, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;35(4):365–77. doi: 10.1053/j.seminoncol.2008.04.007.

  36. Schoch C, Kern W, Kohlmann A, et al. Acute myeloid leukemia with a complex aberrant karyotype is a distinct biological entity characterized by genomic imbalance and a special gene expression profile. Genes Chromos Cancer. 2005;43(3):227–38. doi: 1002/gcc.20193.

  37. Гиндина Т.Л., Мамаев Н.Н., Бархатов И.М. и др. Сложные повреждения хромосом у больных с рецидивами острых лейкозов после аллогенной трансплантации гемопоэтических стволовых клеток. Терапевтический архив. 2012;84(8):61–6. [Gindina TL, Mamaev NN, Barkhatov IM, et al. Complex chromosome damages in patients with recurrent acute leukemias after allogeneic hematopoietic stem cell transplantations. Terapevticheskii arkhiv. 2012;84(8):61–6. (In Russ)]

  38. Bacher U, Haferlach T, Alpermann T, et al. Comparison of cytogenetic clonal evolution patterns following allogeneic hematopoietic transplantation versus convential treatment in patients at relapse of AML. Biol Blood Marrow Transplant. 2010;16(12):1649–57. doi: 10/1016/j.bbmt.2010.06.007.

  39. Chen Y, Kantarjian H, Pierce S, et al. Prognostic significance of 11q23 aberrations in adult acute myeloid leukemia and the role of allogeneic stem cell transplantation. Leukemia. 2013;27(4):836–42. doi: 1038/leu.2012.319.

  40. Yang H, Huang S, Zhu C-Y, et al. The superiority of allogeneic hematopoietic stem cell transplantation over chemotherapy alone in the treatment of acute myeloid leukemia patients with mixed lineage leukemia (MLL) rearrangements. Med Sci Monitor. 2016;22:2315–23. doi: 12659/MSM.899186.

  41. Pigneux A, Labopin M, Maertens J, et al. Outcome of allogeneic hematopoietic stem-cell transplantation in adult patients with AML and 11q23/MLL rearragement (MLL-r-AML). Leukemia. 2015;29(12):2375–81. doi: 1038/leu.2015.143.

  42. Gindina T, Mamaev N, Alyanskiy A, et al. Outcome of allogeneic hematopoietic stem cell transplantation in patients with KMT2A (MLL)-related leukemia, depending on number of transplanted CD34+ cells. Bone Marrow Transplant. 2015;50(Suppl 1):S481.

  43. Гиндина Т.Л., Мамаев Н.Н., Николаева Е.С. и др. Исход аллогенной трансплантации гемопоэтических стволовых клеток при острых миелоидных лейкозах с гипердиплоидным кариотипом. Клиническая онкогематология. 2016;9(4):383–90. doi: 21320/2500-2139-2016-9-4-383-390.

    [Gindina TL, Mamaev NN, Nikolaeva ES, et al. Outcome of Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemias with Hyperdiploid Karyotype. Clinical oncohematology. 2016;9(4):383–90. doi: 10.21320/2500-2139-2016-9-4-383-390. (In Russ)]

  44. Chevallier P, Labopin M, Nagler A, et al. Outcome after allogeneic transplantation for adult acute myeloid leukemia patients exhibiting isolated trisomy 8 chromosomal abnormality: a survey on behalf of the ALWP of the EBMT. Bone Marrow Transplant. 2009;44(9):589–94. doi: 10.1038/bmt.2009.68.

  45. Konuma T, Kondo T, Yamashita T, et al. Outocome of allogeneic hematopoietic stem cell transplantation in adult patients with acute myeloid leulkemia harboring trisomy 8. Ann Hematol. 2017;96(3):469–78. doi: 1007/s00277-016-2009-2.

  46. Herold T, Metzeler KH, Vosberg S, et al. Isolated trisomy 13 defines a homogenous AML subgroup with high frequency of mutations in splisome genes and poor prognosis. Blood. 2014;124(8):1304–11. doi: 10.1182/blood-2013-12-540716.

  47. Mohr B, Schetelig J, Schafer-Eckart K, et al. Impact of allogeneic haematopoietic stem cell transplantation in patients with abnl(17p) acute myeloid leukemia. Br J Haematol. 2013;161(2):237–44. doi: 10.1111/bjh.12253.

  48. Middeke JM, Fang M, Cornellisen JJ, et al. Outcome of patients with abnl(17p) acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Blood. 2014;123(19):2960–7. doi: 10.1182/blood-201312-544957.

  49. Vey N, Thomas X, Picard C, et al. Allogeneic stem cell transplantation improves the outcome of adults with t(1;19)/E2A-PBX1 and t(4;11)/MLL-AF4 positive B-cell acute lymphoblastic leukemia: results of the prospective multicenter LALA-94 study. Leukemia. 2006;20:2155–61. doi: 10.1038/sj.leu.2404420.

  50. Marks DI, Moorman AV, Chilton L, et al. The clinical characteristics, therapy and outcome of 85 adults with acute lymphoblastic leukemia and t(4;11)(q21q23)/MLL-AFF1 prospectively treated in the UKALLXII/ECOG2993 trial. 2013;98(6):945–52. doi: 10.3324/haematol.2012.081877.

  51. Cimino G, Cenfra N, Elia L, et al. The therapeutic response and clinical outcome of adults with ALL1(MLL)/AF4 fusion positive acute lymphoblastic leukemia according to the GIMEMA experience. Haematologica. 2010;95(5):837–40. doi: 10.3324/haematol.2009.009035.

  52. Kato M, Hasegawa D, Koh K, et al. Allogeneic haematopoietic stem cell transplantation for infant acute lymphoblastic leukemia with KMT2A (MLL) rearrangements: a retrospective study from the paediatric acute lymphoblastic leukemia working group of the Japan Society for Haematopoietic Cell Transplantation. Br J Haematol. 2014;168(4):564–70. doi: 10.1111/bjh.13174.

  53. Ribera JM, Oriol A, Gonzalez M, et al. Concurrent intensive chemotherapy and imatinib before and after stem cell transplantation in newly diagnosed Philadelphia chromosome‐positive acute lymphoblastic leukemia. Final results of the CSTIBES02 trial. 2010;95(1):87–95. doi: 10.3324/haematol.2009.011221.

  54. Ribera JM, Garcia O, Montesinos P, et al. Treatment of young patients with Philadelphia chromosome-positive acute lymphoblastic leukemia using increased dose of imatinib and deintensified chemotherapy before allogeneic stem cell transplantation. Br J Haematol. 2012;159(1):78–81. doi: 10.1111/j.1365-2141.2012.09240.x.

  55. Kebriaie P, Saliba R, Rondon G, et al. Long-term follow-up of allogeneic hematopoietic stem cell transplantation for patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: impact of tyrosine kinase inhibitors on treatment outcomes. Biol Blood Marrow Transplant. 2012;18(4):584–92. doi: 10.1016/j.bbmt.2011.08.011.

  56. Aldoss I, Stiller T, Cao TM, et al. Impact of additional cytogenetic abnormalities in adults with Philadelphia chromosome-positive acute lymphoblastic leukemia undergoing allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2015;21(7):1326–9. doi: 10.1016/j.bbmt.2015.03.021.

  57. Chiaretti S, Foa R. Management of adult Ph‐positive acute lymphoblastic leukemia. 2015;2015(1):406–13. doi: 10.1182/asheducation‐2015.1.406.

  58. Giebel S, Labopin M, Gorin NC, et al. Improving results of autologous stem cell transplantation for Philadelphia-positive acute lymphoblastic leukemia in the era of tyrosine kinase inhibitors: A report from the Acute Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Eur J Cancer. 2014;50(2):411–7. doi: 10.1016/j.ejca.2013.08.027.

  59. Giebel S, Labopin M, Potter M, et al. Comparable results of autologous and allogeneic haematopoietic stem cell transplantation for adults with Philadelphia-positive acute lymphoblastic leukaemia in first complete molecular remission: An analysis by the Acute Leukemia Working Party of the EBMT. Eur J Cancer. 2018;96:73–81. doi: 10.1016/j.ejca.2018.03.018.

  60. Gindina TL, Mamaev NN, Nikolaeva ES, et al. Results of allogeneic hematopoietic stem cell transplantation in a mixed cohort of patients with Ph-positive acute lymphoblastic leukemia. Cellular Therapy and Transplantation. 2017;6(1):10–9. doi: 10.18620/ctt-1866-8836-2017-6-1-10-19.

  61. Maino E, Scattolin AM, Viero P, et al. Modern immunotherapy of adult B-lineage acute lymphoblastic leukemia with monoclonal antibodies and chimeric antigen receptor modified T cells. Mediterr J Hematol Infect Dis. 2015;7(1):e2015001. doi: 10.4884/mjhid.2015001.

  62. Zhang J-P, Zhang R, Tsao Sh-T, et al. Sequential allogeneic and autologous CAR-T–cell therapy to treat an immune-compromised leukemic patient. Blood Adv. 2018;2(14):1691–5. doi: 10.1182/bloodadvances.2018017004.

  63. Ziser R. Introduction to a review series on strategies to improve GVL effects. Blood. 2018;131(10):1039. doi: 10.1182/blood-2017-11-814467.

Current Issues in Carcinogenesis

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

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

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

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

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

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


ABSTRACT

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

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

Received: June 27, 2018

Accepted: December 20, 2018

Read in PDF 


REFERENCES

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

Clinical and Prognostic Value of Molecular Markers of Diffuse Large B-Cell Lymphoma

SM Rastorguev1, DA Koroleva2, ES Boulygina1, SV Tsygankova1, NG Goncharov1, OS Naraikin1, NG Gabeeva2, EE Zvonkov2, AV Nedoluzhko1

1 National Research Center “Kurchatov Institute”, 1 Akademika Kurchatova sq., Moscow, Russian Federation, 123182

2 National Medical Hematology Research Center, 4а Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167

For correspondence: Artem Valer’evich Nedoluzhko, PhD in Biology, 1 Akademika Kurchatova sq., Moscow, Russian Federation, 123182; Tel.: +7(916)670-55-95; e-mail: nedoluzhko@gmail.com

For citation: Rastorguev SM, Koroleva DA, Bulygina ES, et al. Clinical and Prognostic Value of Molecular Markers of Diffuse Large B-Cell Lymphoma. Clinical oncohematology. 2019:12(1):95–100.

DOI: 10.21320/2500-2139-2019-12-1-95-100


ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid tumor in adults which is associated with approximately 30–40 % of all non-Hodgkin’s lymphomas. Diagnostic criteria include diffuse growth of large anaplastic tumor cells, expression of В-cell markers, and a high proliferative index. Due to the development of molecular genetic technologies it became obvious that underlying cause of clinical diversity is a huge amount of genetic failures which determine epigenetic modification of gene expression, activation variability of certain signaling pathways, and immunological properties of tumor cells. The study and systemization of molecular markers present a significant trend in DLBCL diagnosis and treatment. This review discusses most important molecular markers and current view on their clinical value.

Keywords: lymphoma, DLBCL, B-cells, transcriptomics, gene expression, epigenomics, genomics.

Received: July 3, 2018

Accepted: December 10, 2018

Read in PDF 


REFERENCES

  1. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503–11. doi: 10.1038/35000501.

  2. Rosenwald A, Alizadeh AA, Widhopf G, et al. Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med. 2001;194(11):1639–48. doi: 1084/jem.194.11.1639.

  3. Staudt LM, Dave S. The biology of human lymphoid malignancies revealed by gene expression profiling. Adv Immunol. 2005;87:163–208. doi: 10.1016/S0065-2776(05)87005-1.

  4. Звонков Е.Е., Морозова А.К., Кравченко С.К. и др. Восьмилетний опыт применения модифицированной программы NHL-BFM-90 в лечении взрослых больных первичной диффузной В-крупноклеточной лимфомой желудка. Гематология и трансфузиология. 2012;57(3):47–8.

    [Zvonkov EE, Morozova AK, Kravchenko SK, et al. Eight-year experience of using the modified NHL-BFM-90 program for treatment of adult patients with primary diffuse large B-cell gastric lymphoma. Gematologiya i transfuziologiya. 2012;57(3):47–8. (In Russ)]

  5. Магомедова А.У., Кравченко С.К., Кременецкая A.M. и др. Девятилетний опыт лечения больных диффузной В-крупноклеточной лимфосаркомой. Терапевтический архив. 2011;83(7):5–10.

    [Magomedova AU, Kravchenko SK, Kremenetskaya AM, et al. Nine-year experience in the treatment of patients with diffuse large B-cell lymphosarcoma. Terapevticheskii arkhiv. 2011;83(7):5–10. (In Russ)]

  6. Гаврилина О.А., Габеева Н.Г., Морозова А.К. и др. Роль высокодозной химиотерапии и трансплантации аутологичных стволовых клеток крови у пациентов с диффузной В-крупноклеточной лимфомой. Терапевтический архив. 2013;85(7):90–7.

    [Gavrilina OA, Gabeeva NG, Morozova AK, et al. Role of high-dose chemotherapy and autologous blood stem cell transplantation in patients with diffuse large B-cell lymphoma. Terapevticheskii arkhiv. 2013;85(7):90–7. (In Russ)]

  7. Габеева Н.Г., Королева Д.А., Беляева А.В. и др. Диффузная В-крупноклеточная лимфома с сочетанной реаранжировкой генов c-MYC и BCL6 с первичным поражением кожи: собственное наблюдение и обзор литературы. Терапевтический архив. 2017;89(7):85–92.

    [Gabeeva NG, Koroleva DA, Belyaeva AV, et al. Diffuse large B-cell lymphoma with concomitant c-MYC and BCL6 gene rearrangements with primary skin involvement: A case report and a review of literature. Terapevticheskii arkhiv. 2017;89(7):85–92. (In Russ)]

  8. Martelli M, Ferreri AJ, Agostinelli C, et al. Diffuse large B-cell lymphoma. Crit Rev Oncol Hematol. 2013;87(2):146–71. doi: 10.1016/j.critrevonc.2012.12.009.

  9. Cohen M, Vistarop AG, Huaman F, et al. Epstein-Barr virus lytic cycle involvement in diffuse large B cell lymphoma. Hematol Oncol. 2017;36(1):98–103. doi: 10.1002/hon.2465.

  10. Lenz G, Wright G, Dave SS, et al. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med. 2008;359(22):2313–23. doi: 10.1056/NEJMoa0802885.

  11. Wright G, Tan B, Rosenwald A, et al. A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc Natl Acad Sci USA. 2003;100(17): 9991–6. doi: 10.1073/pnas.1732008100.

  12. Skryabin KG, Prokhortchouk EB, Mazur AM, et al. Combining Two Technologies for Full Genome Sequencing of Human. Acta Nat. 2009;1(3):102–7.

  13. Artemov AV, Boulygina ES, Tsygankova SV, et al. Study of Alzheimer Family Case Reveals Hemochromotosis-Associated HFE Mutation. Hum Gen Var. 2014;1(1):14004. doi: 10.1038/hgv.2014.4.

  14. Scelo G, Riazalhosseini Y, Greger L, et al. Variation in genomic landscape of clear cell renal cell carcinoma across Europe. Nat Commun. 2014;5(1):5135. doi: 10.1038/ncomms6135.

  15. Shipp MA, Ross KN, Tamayo P, et al. Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat Med. 2002;8(1):68–74. doi: 10.1038/nm0102-68.

  16. 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.

  17. Lawrie CH, Soneji S, Marafioti T, et al. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int J Cancer. 2007;121(5):1156–61. doi: 10.1002/ijc.22800.

  18. Malumbres R, Sarosiek KA, Cubedo E, et al. Differentiation stage–specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas. Blood. 2009;113(16):3754–64. doi: 10.1182/blood-2008-10-184077.

  19. Zhu D, Fang C, Li X, et al. Predictive analysis of long non-coding RNA expression profiles in diffuse large B-cell lymphoma. Oncotarget. 2017;8(14):23228–36. doi: 10.18632/oncotarget.15571.

  20. Peng W, Fan H, Wu G, et al. Upregulation of long noncoding RNA PEG10 associates with poor prognosis in diffuse large B cell lymphoma with facilitating tumorigenicity. Clin Exp Med. 2016;16(2):177–82. doi: 10.1007/s10238-015-0350-9.

  21. Peng W, Feng J. Long noncoding RNA LUNAR1 associates with cell proliferation and predicts a poor prognosis in diffuse large B-cell lymphoma. Biomed Pharmacother. 2016;77:65–71. doi: 10.1016/j.biopha.2015.12.001.

  22. Peng W, Wu J, Feng J. Long noncoding RNA HULC predicts poor clinical outcome and represents pro-oncogenic activity in diffuse large B-cell lymphoma. Biomed Pharmacother. 2016;79:188–93. doi: 10.1016/j.biopha.2016.02.032.

  23. Yan Y, Han J, Li Z, et al. Elevated RNA expression of long non-coding HOTAIR promotes cell proliferation and predicts a poor prognosis in patients with diffuse large B cell lymphoma. Mol Med Rep. 2016;13(6):5125–31. doi: 10.3892/mmr.2016.5190.

  24. Li L-J, Chai Y, Guo X-J, et al. The effects of the long non-coding RNA MALAT-1 regulated autophagy-related signaling pathway on chemotherapy resistance in diffuse large B-cell lymphoma. Biomed Pharmacother. 2017;89:939–48. doi: 10.1016/j.biopha.2017.02.011.

  25. Sun J, Cheng L, Shi H, et al. A potential panel of six-long non-coding RNA signature to improve survival prediction of diffuse large-B-cell lymphoma. Sci Rep. 2016;6(1):27842. doi: 10.1038/srep27842.

  26. Verma A, Jiang Y, Du W, et al. Transcriptome sequencing reveals thousands of novel long non-coding RNAs in B cell lymphoma. Gen Med. 2015;7(1):110. doi: 10.1186/s13073-015-0230-7.

  27. Gutierrez-Garcia G, Cardesa-Salzmann T, Climent F, et al. Gene-expression profiling and not immunophenotypic algorithms predicts prognosis in patients with diffuse large B-cell lymphoma treated with immunochemotherapy. Blood. 2011;117(18):4836–43. doi: 10.1182/blood-2010-12-322362.

  28. Schuetz JM, Johnson NA, Morin RD, et al. BCL2 mutations in diffuse large B-cell lymphoma. Leukemia. 2012;26(6):1383–90. doi: 10.1038/leu.2011.378.

  29. Greenough A, Moffitt A, Jima D, et al. Strand-Specific Total RNA Sequencing Establishes the Complete Transcriptome and Alternative Splicing Repertoire in Diffuse Large B Cell Lymphoma. Blood. 2014;124(21):864.

  30. Park HY, Lee SB, Yoo HY, et al. Whole-Exome and Transcriptome Sequencing of Refractory Diffuse Large B-Cell Lymphoma. Oncotarget. 2016;7(52): 86433–45. doi: 10.18632/oncotarget.13239.

  31. Dekker JD, Park D, Shaffer AL, et al. Subtype-Specific Addiction of the Activated B-Cell Subset of Diffuse Large B-Cell Lymphoma to FOXP1. Proc Natl Acad Sci USA. 2016;113(5):E577–86. doi: 10.1073/pnas.1524677113.

  32. Reddy A, Zhang J, Davis NS, et al. Genetic and Functional Drivers of Diffuse Large B Cell Lymphoma. Cell. 2017;171(2):481–94.e15. doi: 10.1016/j.cell.2017.09.027.

  33. Saez AI, Saez AJ, Artiga MJ, et al. Building an outcome predictor model for diffuse large B-cell lymphoma. Am J Pathol. 2004;164(2):613–22. doi: 10.1016/S0002-9440(10)63150-1.

  34. Campo E. MYC in DLBCL: partners matter. Blood. 2015;126(22):2439–40. doi: 10.1182/blood-2015-10-671362.

  35. Schmitz R, Wright GW, Huang DW, et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N Engl J Med. 2018;378(15):1396–407. doi: 10.1056/NEJMoa1801445.

  36. Dubois S, Viailly PJ, Mareschal S, et al. Next-generation sequencing in diffuse large B-cell lymphoma highlights molecular divergence and therapeutic opportunities: a LYSA study. Clin Cancer Res. 2016;22(12):2919–28. doi: 10.1158/1078-0432.CCR-15-2305.

  37. Lohr JG, Stojanov P, Lawrence MS, et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc Natl Acad Sci USA. 2012;109(10):3879–84. doi: 10.1073/pnas.1121343109.

  38. Morin RD, Mungall K, Pleasance E, et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood. 2013;122(7):1256–65. doi: 10.1182/blood-2013-02-483727.

  39. Pasqualucci L, Trifonov V, Fabbri G, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet. 2011;43(9):830–7. doi: 10.1038/ng.892.

  40. 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.

  41. Yeh P, Hunter T, Sinha D, Ftouni S, et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nat Commun. 2017;8:14756. doi: 10.1038/ncomms14756.

  42. Khare D, Goldschmidt N, Bardugo A, et al. Plasma microRNA profiling: Exploring better biomarkers for lymphoma surveillance. PLoS One. 2017;12(11):e0187722. doi: 10.1371/journal.pone.0187722.

  43. Meng Y, Quan L, Liu A. Identification of key microRNAs associated with diffuse large B-cell lymphoma by analyzing serum microRNA expressions. Gene. 2018;642:205–11. doi: 10.1016/j.gene.2017.11.022.

  44. 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.

  45. Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359(6378):926–30. doi: 10.1126/science.aar3247.

  46. Shaknovich R, Melnick A. Epigenetics and B-Cell Lymphoma. Curr Opin Hematol. 2011;18(4):293–9. doi: 10.1097/MOH.0b013e32834788cf.

  47. Shaknovich R, Geng H, Johnson NA, et al. DNA methylation signatures define molecular subtypes of diffuse large B-cell lymphoma. Blood. 2010;116(20):e81–9. doi: 10.1182/blood-2010-05-285320.

  48. Lai AY, Fatemi M, Dhasarathy A, et al. DNA methylation prevents CTCF-mediated silencing of the oncogene BCL6 in B cell lymphomas. J Exp Med. 2010;207(9):1939–50. doi: 10.1084/jem.20100204.

  49. 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 Epigen. 2016;8(1):5. doi: 10.1186/s13148-016-0261-y.

  50. Wedge E, Hansen JW, Garde C, et al. Global hypomethylation is an independent prognostic factor in diffuse large B cell lymphoma. Am J Hematol. 2017;92(7):689–94. doi: 10.1002/ajh.24751.

  51. Krajnovic M, Jovanovic MP, Mihaljevic B, et al. Hypermethylation of p15 Gene in Diffuse – Large B‐Cell Lymphoma: Association with Less Aggressiveness of the Disease. Clin Transl Sci. 2014;7(5):384–90. doi: 10.1111/cts.12162.

  52. Chambwe N, Kormaksson M, Geng H, et al. Variability in DNA methylation defines novel epigenetic subgroups of DLBCL associated with different clinical outcomes. Blood. 2014;123(11):1699–708. doi: 10.1182/blood-2013-07-509885.

  53. Clozel T, Yang S, Elstrom RL, et al. Mechanism-based epigenetic chemosensitization therapy of diffuse large B-cell lymphoma. Cancer Discov. 2013;3(9):1002–19. doi: 10.1158/2159-8290.CD-13-0117.

  54. Pan H, Jiang Y, Boi M, et al. Epigenomic evolution in diffuse large B-cell lymphomas. Nat Commun. 2015;6(1):6921. doi: 10.1038/ncomms7921.

  55. Jing L, Su L, Ring BZ. Ethnic Background and Genetic Variation in the Evaluation of Cancer Risk: A Systematic Review. PLoS ONE. 2014;9(6):e97522. doi: 10.1371/journal.pone.0097522.

  56. Li Y, Wang Y, Wang Z, et al. Racial Differences in Three Major NHL Subtypes: Descriptive Epidemiology. Cancer Epidemiol. 2015;39(1):8–13. doi: 10.1016/j.canep.2014.12.001.

 

Immunological Synapse in the Biology of Chronic Lymphocytic Leukemia

DS Badmazhapova, IV Gal’tseva, EE Zvonkov

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

For correspondence: Darima Semunkoevna Badmazhapova, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(929)562-93-41; e-mail: badmazhapova-darima@mail.ru

For citation: Badmazhapova DS, Gal’tseva IV, Zvonkov EE. Immunological Synapse in the Biology of Chronic Lymphocytic Leukemia. Clinical oncohematology. 2018;11(4):313–8.

DOI: 10.21320/2500-2139-2018-11-4-313-318


ABSTRACT

Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disease manifested by accumulation of tumor B-cells with characteristic immunophenotype (CD19+CD5+CD23+) in bone marrow, peripheral blood and secondary lymphoid organs. The clinical course of CLL is heterogeneous. This is the most prevalent leukemia among older-aged patients. Despite the use of novel drugs refractory forms of disease remain. The latest discoveries in immunology enabled understanding of some mechanisms of tumor evasion from immune surveillance. The interaction of immune system cells occurs due to the development of immunological synapse that predominantly depends on the family of CD28/В7 molecules, the so-called immune checkpoints able to control the activating and inhibiting mechanisms of cells. The acquisition of tumor phenotype is a multistage process, in which cells obtain unique biological properties including the ability of being invisible to the immune system. As opposed to solid tumors in lymphoproliferative diseases tumor B-cells are able to express major histocompatibility complex class II and CD80 and CD86 co-stimulatory molecules. It proves their ability to present antigens to T-cells. Co-inhibitory molecules on the surface of tumor cells is a factor contributing to the inhibition of immune response. The present paper reviews current conceptions of biological properties and immunological interactions of CLL cells with the microenvironmental cells.

Keywords: chronic lymphocytic leukemia, immunological synapse, immune system.

Received: March 15, 2018

Accepted: June 29, 2018

Read in PDF 


REFERENCES

  1. Hallek M, Cheson BD, Catovsky D, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood. 2008;111(12):5446–56. doi: 10.1182/blood-2007-06-093906.

  2. Eichhorst B, Robak T, Montserrat E, et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v78–v84. doi: 10.1093/annonc/mdv303.

  3. 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.

  4. Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3(11):991–8. doi: 10.1038/ni1102-991.

  5. Mellor AL, Munn DH. Tryptophan catabolism and regulation of adaptive immunity. J Immunol. 2003;170(12):5809–13. doi: 4049/jimmunol.170.12.5809.

  6. Vladimirova R, Popova D, Vikentieva E, et al. Chronic Lymphocytic Leukemia — Microenvironment and B Cells. In: Guenova M, Balatzenko G, eds. Leukemias: Updates and New Insights [Internet]; 2015. рр. 247–76. doi: 10.5772/60761. Available from: https://www.intechopen.com/books/leukemias-updates-and-new-insights/chronic-lymphocytic-leukemia-microenvironment-and-b-cells (accessed 31.05.2018).

  7. Ярилин А.А. Иммунология: учебник. M.: ГЭОТАР-Медиа, 2010. С. 394–403.

    [Yarilin AA. Immunologiya: uchebnik. (Immunology: a manual.) Moscow: GEOTAR-Media Publ.; 2010. pp. 394–403. (In Russ)]

  8. Kupfer A, Kupfer H. Imaging immune cell interactions and functions: SMACs and the immunological synapse. Semin Immunol. 2003;15(6):295–300. doi: 10.1016/j.smim.2003.09.001.

  9. Dustin ML. Modular design of immunological synapses and kinapses. Cold Spring Harb Perspect Biol. 2009;1(1):a002873. doi: 10.1101/cshperspect.a002873.

  10. Janeway C, Travers P, Walport M, et al. Immunobiology. The immune system in health and disease, 6th edn. Garland Science; 2005.

  11. Burger JA. Nurture versus nature: the microenvironment in chronic lymphocytic leukemia. Hematology Am Soc Hematol Educ Program. 2011;1:96–103. doi: 10.1182/asheducation-2011.1.96.

  12. Pasikowska M, Walsby E, Apollonio B, et al. Phenotype and immune function of lymph node and peripheral blood CLL cells are linked to transendothelial migration. 2016;128(4):563–73. doi: 10.1182/blood-2016-01-683128.

  13. Hofbauer JP, Heyder C, Denk U, et al. Development of CLL in the TCL1 transgenic mouse model is associated with severe skewing of the T-cell compartment homologous to human CLL. Leukemia. 2011;25(9):1452–8. doi: 10.1038/leu.2011.111.

  14. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23(1):515–48. doi:1146/annurev.immunol.23.021704.115611.

  15. Sansom DM. CD28, CTLA-4 and their ligands: who does what and to whom? 2000;101(2):169–77. doi: 10.1046/j.1365-2567.2000.00121.x.

  16. Boussiotis VA, Freeman GJ, Gribben GJ, et al. The role of B7-1/B7-2:CD28/CTLA-4 pathways in the prevention of anergy, induction of productive immunity and downregulated of the immune response. Immunol Rev. 1996;153(1):5–26. doi: 10.1111/j.1600-065x.1996.tb00918.x.

  17. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209(6):1201–17. doi: 10.1084/jem.20112741.

  18. Sheppard KA, Fitz LJ, Lee JM, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3 zeta signalosome and downstream signaling to PKC theta. FEBS Lett. 2004;574(1–3):37–41. doi: 10.1016/j.febslet.2004.07.083.

  19. Thibult M-L, Mamessier E, Gertner-Dardenne J, et al. PD-1 is a novel regulator of human B-cell activation. Int Immunol. 2013;25(2):129–37. doi: 10.1093/intimm/dxs098.

  20. Wang K, Wei G, Liu D. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Exp Hematol Oncol. 2012;1(1):36. doi: 10.1186/2162-3619-1-36.

  21. Mills DM, Stolpa JC, Cambier JC. Modulation of MHC class II signal transduction by CD19. Adv Exp Med Biol. 2007;596:139–48. doi: 1007/0-387-46530-8_12.

  22. Kuijpers TW, Bende RJ, Baars PA, et al. CD20 deficiency in humans results in impaired T cell-independent antibody responses. J Clin Invest. 2010;120(1):214–22. doi: 1172/JCI40231.

  23. Nitschke L. CD22 and Siglec-G: B-cell inhibitory receptors with distinct functions. Immunol Rev. 2009;230(1):128–43. doi: 1111/j.1600-065X.2009.00801.x.

  24. Cerutti A, Kim EC, Shah S, et al. Dysregulation of CD30+ T cells by leukemia impairs isotype switching in normal B cells. Nat Immunol. 2001;2(2):150–6. doi: 10.1038/84254.

  25. Agata Y, Kawasaki A, Nishimura H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8(5):765–72. doi: 10.1093/intimm/8.5.765.

  26. Chinai JM, Janakiram M, Chen F, et al. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol Sci. 2015;36(9):587–95. doi: 10.1016/j.tips.2015.06.005.

  27. Majolini MB, D’Elios MM, Galieni P, et al. Expression of the T-cell-specific tyrosine kinase Lck in normal B-1 cells and in chronic lymphocytic leukemia B cells. Blood. 1998;91(9):3390–6.

  28. Ramsay AG, Johnson AJ, Lee AM, et al. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest. 2008;118(7):2427–37. doi: 10.1172/JCI35017.

  29. Caligaris-Cappio F, Bertilaccio MT, Scielzo C. How the microenvironment wires the natural history of chronic lymphocytic leukemia. Semin Cancer Biol. 2014;24:43–8. doi: 10.1016/j.semcancer.2013.06.010.

  30. Damle RN, Calissano C, Chiorazzi N. Chronic lymphocytic leukaemia: a disease of activated monoclonal B cells. Clin Haematol. 2010;23(1):33–45. doi: 10.1016/j.beha.2010.02.001.

  31. Lauria F, Foa R, Catovsky D. Increase in T gamma lymphocytes in B-cell chronic lymphocytic leukaemia. Scand J Haematol. 1980;24(2):187–90. doi:1111/j.1600-0609.1980.tb02366.x.

  32. Herrmann F, Lochner A, Philippen H, et al. Imbalance of T cell subpopulations in patients with chronic lymphocytic leukaemia of the B cell type. Clin Exp Immunol. 1982;49(1):157–62.

  33. Mills KH, Worman CP, Cawley JC. T-cell subsets in B-chronic lymphocytic leukaemia (CLL). Br J Haematol. 1982;50(4):710–2. doi:1111/j.1365-2141.1982.tb01974.x.

  34. Platsoucas CD, Galinski M, Kempin S, et al. Abnormal T lymphocyte subpopulations in patients with B cell chronic lymphocytic leukemia: an analysis by monoclonal antibodies. J Immunol. 1982;129(5):2305–12.

  35. Pizzolo G, Chilosi M, Ambrosetti A, et al. Immunohistologic study of bone marrow involvement in B-chronic lymphocytic leukemia. Blood. 1983;62(6):1289–96.

  36. Ghia P, Strola G, Granziero L, et al. Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22. Eur J Immunol. 2002;32(5):1403–13. doi: 10.1002/1521-4141(200205)32:5<1403::aid-immu1403>3.0.co;2-y.

  37. Bagnara D, Kaufman MS, Calissano C, et al. A novel adoptive transfer model of chronic lymphocytic leukemia suggests a key role for T lymphocytes in the disease. Blood. 2011;117(20):5463–72. doi: 10.1182/blood-2010-12-324210.

  38. Qorraj M, Bottcher M, Mougiakakos D. PD-L1/PD-1: new kid on the “immune metabolic” block. Oncotarget. 2017;8(43):73364–5. doi: 10.18632/oncotarget.20639.

  39. Burger JA, Tsukada N, Burger M, et al. Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1. Blood. 2000;96(8):2655–63.

  40. Tsukada N, Burger JA, Zvaifler NJ, Kipps TJ. Distinctive features of “nurselike” cells that differentiate in the context of chronic lymphocytic leukemia. Blood. 2002;99(3):1030–7. doi: 10.1182/blood.V99.3.1030.

  41. Schiemann B, Gommerman JL, Vora K, et al. An essential role for BAFF in the normal development of B cells through a BCMA-independent pathway. Science. 2001;293(5537):2111–4. doi: 1126/science.1061964.

  42. Schneider P, Takatsuka H, Wilson A, et al. Maturation of marginal zone and follicular B cells requires B cell activating factor of the tumor necrosis factor family and is independent of B cell maturation antigen. J Exp Med. 2001;194(11):1691–7. doi: 10.1084/jem.194.11.1691.

  43. Mackay F, Schneider P, Rennert P, et al. BAFF and APRIL: a tutorial on B cell survival. Annu Rev Immunol. 2003;21(1):231–64. doi: 1146/annurev.immunol.21.120601.141152.

  44. Walton JA, Lydyard PM, Nathwani A, et al. Patients with B cell chronic lymphocytic leukaemia have an expanded population of CD4 perforin expressing T cells enriched for human cytomegalovirus specificity and an effector-memory phenotype. Br J Haematol. 2010;148(2):274–84. doi: 10.1111/j.1365-2141.2009.07964.x.

  45. Nunes C, Wong R, Mason M, et al. Expansion of a CD8(+) PD-1(+) replicative senescence phenotype in early stage CLL patients is associated with inverted CD4:CD8 ratios and disease progression. Clin Cancer Res. 2012;18(3):678–87. doi: 10.1158/1078-0432.CCR-11-2630.

  46. Brown JA, Dorfman DM, Ma FR, et al. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol. 2003;170(3):1257–66. doi: 10.4049/jimmunol.170.3.1257.

  47. Ramsay AG, Clear AJ, Fatah R, et al. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood. 2012;120(7):1412–21. doi: 10.1182/blood-2012-02-411678.

  48. Grzywnowicz M, Karabon L, Karczmarczyk A, et al. The function of a novel immunophenotype candidate molecule PD-1 in chronic lymphocytic leukemia. Leuk Lymphoma. 2015;56(10):2908–13. doi: 10.3109/10428194.2015.1017820.

  49. Li J, Pang N, Zhang Z, et al. PD-1/PD-L1 expression and its implications in patients with chronic lymphocytic leukemia. Zhonghua Xue Ye Xue Za Zhi. 2017;38(03):198–203. doi: 10.3760/cma.j.issn.0253-2727.2017.03.005.

  50. Brusa D, Serra S, Coscia M, et al. The PD-1/PD-L1 axis contributes to T-cell dysfunction in chronic lymphocytic leukemia. Haematologica. 2013;98(6):953–63. doi: 10.3324/haematol.2012.077537.

  51. Xerri L, Chetaille B, Seriari N, et al. Programmed death 1 is a marker of angioimmunoblastic T-cell lymphoma and B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia. Hum Pathol. 2008;39(7):1050–8. doi: 10.1016/j.humpath.2007.11.012.

  52. Panayiotidis P, Jones D, Ganeshaguru K, et al. Human bone marrow stromal cells prevent apoptosis and support the survival of chronic lymphocytic leukaemia cells in vitro. Br J Haematol. 1996;92(1):97–103. doi: 10.1046/j.1365-2141.1996.00305.x.

  53. Burger M, Hartmann T, Krome M, et al. Small peptide inhibitors of the CXCR4 chemokine receptor (CD184) antagonize the activation, migration and antiapoptotic responses of CXCL12 in chronic lymphocytic leukemia B cells. Blood. 2005;106(5):1824–30. doi: 10.1182/blood-2004-12-4918.

Immune Checkpoint Inhibitors in the Treatment of Lymphomas

KV Lepik

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

For correspondence: Kirill Viktorovich Lepik, 6/8 L’va Tolstogo str., Saint Petersburg, Russian Federation, 197022; e-mail: lepikkv@gmail.com

For citation: Lepik KV. Immune Checkpoint Inhibitors in the Treatment of Lymphomas. Clinical oncohematology. 2018;11(4):303–12.

DOI: 10.21320/2500-2139-2018-11-4-303-312


ABSTRACT

Programmed death receptors and ligands (PD-1 and PD-L1) are the best studied immune checkpoints (ICP) and are considered to be key factors of immune response control. The ability of tumor cells to affect the ICP receptors is one of the principal mechanisms of suppressing antitumor immunity. The development of ICP inhibitors creates an opportunity to control and activate immune response and opens new perspectives for immunotherapy of cancers, including lymphomas. The paper reviews the biological background for the use of ICP inhibitors in the treatment of classical Hodgkin’s and non-Hodgkin’s lymphomas and summarizes the clinical experience of their use. The new approaches for the creation of combination regimens with ICP are also highlighted.

Keywords: immune checkpoints (ICP), PD-1, PD-L1, classical Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, ICP inhibitors.

Received: March 25, 2018

Accepted: July 23, 2018

Read in PDF 


REFERENCES

  1. Walunas TL, Bakker CY, Bluestone JA. CTLA 4 ligation blocks CD28 dependent T cell activation. J Exp Med. 1996;183(6):2541–50.

  2. Freeman GJ, Long AJ, Iwai Y. Engagement of the Pd-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192(7):1027–34. doi: 1084/jem.192.7.1027.

  3. Greaves P, Gribben JG. The role of B7 family molecules in hematologic malignancy. Blood. 2013;121(5):734–44. doi: 1182/blood-2012-10-385591.

  4. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311–9. doi: 1056/NEJMoa1411087.

  5. Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 2008;26(1):677–704. doi: 1146/annurev.immunol.26.021607.090331.

  6. Lee SJ, Jang BC, Lee SW, et al. Interferon regulatory factor-1 is prerequisite to the constitutive expression and IFN-gamma-induced upregulation of B7-H1 (CD274). FEBS Lett. 2006;580(3):755–62. doi: 1016/j.febslet.2005.12.093.

  7. Liu J, Hamrouni A, Wolowiec D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-gamma and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood. 2007;110(1):296–304. doi: 1182/blood-2006-10-051482.

  8. Fife BT, Pauken KE, Eagar TN, et al. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol. 2009;10(11):1185–92. doi: 1038/ni.1790.

  9. Yokosuka T, Takamatsu M, Kobayashi-Imanishiet W, et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209(6):1201–17. doi: 1084/jem.20112741.

  10. Chemnitz JM, Parry RV, Nicholset KE, et al. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173(2):945–54. doi: 4049/jimmunol.173.2.945.

  11. Nurieva R, Thomas S, Nguyen T, et al. T-cell tolerance or function is determined by combinatorial costimulatory signals. EMBO J. 2006;25(11):2623–33. doi: 1038/sj.emboj.7601146.

  12. Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7 H1 promotes T cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8(8):793–800. doi: 1038/nm730.

  13. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23. doi: 1056/NEJMoa1003466.

  14. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–30. doi: 10.1056/NEJMoa1412082.

  15. Weber JS, D’Angelo SP, Minor D, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA 4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–84. doi: 1016/S1470-2045(15)70076-8.

  16. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–30. doi: 1200/JCO.2013.53.0105.

  17. 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.

  18. 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.

  19. Kuppers R. The biology of Hodgkin’s lymphoma. Nat Rev Cancer. 2009;9(1):15–27. doi: 10.1038/nrc2542.

  20. 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.

  21. Chen BJ, Chapuy B, Ouyang J, et al. PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin Cancer Res. 2013;19(13):3462–73. doi: 10.1158/1078-0432.CCR-13-0855.

  22. 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.

  23. 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.

  24. Paydas S, Bagir E, Seydaoglu G, et al. Programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1), and EBV-encoded RNA (EBER) expression in Hodgkin lymphoma. Ann Hematol. 2015;94(9):1545–52. doi: 10.1007/s00277-015-2403-2.

  25. Hollander P, Kamper P, Smedby KE, et al. High proportions of PD-1+ and PD-L1+ leukocytes in classical Hodgkin lymphoma microenvironment are associated with inferior outcome. Blood Adv. 2017;1(18):1427–39. doi: 10.1182/bloodadvances.2017006346.

  26. Younes A, Santoro A, Shipp M, et al. Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol. 2016;17(9):1283–94. doi: 10.1016/S1470-2045(16)30167-X.

  27. Armand P, Shipp MA, Ribrag V, et al. Pembrolizumab in Patients with Classical Hodgkin Lymphoma after Brentuximab Vedotin Failure: Long-Term Efficacy from the Phase 1b Keynote-013 Study. Blood. 2016;128:1108, abstract.

  28. 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.

  29. Engert A, Fanale M, Santoro A, et al. Nivolumab for relapsed/refractory classical Hodgkin lymphoma after autologous transplant: full results after extended follow-up of the multicohort multicenter phase 2 CheckMate 205 trial. EHA conference 2017. Abstract S412.

  30. Armand P, Shipp MA, Ribrag V, et al. Programmed Death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab vedotin failure. J Clin Oncol. 2016; 34(31):3733–9. doi: 10.1200/JCO.2016.67.3467.

  31. Chen R, Zinzani PL, Fanale MA, et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J Clin Oncol. 2017;35(19):2125–32. doi: 10.1200/JCO.2016.72.1316.

  32. Tsimberidou AM, Braiteh F, Stewart DJ, Kurzrock R. Ultimate fate of oncology drugs approved by the US Food and Drug Administration without a randomized trial. J Clin Oncol. 2009;27(36):6243–50. doi: 10.1200/JCO.2009.23.6018.

  33. Nishijima TF, Shachar SS, Nyrop KA, Muss HB. Safety and tolerability of PD-1/PD-L1 inhibitors compared with chemotherapy in patients with advanced cancer: a meta-analysis. Oncologist. 2017;22(4):470–9. doi: 10.1634/theoncologist.2016-0419.

  34. Shi M, Roemer MGM, Chapuy B, et al. Expression of programmed cell death 1 ligand 2 (PD-L2) is a distinguishing feature of primary mediastinal (thymic) large B-cell lymphoma and associated with PDCD1LG2 copy gain. Am J Surg Pathol. 2014;38(12):1715–23. doi: 10.1097/PAS.0000000000000297.

  35. Twa DDW, Chan FC, Ben-Neriah S, et al. Genomic rearrangements involving programmed death ligands are recurrent in primary mediastinal large B-cell lymphoma. Blood. 2014;123(13):2062–5. doi: 10.1182/blood-2013-10-535443.

  36. Van Roosbroeck K, Ferreiro JF, Tousseyn T, et al. Genomic alterations of the JAK2 and PDL loci occur in a broad spectrum of lymphoid malignancies. Genes Chromos Cancer. 2016;55(5):428–41. doi: 10.1002/gcc.22345.

  37. Zinzani PL, Ribrag V, Moskowitz CH, et al. Safety and tolerability of pembrolizumab in patients with relapsed/refractory primary mediastinal large B-cell lymphoma. Blood. 2017;130(3):267–70. doi: 10.1182/blood-2016-12-758383.

  38. Zinzani PL, Thieblemont C, Melnichenko V, et al. Efficacy and Safety of Pembrolizumab in Relapsed/Refractory Primary Mediastinal Large B-Cell Lymphoma (rrPMBCL): Updated Analysis of the Keynote-170 Phase 2 Trial. ASH conference 2017. Abstract 2833B.

  39. Chapuy B, Roemer MGM, Stewart C, et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood. 2016;127(7):869–81. doi: 10.1182/blood-2015-10-673236.

  40. Nayak L, Iwamoto FM, LaCasce A, et al. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood. 2017;129(23):3071–3. doi: 10.1182/blood-2017-01-764209.

  41. Eberle FC, Salaverria I, Steidl C, et al. Gray zone lymphoma: chromosomal aberrations with immunophenotypic and clinical correlations. Mod Pathol. 2011;24(12):1586–97. doi: 10.1038/modpathol.2011.116.

  42. Melani C, Major A, Schowinsky J, et al. PD-1 blockade in mediastinal gray-zone lymphoma. N Engl J Med. 2017;377(1):89–91. doi: 10.1056/NEJMc1704767.

  43. Georgiou K, Chen L, Berglund M, et al. Genetic basis of PD-L1 overexpression in diffuse large B-cell lymphomas. Blood. 2016;127(24):3026–34. doi: 10.1182/blood-2015-12-686550.

  44. Kiyasu J, Miyoshi H, Hirata A, et al. Expression of programmed cell death ligand 1 is associated with poor overall survival in patients with diffuse large B-cell lymphoma. Blood. 2015;126(19):2193–201. doi: 10.1182/blood-2015-02-629600.

  45. Chen M, Andreozzi M, Pockaj B, et al. Development and validation of a novel clinical fluorescence in situ hybridization assay to detect JAK2 and PD-L1 amplification. Mod Pathol. 2017;30(11):1516–26. doi: 10.1038/modpathol.2017.86.

  46. Gupta M, Han JJ, Stenson M, et al. Elevated serum IL-10 levels in diffuse large B-cell lymphoma: a mechanism of aberrant JAK2 activation. Blood. 2012;119(12):2844–53. doi: 10.1182/blood-2011-10-388538.

  47. Choi JW, Kim Y, Lee JH, et al. MYD88 expression and L265P mutation in diffuse large B-cell lymphoma. Hum Pathol. 2013;44(7):1375–81. doi: 10.1016/j.humpath.2012.10.026.

  48. Bellucci R, Martin A, Bommarito D, et al. Interferon-γ-induced activation of JAK1 and JAK2 suppresses tumor cell susceptibility to NK cells through upregulation of PD-L1 expression. OncoImmunology. 2015;4(6):e1008824. doi: 10.1080/2162402X.2015.1008824.

  49. Laurent C, Charmpi K, Gravelle P, et al. Several immune escape patterns in non-Hodgkin’s lymphomas. OncoImmunology. 2015;4(8):e1026530. doi: 10.1080/2162402X.2015.1026530.

  50. Andorsky DJ, Yamada RE, Said J, et al. Programmed death ligand 1 is expressed by non-Hodgkin lymphomas and inhibits the activity of tumor-associated T cells. Clin Cancer Res. 2011;17(13):4232–44. doi: 10.1158/1078-0432.CCR-10-2660.

  51. Jo JC, Kim M, Choi Y, et al. Expression of programmed cell death 1 and programmed cell death ligand 1 in extranodal NK/T-cell lymphoma, nasal type. Ann Hematol. 2017;96(1):25–31. doi: 10.1007/s00277-016-2818-4.

  52. Muenst S, Hoeller S, Willi N, et al. Diagnostic and prognostic utility of PD-1 in B cell lymphomas. Dis Markers. 2010;29(1):47–53. doi: 10.1155/2010/404069.

  53. Hu L-Y, Xu X-L, Rao H-L, et al. Expression and clinical value of programmed cell death-ligand 1 (PD-L1) in diffuse large B cell lymphoma: a retrospective study. Chin J Cancer. 2017;36(1):94. doi: 10.1186/s40880-017-0262-z.

  54. Ansell SM, Hurvitz SA, Koenig PA, et al. Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clin Cancer Res. 2009;15(20):6446–53. doi: 10.1158/1078-0432.CCR-09-1339.

  55. Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J Clin Oncol. 2016;34(23):2698–704. doi: 10.1200/JCO.2015.65.9789.

  56. Armand P, Nagler A, Weller EA, et al. Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. J Clin Oncol. 2013;31(33):4199–206. doi: 10.1200/JCO.2012.48.3685.

  57. Palomba ML, Till BG, Park SI, et al. A phase IB study evaluating the safety and clinical activity of atezolizumab combined with obinutuzumab in patients with relapsed or refractory non-Hodgkin lymphoma (NHL). Hematol Oncol. 2017;35(Suppl 2):137–8. doi: 10.1002/hon.2437_126.

  58. Ansell S, Gutierrez ME, Shipp MA, et al. A phase 1 study of nivolumab in combination with ipilimumab for relapsed or refractory hematologic malignancies (CheckMate 039). Blood. 2016;128;22, abstract 183.

  59. Brusa D, Serra S, Coscia M, et al. The PD-1/PD-L1 axis contributes to T-cell dysfunction in chronic lymphocytic leukemia. Haematologica. 2013;98(6):953–63. doi: 10.3324/haematol.2012.077537.

  60. Soma LA, Craig FE, Swerdlow SH. The proliferation center microenvironment and prognostic markers in chronic lymphocytic leukemia/small lymphocytic lymphoma. Hum Pathol. 2006;37(2):152–9. doi: 10.1016/j.humpath.2005.09.029.

  61. Nunes C, Wong R, Mason M, et al. Expansion of a CD8(+)PD-1(+) replicative senescence phenotype in early stage CLL patients is associated with inverted CD4:CD8 ratios and disease progression. Clin Cancer Res. 2012;18(3):678–87. doi: 10.1158/1078-0432.CCR-11-2630.

  62. Ramsay AG, Johnson AJ, Lee AM, et al. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest. 2008;118(7):2427–37. doi: 10.1172/JCI35017.

  63. Berger R, Rotem-Yehudar R, Slama G, et al. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res. 2008;14(10):3044–51. doi: 10.1158/1078-0432.ccr-07-4079.

  64. Ding W, LaPlant BR, Call TG, et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood. 2017;129(26):3419–27. doi: 10.1182/blood-2017-02-765685.

  65. Panjwani P, Charu V, DeLisser M, et al. Programmed death-1 ligands PD-L1 and PD-L2 show distinctive and restricted patterns of expression in lymphoma subtypes. Hum Pathol. 2018;71:91–9. doi: 10.1016/j.humpath.2017.10.029.

  66. Menter T, Bodmer-Haecki A, Dirnhoferet S, et al. Evaluation of the diagnostic and prognostic value of PDL1 expression in Hodgkin and B-cell lymphomas. Hum Pathol. 2016;54:17–24. doi: 10.1016/j.humpath.2016.03.005.

  67. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;131(6):492–9. doi: 10.1038/ni.2035.

  68. Wahlin BE, Aggarwal M, Montes-Moreno S, et al. A unifying microenvironment model in follicular lymphoma: outcome is predicted by programmed death-1—positive, regulatory, cytotoxic, and helper T cells and macrophages. Clin Cancer Res. 2010;16(2):637–50. doi: 10.1158/1078-0432.CCR-09-2487.

  69. Myklebust JH, Irish JM, Brody J, et al. High PD-1 expression and suppressed cytokine signaling distinguish T cells infiltrating follicular lymphoma tumors from peripheral T cells. Blood. 2013;121(8):1367–76. doi: 10.1182/blood-2012-04-421826.

  70. Smeltzer JP, Jones JM, Ziesmer SC, et al. Pattern of CD14+ follicular dendritic cells and PD1+ T cells independently predicts time to transformation in follicular lymphoma. Clin Cancer Res. 2014;20(11):2862–72. doi: 10.1158/1078-0432.CCR-13-2367.

  71. Carreras J, Lopez-Guillermo A, Roncador G, et al. High numbers of tumor-infiltrating programmed cell death 1-positive regulatory lymphocytes are associated with improved overall survival in follicular lymphoma. J Clin Oncol. 2009;27(9):1470–6. doi: 10.1200/JCO.2008.18.0513.

  72. Richendollar BG, Pohlman B, Elson P, et al. Follicular programmed death 1-positive lymphocytes in the tumor microenvironment are an independent prognostic factor in follicular lymphoma. Hum Pathol. 2011;42(4):552–7. doi: 10.1016/j.humpath.2010.08.015.

  73. Yang ZZ, Grote DM, Ziesmer SC, et al. PD-1 expression defines two distinct T-cell sub-populations in follicular lymphoma that differentially impact patient survival. Blood Cancer J. 2015;5:e281. doi: 10.1038/bcj.2015.1.

  74. Horning SJ, Rosenberg SA. The natural history of initially untreated low-grade non-Hodgkin’s lymphomas. N Engl J Med. 1984;311(23):1471–5. doi: 10.1056/NEJM198412063112303.

  75. Berger R, Rotem-Yehudar R, Slama G, et al. Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res. 2008;14(10):3044–51. doi: 10.1158/1078-0432.CCR-07-4079.

  76. Westin JR, Chu F, Zhang M, et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol. 2014;15(1):69–77. doi: 10.1016/S1470-2045(13)70551-5.

  77. Cheson BD, Leonard JP. Monoclonal antibody therapy for B-cell non-Hodgkin’s lymphoma. N Engl J Med. 2008;359(6):613–26. doi: 10.1056/NEJMra0708875.

  78. Nastoupil LJ, Westin J, Fowler N, et al. High response rates with pembrolizumab in combination with rituximab in patients with relapsed follicular lymphoma: interim results of an on open-label, phase II study. Hematol Oncol. 2017;35(Suppl 2):120–1. doi: 10.1002/hon.2437_108.

  79. Zaja F, Tabanelli V, Agostinelli C. CD38, BCL-2, PD-1, and PD-1L expression in nodal peripheral T-cell lymphoma: Possible biomarkers for novel targeted therapies? Am J Hematol. 2017;92(1):E1–E2. doi: 10.1002/ajh.24571.

  80. Xerri L, Chetaille B, Serriari N. Programmed death 1 is a marker of angioimmunoblastic T-cell lymphoma and B-cell small lymphocytic lymphoma/chronic lymphocytic leukemia. Hum Pathol. 2008;39(7):1050–8. doi: 10.1016/j.humpath.2007.11.012.

  81. Wilcox RA, Feldman AL, Wada DA, et al. B7-H1 (PD-L1, CD274) suppresses host immunity in T-cell lymphoproliferative disorders. Blood. 2009;114(10):2149–58. doi: 10.1182/blood-2009-04-216671.

  82. Vranic S, Ghosh N, Kimbrough J. PD-L1 Status in Refractory Lymphomas. PLoS One. 2016;11(11):e0166266. doi: 10.1371/journal.pone.0166266.

  83. Merryman RW, Armand P, Wright KT, Rodig SJ. Checkpoint blockade in Hodgkin and non-Hodgkin lymphoma. Blood Adv. 2017;1(26):2643–54. doi: 10.1182/bloodadvances.2017012534.

  84. Marzec M, Zhang Q, Goradia A, et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc Natl Acad Sci USA. 2008;105(52):20852–7. doi: 10.1073/pnas.0810958105.

  85. Brown JA, Dorfman DM, Ma FR, et al. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol. 2003;170(3):1257–66. doi: 10.4049/jimmunol.170.3.1257.

  86. Hebart H, Lang P, Woessmann W. Nivolumab for Refractory Anaplastic Large Cell Lymphoma: A Case Report. Ann Intern Med. 2016;165(8):607–8. doi: 10.7326/116-0037.

  87. Cetinozman F, Jansen PM, Willemze R. Expression of programmed death-1 in primary cutaneous CD4-positive small/medium-sized pleomorphic T-cell lymphoma, cutaneous pseudo-T-cell lymphoma, and other types of cutaneous T-cell lymphoma. Am J Surg Pathol. 2012;36(1):109–16. doi: 10.1097/PAS.0b013e318230df87.

  88. Xia Y, Medeiros JL, Young KH. Signaling pathway and dysregulation of PD1 and its ligands in lymphoid malignancies. Biochim Biophys Acta. 2016;1865(1):58–71. doi: 10.1016/j.bbcan.2015.09.002.

  89. Cetinozman F, Jansen PM, Vermeer MH, et al. Differential expression of programmed death-1 (PD-1) in Sezary syndrome and mycosis fungoides. Arch Dermatol. 2012;148(12):1379. doi: 10.1001/archdermatol.2012.2089.

  90. Khodadoust M, Rook AH, Porcu P, et al. Pembrolizumab for treatment of relapsed/refractory mycosis fungoides and Sezary syndrome: clinical efficacy in a Citn multicenter phase 2 study. Blood. 2016;128:22, abstract 181.

  91. Kwong YL, Chan TSY, Tan D, et al. PD1 blockade with pembrolizumab is highly effective in relapsed or refractory NK/T-cell lymphoma failing l-asparaginase. Blood. 2017;129(17):2437–42. doi: 10.1182/blood-2016-12-756841.

  92. Chan TSY, Li J, Loong F, et al. PD1 blockade with low-dose nivolumab in NK/T cell lymphoma failing L-asparaginase: efficacy and safety. Ann Hematol. 2018;97(1):193–6. doi: 10.1007/s00277-017-3127-2.

  93. Four M, Cacheux V, Tempier A, et al. PD1 and PDL1 expression in primary central nervous system diffuse large B-cell lymphoma are frequent and expression of PD1 predicts poor survival. Hematol Oncol. 2017;35(4):487–96. doi: 10.1002/hon.2375.

  94. Pelland K, Mathews S, Kamath A, et al. Dendritic Cell Markers and PD-L1 are Expressed in Mediastinal Gray Zone Lymphoma. Appl Immunohistochem Mol Morphol. 2017. doi: 10.1097/PAI.0000000000000615. [Epub ahead of print]

  95. Park JH, Han JH, Kanget HY, et al. Expression of follicular helper T-cell markers in primary cutaneous T-cell lymphoma. Am J Dermatopathol. 201;36(6):465–70. doi: 10.1097/DAD.0b013e3182a72f8c.

Theory and Practice of Immunotherapy Directed against the PRAME Antigen

VA Misyurin

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

For correspondence: Vsevolod Andreevich Misyurin, PhD in Biology, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(985)4363019; e-mail: vsevolod.misyurin@gmail.com

For citation: Misyurin VA. Theory and Practice of Immunotherapy Directed against the PRAME Antigen. Clinical oncohematology. 2018;11(2):138–49.

DOI: 10.21320/2500-2139-2018-11-2-138-149


ABSTRACT

The preferentially expressed antigen of melanoma (PRAME) is a significant target for monoclonal antibodies and an oncospecific marker known for its activity on all the tumor cell differentiation stages and its eliciting of a spontaneous T-cell response. Since PRAME protein is active in approximately every second patient with solid tumors and oncohematological diseases, anti-PRAME immunotherapy is very promising. In current review the mechanism of spontaneous immune response against PRAME is discussed as well as the role of this antigen in immunosurveillance. The review deals with the PRAME-specific T-cell genesis and risk assessment of immunotherapy directed against PRAME-positive cells. The risks and benefits of various immunotherapy approaches including the use of dendritic cell vaccines, PRAME vaccination, development of specific T-cells, and development of specific monoclonal antibodies were analysed. Possible causes of treatment failure are analysed, and methods of overcoming them are suggested. The literature search in the Pubmed, Scopus, and eLibrary databases, with the use of “PRAME” as a keyword was performed. Only publications related to various aspects of immunotherapy and anti-PRAME-specific agents were included in the review.

Keywords: PRAME, immunotherapy, dendritic cell vaccines, peptide vaccines, T-cell vaccines, therapeutic antibodies.

Received: December 19, 2017

Accepted: February 5, 2018

Read in PDF 


REFERENCES

  1. Lehmann F, Marchand M, Hainaut P, et al. Differences in the antigens recognized by cytolytic T cells on two successive metastases of a melanoma patient are consistent with immune selection. Eur J Immunol. 1995;25(2):340–7. doi: 10.1002/eji.1830250206.
  2. Ikeda H, Lethe B, Lehmann F, et al. Characterization of an Antigen That Is Recognized on a Melanoma Showing Partial HLA Loss by CTL Expressing an NK Inhibitory Receptor. Immunity. 1997;6(2):199–208. doi: 10.1016/s1074-7613(00)80426-4.
  3. Rezvani K, Yong AS, Tawab A, et al. Ex vivo characterization of polyclonal memory CD8 T-cell responses to PRAME-specific peptides in patients with acute lymphoblastic leukemia and acute and chronic myeloid leukemia. Blood. 2009;113(10):2245–55. doi: 10.1182/blood-2008-03-144071.
  4. Lutz M, Worschech A, Alb M, et al. Boost and loss of immune responses against tumor-associated antigens in the course of pregnancy as a model for allogeneic immunotherapy. Blood. 2015;125(2):261–72. doi: 10.1182/blood-2014-09-601302.
  5. LaVoy EC, Bollard CM, Hanley PJ, et al. A single bout of dynamic exercise enhances the expansion of MAGE-A4 and PRAME-specific cytotoxic T-cells from healthy adults. Exerc Immunol Rev. 2015;21:144–53.
  6. Saldanha-Araujo F, Haddad R, Zanette DL, et al. Cancer/Testis Antigen Expression on Mesenchymal Stem Cells Isolated from Different Tissues. Anticancer Res. 2010;30(12):5023–7. doi: 10.1007/978-94-007-4798-2_11.
  7. Kirkin AF, Dzhandzhugazyan K, Zeuthen J. The Immunogenic Properties of Melanoma-Associated Antigens Recognized by Cytotoxic T Lymphocytes. Exp Clin Immunogenet. 1998;15(1):19–32. doi: 10.1159/000019050.
  8. Luetkens T, Schafhausen P, Uhlich F, et al. Expression, epigenetic regulation, and humoral immunogenicity of cancer-testis antigens in chronic myeloid leukemia. Leuk Res. 2010;34(12):1647–55. doi: 10.1016/j.leukres.2010.03.039.
  9. Luetkens T, Kobold S, Cao Y, et al. Functional autoantibodies against SSX-2 and NY-ESO-1 in multiple myeloma patients after allogeneic stem cell transplantation. Cancer Immunol Immunother. 2014;63(11):1151–62. doi: 10.1007/s00262-014-1588-x.
  10. Kessler JH, Beekman NJ, Bres-Vloemans SA, et al. Efficient Identification of Novel HLA-A*0201–presented Cytotoxic T Lymphocyte Epitopes in the Widely Expressed Tumor Antigen PRAME by Proteasome-mediated Digestion Analysis. J Exp Med. 2001;193(1):73–88. doi: 10.1084/jem.193.1.73.
  11. Quintarelli C, Dotti G, Hasan ST, et al. High-avidity cytotoxic T lymphocytes specific for a new PRAME-derived peptide can target leukemic and leukemic-precursor cells. Blood. 2011;117(12):3353–62. doi: 10.1182/blood-2010-08-300376.
  12. Kessler JH, Mommaas B, Mutis T, et al. Competition-Based Cellular Peptide Binding Assays for 13 Prevalent HLA Class I Alleles Using Fluorescein-Labeled Synthetic Peptides. Hum Immunol. 2003;64(2):245–55. doi: 10.1016/S0198-8859(02)00787-5.
  13. Kawahara M, Hori T, Matsubara Y, et al. Identification of HLA class I–restricted tumor-associated antigens in adult T cell leukemia cells by mass spectrometric analysis. Exp Hematol. 2006;34(11):1496–504. doi: 10.1016/j.exphem.2006.06.010.
  14. Kessler JH, Khan S, Seifert U, et al. Antigen processing by nardilysin and thimet oligopeptidase generates cytotoxic T cell epitopes. Nat Immunol. 2011;12(1):45–53. doi: 10.1038/ni.1974.
  15. Grunebach F, Mirakaj V, Mirakaj V, et al. BCR-ABL Is Not an Immunodominant Antigen in Chronic Myelogenous Leukemia. Cancer Res. 2006;66(11):5892–900. doi: 10.1158/0008-5472.CAN-05-2868.
  16. Greiner J, Schmitt M, Li L, et al. Expression of tumor-associated antigens in acute myeloid leukemia: implications for specific immunotherapeutic approaches. Blood. 2006;108(13):4109–17. doi: 10.1182/blood-2006-01-023127.
  17. Weber G, Caruana I, Rouce RH, et al. Generation of tumor antigen-specific T cell lines from pediatric patients with acute lymphoblastic leukemia – implications for immunotherapy. Clin Cancer Res. 2013;19(18):5079–91. doi: 10.1158/1078-0432.CCR-13-0955.
  18. Schneider V, Zhang L, Rojewski M, et al. Leukemic progenitor cells are susceptible to targeting by stimulated cytotoxic T cells against immunogenic leukemia-associated antigens. Int J Cancer. 2015;137(9):2083–92. doi: 10.1002/ijc.29583.
  19. Babiak A, Steinhauser M, Gotz M, et al. Frequent T cell responses against immunogenic targets in lung cancer patients for targeted immunotherapy. Oncol Rep. 2014;31(1):384–90. doi: 10.3892/or.2013.2804.
  20. Greiner J, Ringhoffer M, Simikopinko O, et al. Simultaneous expression of different immunogenic antigens in acute myeloid leukemia. Exp Hematol. 2000;28(12):1413–22. doi: 10.1016/S0301-472X(00)00550-6.
  21. Griffioen M, Kessler JH, Borghi M, et al. Detection and Functional Analysis of CD8+ T Cells Specific for PRAME: a Target for T-Cell Therapy. Clin Cancer Res. 2006;12(10):3130–6. doi: 10.1158/1078-0432.CCR-05-2578.
  22. Yao J, Caballero OL, Yung WK, et al. Tumor subtype-specific cancer-testis antigens as potential biomarkers and immunotherapeutic targets for cancers. Cancer Immunol Res. 2014;2(4):371–9. doi: 10.1158/2326-6066.CIR-13-0088.
  23. Qin YZ, Zhu HH, Liu YR, et al. PRAME and WT1 transcripts constitute a good molecular marker combination for monitoring minimal residual disease in myelodysplastic syndromes. Leuk Lymphoma. 2013;54(7):1442–9. doi: 10.3109/10428194.2012.743656.
  24. Gutierrez-Cosio S, de la Rica L, Ballestar E, et al. Epigenetic regulation of PRAME in acute myeloid leukemia is different compared to CD34+ cells from healthy donors: Effect of 5-AZA treatment. Leuk Res. 2012;36(7):895–9. doi: 10.1016/j.leukres.2012.02.030.
  25. Greiner J, Ringhoffer M, Taniguchi M, et al. mRNA expression of leukemia-associated antigens in patients with acute myeloid leukemia for the development of specific immunotherapies. Int J Cancer. 2004;108(5):704–11. doi: 10.1002/ijc.11623.
  26. Paydas S, Tanriverdi K, Yavuz S, et al. PRAME mRNA Levels in Cases With Acute Leukemia: Clinical Importance and Future Prospects. Am J Hematol. 2005;79(4):257–61.
  27. Gerber JM, Qin L, Kowalski J, et al. Characterization of chronic myeloid leukemia stem cells. Am J Hematol. 2011;86(1):31–7. doi: 10.1002/ajh.21915.
  28. Yong AS, Keyvanfar K, Eniafe R, et al. Hematopoietic stem cells and progenitors of chronic myeloid leukemia express leukemia-associated antigens: implications for the graft-versus-leukemia effect and peptide vaccine-based immunotherapy. Leukemia. 2008;22(9):1721–7. doi: 10.1038/leu.2008.161.
  29. Steger B, Milosevic S, Doessinger G, et al. CD4+ and CD8+ T-cell reactions against leukemia-associated- or minor-histocompatibility-antigens in AML-patients after allogeneic SCT. Immunobiology. 2014;219(4):247–60. doi: 10.1016/j.imbio.2013.10.008.
  30. Doolan P, Clynes M, Kennedy S, et al. Prevalence and prognostic and predictive relevance of PRAME in breast cancer. Breast Cancer Res Treat. 2008;109(2):359–65. doi: 10.1007/s10549-007-9643-3.
  31. Altvater B, Kailayangiri S, Theimann N, et al. Common Ewing sarcoma-associated antigens fail to induce natural T cell responses in both patients and healthy individual. Cancer Immunol Immunother. 2014;63(10):1047–60. doi: 10.1007/s00262-014-1574-3.
  32. Hughes A, Clarson J, Tang C, et al. CML patients with deep molecular responses to TKI have restored immune effectors and decreased PD-1 and immune suppressors. Blood. 2017;129(9):1166–1176. doi: 10.1182/blood-2016-10-745992.
  33. Schmitt M, Li L, Giannopoulos K, et al. Chronic myeloid leukemia cells express tumor-associated antigens eliciting specific CD8+ T-cell responses and are lacking costimulatory molecules. Exp Hematol. 2006;34(12):1709–19. doi: 10.1016/j.exphem.2006.07.009.
  34. Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8(1):59–73. doi: 10.1038/nri2216.
  35. Morandi F, Chiesa S, Bocca P, et al. Tumor mRNA–Transfected Dendritic Cells Stimulate the Generation of CTL That Recognize Neuroblastoma-Associated Antigens and Kill Tumor Cells: Immunotherapeutic Implications. Neoplasia. 2006;8(10):833–42. doi: 10.1593/neo.06415.
  36. Winkler C, Steingrube DS, Altermann W, et al. Hodgkin’s lymphoma RNA-transfected dendritic cells induce cancer/testis antigen-specific immune responses. Cancer Immunol Immunother. 2012;61(10):1769–79. doi: 10.1007/s00262-012-1239-z.
  37. Gerdemann U, Katari U, Christin AS, et al. Cytotoxic T Lymphocytes Simultaneously Targeting Multiple Tumor-associated Antigens to Treat EBV Negative Lymphoma. Mol Ther. 2011;19(12):2258–68. doi: 10.1038/mt.2011.167.
  38. Mohamed YS, Bashawri LA, Vatte C, et al. The in vitro generation of multi-tumor antigen-specific cytotoxic T cell clones: Candidates for leukemia adoptive immunotherapy following allogeneic stem cell transplantation. Mol Immunol. 2016;77:79–88. doi: 10.1016/j.molimm.2016.07.012.
  39. Li L, Schmitt A, Reinhardt P, et al. Reconstitution of CD40 and CD80 in dendritic cells generated from blasts of patients with acute myeloid leukemia. Cancer Immun. 2003;3:8.
  40. Li L, Reinhardt P, Schmitt A, et al. Dendritic cells generated from acute myeloid leukemia (AML) blasts maintain the expression of immunogenic leukemia associated antigens. Cancer Immunol Immunother. 2005;54(7):685–93. doi: 10.1007/s00262-004-0631-8.
  41. Li L, Giannopoulos K, Reinhardt P, et al. Immunotherapy for patients with acute myeloid leukemia using autologous dendritic cells generated from leukemic blasts. Int J Oncol. 2006;28(4):855–61. doi: 10.3892/ijo.28.4.855.
  42. Altvater B, Pscherer S, Landmeier S, et al. Activated human γδ T cells induce peptide-specific CD8+ T-cell responses to tumor-associated self-antigens. Cancer Immunol Immunother. 2012;61(3):385–96. doi: 10.1007/s00262-011-1111-6.
  43. Matsushita M, Ikeda H, Kizaki M, et al. Quantitative monitoring of the PRAME gene for the detection of minimal residual disease in leukaemia. Br J Haematol. 2001;112(4):916–26. doi: 10.1046/j.1365-2141.2001.02670.x.
  44. van den Ancker W, Ruben JM, Westers TM, et al. Priming of PRAME- and WT1-specific CD8+ T cells in healthy donors but not in AML patients in complete remission. Oncoimmunology. 2013;2(4):e23971. doi: 10.4161/onci.23971.
  45. Yao Y, Zhou J, Wang L, et al. Increased PRAME-Specific CTL Killing of Acute Myeloid Leukemia Cells by Either a Novel Histone Deacetylase Inhibitor Chidamide Alone or Combined Treatment with Decitabine. PLoS One. 2013;8(8):e70522. doi: 10.1371/journal.pone.0070522.
  46. Zhang M, Graor H, Visioni A, et al. T Cells Derived From Human Melanoma Draining Lymph Nodes Mediate Melanoma-specific Antitumor Responses In Vitro and In Vivo in Human Melanoma Xenograft Model. J Immunother. 2015;38(6):229–38. doi: 10.1097/CJI.0000000000000078.
  47. Yan M, Himoudi N, Basu BP, et al. Increased PRAME antigen-specific killing of malignant cell lines by low avidity CTL clones, following treatment with 5-Aza-20-Deoxycytidine. Cancer Immunol Immunother. 2011;60(9):1243–55. doi: 10.1007/s00262-011-1024-4.
  48. Quintarelli C, Dotti G, De Angelis B, et al. Cytotoxic T lymphocytes directed to the preferentially expressed antigen of melanoma (PRAME) target chronic myeloid leukemia. Blood. 2008;112(5):1876–85. doi: 10.1182/blood-2008-04-150045.
  49. Amir AL, van der Steen DM, van Loenen MM, et al. PRAME-Specific Allo-HLA–Restricted T Cells with Potent Antitumor Reactivity Useful for Therapeutic T-Cell Receptor Gene Transfer. Clin Cancer Res. 2011;17(17):5615–25. doi: 10.1158/1078-0432.CCR-11-1066.
  50. van Loenen MM, de Boer R, Hagedoorn RS, et al. Multi-cistronic vector encoding optimized safety switch for adoptive therapy with T-cell receptor-modified T cells. Gene Ther. 2013;20(8):861–7. doi: 10.1038/gt.2013.4.
  51. Spel L, Boelens JJ, van der Steen DM, et al. Natural killer cells facilitate PRAME-specific T-cell reactivity against neuroblastoma. Oncotarget. 2015;6(34):35770–81. doi: 10.18632/oncotarget.5657.
  52. Weber JS, Vogelzang NJ, Ernstoff MS, et al. A Phase 1 Study of a Vaccine Targeting Preferentially Expressed Antigen in Melanoma and Prostate-specific Membrane Antigen in Patients With Advanced Solid Tumors. J Immunother. 2011;34(7):556–67. doi: 10.1097/CJI.0b013e3182280db1.
  53. Garcon N, Silvano J, Kuper CF, et al. Non-clinical safety evaluation of repeated intramuscular administration of the AS15 immunostimulant combined with various antigens in rabbits and cynomolgus monkeys. J Appl Toxicol. 2016;36(2):238–56. doi: 10.1002/jat.3167.
  54. Gerard C, Baudson N, Ory T, et al. A Comprehensive Preclinical Model Evaluating the Recombinant PRAME Antigen Combined With the AS15 Immunostimulant to Fight Against PRAME-expressing Tumors. J Immunother. 2015;38(8):311–20. doi: 10.1097/CJI.0000000000000095.
  55. Pujol JL, De Pas T, Rittmeyer A, et al. Safety and Immunogenicity of the PRAME Cancer Immunotherapeutic in Patients with Resected Non–Small Cell Lung Cancer: A Phase I Dose Escalation Study. J Thorac Oncol. 2016;11(12):2208–17. doi: 10.1016/j.jtho.2016.08.120.
  56. Gutzmer R, Rivoltini L, Levchenko E, et al. Safety and immunogenicity of the PRAME cancer immunotherapeutic in metastatic melanoma: results of a phase I dose escalation study. ESMO Open. 2016;1(4):e000068.
  57. Blais N, Martin D, Palmantier RM. Vaccin. Patent PCT/EP2008/050290. Available from: https://patentscope.wipo.int/search/ru/detail.jsf?docId=WO2008087102&redirectedID=true. (accessed 08.12.2017).
  58. Chang AY, Dao T, Gejman RS, et al. A therapeutic T cell receptor mimic antibody targets tumor-associated PRAME peptide/HLA-I antigens. J Clin Invest. 2017;127(7):2705–18. doi: 10.1172/JCI92335.
  59. Pankov D, Sjostrom L, Kalidindi T, et al. In vivo immuno-targeting of an extracellular epitope of membrane bound preferentially expressed antigen in melanoma (PRAME). Oncotarget. 2017;8(39):65917–31. doi: 10.18632/oncotarget.19579.
  60. Финашутина Ю.П., Мисюрин А.В., Ахлынина Т.В. и др. Получение рекомбинантного раково-тестикулярного белка PRAME и моноклональных антител к нему. Российский биотерапевтический журнал. 2015;14(3):29–36.[Finashutina YuP, Misyurin AV, Akhlynina TV, et al. Production of recombinant PRAME cancer testis antigen and its specific monoclonal antibodies. Rossiiskii bioterapevticheskii zhurnal. 2015;14(3):29–36. (In Russ)]
  61. Мисюрин А.В., Финашутина Ю.П. Антигенная композиция и ее терапевтическое применение для профилактики и лечения онкологических заболеваний, рекомбинантная плазмидная ДНК, обеспечивающая синтез гибридного белка, а также способ получения белка. Патент РФ на изобретение № 2590701/13.04.29. Бюл. № 19. Доступно по: http://www.fips.ru/cdfi/fips.dll/en?ty=29&docid=2590701. Ссылка активна на 08.12.2017.[Misyurin AV, Finashutina YuP. Antigennaya kompozitsiya i ee terapevticheskoe primenenie dlya profilaktiki i lecheniya onkologicheskikh zabolevanii, rekombinantnaya plazmidnaya DNK, obespechivayushchaya sintez gibridnogo belka, a takzhe sposob polucheniya belka. Patent RUS No. 2590701/13.04.29. Byul. No. 19. Available from: http://www.fips.ru/cdfi/fips.dll/en?ty=29&docid=2590701. (accessed 08.12.2017) (In Russ)]
  62. Лыжко Н.А., Ахлынина Т.В., Мисюрин А.В. и др. Повышение уровня экспрессии гена PRAME в опухолевых клетках сопровождается локализацией белка в клеточном ядре. Российский биотерапевтический журнал. 2015;14(4):19–30.[Lyzhko NA, Ahlynina TV, Misyurin AV, et al. The increased PRAME expression in cancer cells is associated with deposit of the protein in cell nucleus. Rossiiskii bioterapevticheskii zhurnal. 2015;14(4):19–30. (In Russ)]
  63. Лыжко Н.А., Мисюрин В.А., Финашутина Ю.П. и др. Проявление цитостатического эффекта моноклональных антител к белку PRAME. Российский биотерапевтический журнал. 2016;15(4):53–8. doi: 10.17650/1726-9784-2016-15-4-53-58.[Lyzhko NA, Misyurin VA, Finashutina YuP, et al. Development of cytostatic effect of monoclonal antibodies to the protein PRAME. Rossiiskii bioterapevticheskii zhurnal. 2016;15(4):53–8. doi: 10.17650/1726-9784-2016-15-4-53-58. (In Russ)]
  64. Dillman RO. Cancer immunotherapy. Cancer Biother Radiopharm 2011;26:1–64. doi: 10.1089/cbr.2010.0902.
  65. Theisen D, Murphy K. The role of cDC1s in vivo: CD8 T cell priming through cross-presentation. F1000Res. 2017;6:98. doi: 10.12688/f1000research.9997.1.
  66. Epping MT, Wang L, Edel MJ, et al. The human tumor antigen PRAME is a dominant repressor of retinoic acid receptor signaling. Cell. 2005;122(6):835–47. doi: 10.1016/j.cell.2005.07.003.
  67. De Carvalho DD, Mello BP, Pereira WO, Amarante-Mendes GP. PRAME/EZH2-mediated regulation of TRAIL: a new target for cancer therapy. Curr Mil Med. 2013;13(2):296–304. doi: 10.2174/1566524011313020006.
  68. Мисюрин В.А. Клиническое значение экспрессии гена PRAME при онкогематологических заболеваниях. Клиническая онкогематология. 2018;11(1):26–33. doi: 10.21320/2500-2139-2018-11-1-26-33.[Misyurin VA. Clinical Significance of the PRAME Gene Expression in Oncohematological Diseases. Clinical oncohematology.2018;11(1):26–33. doi: 10.21320/2500-2139-2018-11-1-26-33. (In Russ)]

Clinical Significance of the PRAME Gene Expression in Oncohematological Diseases

VA Misyurin

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

For correspondence: Vsevolod Andreevich Misyurin, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(985)436-30-19; e-mail: vsevolod.misyurin@gmail.com

For citation: Misyurin AV. Clinical Significance of the PRAME Gene Expression in Oncohematological Diseases. Clinical oncohematology. 2018;11(1):26–33.

DOI: 10.21320/2500-2139-2018-11-1-26-33


ABSTRACT

Although the PRAME activity was first discovered in solid tumors, this gene is very frequently expressed in oncohematological diseases. PRAME can be regarded as a reliable biomarker of tumor cells. Determination of PRAME transcripts is used in residual disease monitoring and molecular relapse diagnostics. Experimentation with PRAME expressing lines of leukemia cells yielded controversial results. Therefore, it is hardly possible to estimate the prognostic value of PRAME activity in oncohematological diseases. In chronic myeloproliferative disease and chronic myeloid leukemia, however, PRAME activity proves to be a predictor of negative prognosis, and on the contrary, it can be regarded as a positive prognostic factor in acute myeloid or lymphoid leukemia. Despite many clinical studies prognostic value of PRAME expression in some diseases requires further investigation. The present literature review contains the data concerning PRAME expression in oncohematological diseases.

Keywords: PRAME, leukemia, lymphoma, prognosis.

Received: September 14, 2017

Accepted: December 2, 2017

Read in PDF 


REFERENCES

  1. Ikeda H, Lethe B, Lehmann F, et al. Characterization of an antigen that is recognized on a melanoma showing partial HLA loss by CTL expressing an NK inhibitory receptor. Immunity. 1997;6(2):199–208. doi: 10.1016/S1074-7613(00)80426-4.
  2. Greiner J, Ringhoffer M, Simikopinko O, et al. Simultaneous expression of different immunogenic antigens in acute myeloid leukemia. Exp Hematol. 2000;28(12):1413–22. doi: 10.1016/S0301-472X(00)00550-6.
  3. Epping MT, Wang L, Edel MJ, et al. The human tumor antigen PRAME is a dominant repressor of retinoic acid receptor signaling. Cell. 2005;122(6):835–47. doi: 10.1016/j.cell.2005.07.003.
  4. De Carvalho DD, Mello BP, Pereira WO, Amarante-Mendes GP. PRAME/EZH2-mediated regulation of TRAIL: a new target for cancer therapy. Curr Mol Med. 2013;13(2):296–304. doi: 10.2174/156652413804810727.
  5. Costessi A, Mahrour N, Tijchon E, et al. The tumour antigen PRAME is a subunit of a Cul2 ubiquitin ligase and associates with active NFY promoters. EMBO J. 2011;30(18):3786–98. doi: 10.1038/emboj.2011.262.
  6. Kim HL, Seo YR. Molecular and genomic approach for understanding the gene-environment interaction between Nrf2 deficiency and carcinogenic nickel-induced DNA damage. Oncol Rep. 2012;28(6):1959–67. doi: 10.3892/or.2012.2057.
  7. Yao J, Caballero OL, Yung WK, et al. Tumor subtype-specific cancer-testis antigens as potential biomarkers and immunotherapeutic targets for cancers. Cancer Immunol Res. 2014;2(4):371–9. doi: 10.1158/2326-6066.CIR-13-0088.
  8. van Baren N, Chambost H, Ferrant A, et al. PRAME, a gene encoding an antigen recognized on a human melanoma by cytolytic T cells, is expressed in acute leukaemia cells. Br J Haematol. 1998;102(5):1376–9. doi: 10.1046/j.1365-2141.1998.00982.x.
  9. Oehler VG, Guthrie KA, Cummings CL, et al. The preferentially expressed antigen in melanoma (PRAME) inhibits myeloid differentiation in normal hematopoietic and leukemic progenitor cells. Blood. 2009;114(15):3299–308. doi: 10.1182/blood-2008-07-170282.
  10. Roman-Gomez J, Jimenez-Velasco A, Agirre X, et al. Epigenetic regulation of PRAME gene in chronic myeloid leukemia. Leuk Res. 2007;31(11):1521–8. doi: 10.1016/j.leukres.2007.02.016.
  11. Ortmann CA, Eisele L, Nuckel H, et al. Aberrant hypomethylation of the cancer–testis antigen PRAME correlates with PRAME expression in acute myeloid leukemia. Ann Hematol. 2008;87(10):809–18. doi: 10.1007/s00277-008-0514-8.
  12. Gutierrez-Cosio S, de la Rica L, Ballestar E, et al. Epigenetic regulation of PRAME in acute myeloid leukemia is different compared to CD34+ cells from healthy donors: Effect of 5-AZA treatment. Leuk Res. 2012;36(7):895–9. doi: 10.1016/j.leukres.2012.02.030.
  13. Arons E, Suntum T, Margulies I, et al. PRAME expression in Hairy Cell Leukemia. Leuk Res. 2008;32(9):1400–6. doi: 10.1016/j.leukres.2007.12.010.
  14. Steinbach D, Schramm A, Eggert A, et al. Identification of a Set of Seven Genes for the Monitoring of Minimal Residual Disease in Pediatric Acute Myeloid Leukemia. Clin Cancer Res. 2006;12(8):2434–41. doi: 10.1158/1078-0432.CCR-05-2552.
  15. Matsushita M, Ikeda H, Kizaki M, et al. Quantitative monitoring of the PRAME gene for the detection of minimal residual disease in leukaemia. Br J Haematol. 2001;112(4):916–26. doi: 10.1046/j.1365-2141.2001.02670.x.
  16. Tajeddine N, Millard I, Gailly P, Gala JL. Real-time RT-PCR quantification of PRAME gene expression for monitoring minimal residual disease in acute myeloblastic leukaemia. Clin Chem Lab Med. 2006;44(5):548–55. doi: 10.1515/CCLM.2006.106.
  17. Schneider V, Zhang L, Rojewski M, et al. Leukemic progenitor cells are susceptible to targeting by stimulated cytotoxic T cells against immunogenic leukemia-associated antigens. Int J Cancer. 2015;137(9):2083–92. doi: 10.1002/ijc.29583.
  18. Гапонова Т.В., Менделеева Л.П., Мисюрин А.В. и др. Экспрессия опухолеассоциированных генов PRAME, WT1 и XIAP у больных множественной миеломой. Онкогематология. 2009;2:52–7. [Gaponova TV, Mendeleeva LP, Misyurin AV, et al. Expression of PRAME, WT1 and XIAP tumor-associated genes in patients with multiple myeloma. Onkogematologiya. 2009;2:52–7. (In Russ)]
  19. Абраменко И.В., Белоус Н.И., Крячок И.А. и др. Экспрессия гена PRAME при множественной миеломе. Терапевтический архив. 2004;74(7):77–81. [Abramenko IV, Belous NI, Kryachok IA, et al. Expression of PRAME gene in multiple myeloma. Terapevticheskii arkhiv. 2004;74(7):77–81. (In Russ)]
  20. Мисюрин В.А., Мисюрин А.В., Кесаева Л.А. и др. Новые маркеры прогрессирования хронического миелолейкоза. Клиническая онкогематология. 2014;7(2):206–12. [Misyurin VA, Misyurin AV, Kesayeva LA, et al. New molecular markers of CML progression. Klinicheskaya onkogematologiya. 2014;7(2):206–12. (In Russ)]
  21. van Baren N, Brasseur F, Godelaine D, et al. Genes encoding tumor-specific antigens are expressed in human myeloma cells. Blood. 1999;94(4):1156–64.
  22. Pellat-Deceunynck C, Mellerin M., Labarriere N, et al. The cancer germ-line genes MAGE-1, MAGE-3 and PRAME are commonly expressed by human myeloma cells. Eur J Immunol. 2000;30(3):803–9. doi: 10.1002/1521-4141(200003)30:3<803:AID-IMMU803>3.0.CO;2-P.
  23. Andrade VC, Vettore AL, Felix RS, et al. Prognostic impact of cancer/testis antigen expression in advanced stage multiple myeloma patients. Cancer Immun. 2008;8:2.
  24. Qin Y, Lu J, Bao L, et al. Bortezomib improves progression-free survival in multiple myeloma patients overexpressing preferentially expressed antigen of melanoma. Chin Med J (Engl). 2014;127(9):1666–71. doi: 10.3760/cma.j.issn.0366-6999.20132356.
  25. Proto-Siqueira R, Falcao RP, de Souza CA, et al. The expression of PRAME in chronic lymphoproliferative disorders. Leuk Res. 2003;27(5):393–6. doi: 10.1016/S0145-2126(02)00217-5.
  26. Proto-Siqueira R, Figueiredo-Pontes LL, Panepucci RA, et al. PRAME is a membrane and cytoplasmic protein aberrantly expressed in chronic lymphocytic leukemia and mantle cell lymphoma. Leuk Res. 2006;30(11):1333–39. doi: 10.1016/j.leukres.2006.02.031.
  27. Paydas S, Tanriverdi K, Yavuz S, Seydaoglu G. PRAME mRNA levels in cases with chronic leukemia: Clinical importance and review of the literature. Leuk Res. 2007;31(3):365–9. doi: 10.1016/j.leukres.2006.06.022.
  28. Kawano R, Karube K, Kikuchi M, et al. Oncogene associated cDNA microarray analysis shows PRAME gene expression is a marker for response to anthracycline containing chemotherapy in patients with diffuse large B-cell lymphoma. J Clin Exp Hematop. 2009;49(1):1–7. doi: 10.3960/jslrt.49.1.
  29. Mitsuhashi K, Masuda A, Wang YH, et al. Prognostic significance of PRAME expression based on immunohistochemistry for diffuse large B-cell lymphoma patients treated with R-CHOP therapy. Int J Hematol. 2014;100(1):88–95. doi: 10.1007/s12185-014-1593-z.
  30. Schmitt M, Li L, Giannopoulos K, et al. Chronic myeloid leukemia cells express tumor-associated antigens eliciting specific CD8+ T-cell responses and are lacking costimulatory molecules. Exp Hematol. 2006;34(12):1709–19. doi: 10.1016/j.exphem.2006.07.009.
  31. Qian J, Zhu Z.H, Lin J, et al. Hypomethylation of PRAME promoter is associated with poor prognosis in myelodysplastic syndrome. Br J Haematol. 2011;154(1):153–5. doi: 10.1111/j.1365-2141.2011.08585.x.
  32. Ding K, Wang XM, Fu R, et al. PRAME Gene Expression in Acute Leukemia and Its Clinical Significance. Cancer Biol Med. 2012;9(1):73–6. doi: 10.3969/j.issn.2095-3941.2012.01.013.
  33. Greiner J, Ringhoffer M, Taniguchi M, et al. mRNA expression of leukemia-associated antigens in patients with acute myeloid leukemia for the development of specific immunotherapies. Int J Cancer. 2004;108(5):704–11. doi: 10.1002/ijc.11623.
  34. Li L, Reinhardt P, Schmitt A, et al. Dendritic cells generated from acute myeloid leukemia (AML) blasts maintain the expression of immunogenic leukemia associated antigens. Cancer Immunol Immunother. 2005;54(7):685–93. doi: 10.1007/s00262-004-0631-8.
  35. Atanackovic D, Luetkens T, Kloth B, et al. Cancer-testis antigen expression and its epigenetic modulation in acute myeloid leukemia. Am J Hematol. 2011;86(11):918–22. doi: 10.1002/ajh.22141.
  36. Gerber JM, Qin L, Kowalski J, et al. Characterization of chronic myeloid leukemia stem cells. Am J Hematol. 2011;86(1):31–7. doi: 10.1002/ajh.21915.
  37. Qin YZ, Zhu HH, Liu YR, et al. PRAME and WT1 transcripts constitute a good molecular marker combination for monitoring minimal residual disease in myelodysplastic syndromes. Leuk Lymphoma. 2013;54(7):1442–9. doi: 10.3109/10428194.2012.743656.
  38. Steinbach D, Viehmann S, Zintl F, Gruhn B. PRAME gene expression in childhood acute lymphoblastic leukemia. Cancer Genet Cytogenet. 2002;138(1):89–91. doi: 10.1016/S0165-4608(02)00582-4.
  39. Steinbach D, Hermann J, Viehmann S, et al. Clinical implications of PRAME gene expression in childhood acute myeloid leukemia. Cancer Genet Cytogenet. 2002;133(2):118–23. doi: 10.1016/S0165-4608(01)00570-2.
  40. Spanaki A, Perdikogianni C, Linardakis E, Kalmanti M. Quantitative assessment of PRAME expression in diagnosis of childhood acute leukemia. Leuk Res. 2007;31(5):639–42. doi: 10.1016/j.leukres.2006.06.006.
  41. Steinbach D, Bader P, Willasch A, et al. Prospective Validation of a New Method of Monitoring Minimal Residual Disease in Childhood Acute Myelogenous Leukemia. Clin Cancer Res. 2015;21(6):1353–9. doi: 10.1158/1078-0432.CCR-14-1999.
  42. Paydas S, Tanriverdi K, Yavuz S, et al. PRAME mRNA levels in cases with chronic leukemia: Clinical Importance and Future Prospects. Am J Hematol. 2005;79(4):257–61. doi: 10.1002/ajh.20425.
  43. Steinbach D, Pfaffendorf N, Wittig S, Gruhn B. PRAME expression is not associated with down-regulation of retinoic acid signaling in primary acute myeloid leukemia. Cancer Genet Cytogenet. 2007;177(1):51–4. doi: 10.1016/j.cancergencyto.2007.05.011.
  44. Santamaria C, Chillon MC, Garcia-Sanz R, et al. The relevance of preferentially expressed antigen of melanoma (PRAME) as a marker of disease activity and prognosis in acute promyelocytic leukemia. Haematologica. 2008;93(12):1797–805. doi: 10.3324/haematol.13214.
  45. Qin Y, Zhu H, Jiang B, et al. Expression patterns of WT1 and PRAME in acute myeloid leukemia patients and their usefulness for monitoring minimal residual disease. Leuk Res. 2009;33(3):384–90. doi: 10.1016/j.leukres.2008.08.026.
  46. Мисюрин В.А., Лукина А.Е., Мисюрин А.В. и др. Особенности соотношения уровней экспрессии генов PRAME и PML/RARa в дебюте острого промиелоцитарного лейкоза. Российский биотерапевтический журнал. 2014;13(1):9–16. [Misyurin VA, Lukina AE, Misyurin AV, et al. A ratio between gene expression levels of PRAME and PML/RARA at the onset of acute promyelocytic leukemia and clinical features of the disease. Rossiiskii bioterapevticheskii zhurnal. 2014;13(1):9–16. (In Russ)]
  47. Liberante FG, Pellagatti A, Boncheva V, et al. High and low, but not intermediate, PRAME expression levels are poor prognostic markers in myelodysplastic syndrome at disease presentation. Br J Haematol. 2013;162(2):282–5. doi: 10.1111/bjh.12352.
  48. Goellner S, Steinbach D, Schenk T, et al. Childhood acute myelogenous leukaemia: Association between PRAME, apoptosis- and MDR-related gene expression. Eur J Cancer. 2006;42(16):2807–14. doi: 10.1016/j.ejca.2006.06.018.
  49. Tajeddine N, Louis M, Vermylen C, et al. Tumor associated antigen PRAME is a marker of favorable prognosis in childhood acute myeloid leukemia patients and modifies the expression of S100A4, Hsp 27, p21, IL-8 and IGFBP-2 in vitro and in vivo. Leuk Lymphoma. 2008;49(6):1123–31. doi: 10.1080/10428190802035933.
  50. Santamaria CM, Chillon MC, Garcia-Sanz R, et al. Molecular stratification model for prognosis in cytogenetically normal acute myeloid leukemia. Blood. 2009;114(1):148–52. doi: 10.1182/blood-2008-11-187724.
  51. Ercolak V, Paydas S, Bagir E, et al. PRAME Expression and Its Clinical Relevance in Hodgkin’s Lymphoma. Acta Haematol. 2015;134(4):199–207. doi: 10.1159/000381533.
  52. Luetkens T, Kobold S, Cao Y, et al. Functional autoantibodies against SSX‐2 and NY‐ESO‐1 in multiple myeloma patients after allogeneic stem cell transplantation. Cancer Immunol Immunother. 2014;63(11):1151–62. doi: 10.1007/s00262-014-1588-x.
  53. Gunn SR, Bolla AR, Barron LL, et al. Array CGH analysis of chronic lymphocytic leukemia reveals frequent cryptic monoallelic and biallelic deletions of chromosome 22q11 that include the PRAME gene. Leuk Res. 2009;33(9):1276–81. doi: 10.1016/j.leukres.2008.10.010.
  54. Mraz M, Stano Kozubik K, Plevova K, et al. The origin of deletion 22q11 in chronic lymphocytic leukemia is related to the rearrangement of immunoglobulin lambda light chain locus. Leuk Res. 2013;37(7):802–8. doi: 10.1016/j.leukres.2013.03.018.
  55. Staege MS, Banning-Eichenseer U, Weissflog G, et al. Gene expression profiles of Hodgkin’s lymphoma cell lines with different sensitivity to cytotoxic drugs. Exp Hematol. 2008;36(7):886–96. doi: 10.1016/j.exphem.2008.02.014.
  56. Kewitz S, Staege MS. Knock-Down of PRAME Increases Retinoic Acid Signaling and Cytotoxic Drug Sensitivity of Hodgkin Lymphoma Cells. PLoS One. 2013;8(2):e55897. doi: 10.1371/journal.pone.0055897.
  57. Bea S, Salaverria I, Armengol L, et al. Uniparental disomies, homozygous deletions, amplifications, and target genes in mantle cell lymphoma revealed by integrative high-resolution whole-genome profiling. Blood. 2009;113(13):3059–69. doi: 10.1182/blood-2008-07-170183.
  58. Liggins AP, Lim SH, Soilleux EJ, et al. A panel of cancer-testis genes exhibiting broadspectrum expression in haematological malignancies. Cancer Immun. 2010;10:8.
  59. Radich JP, Dai H, Mao M, et al. Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci USA. 2006;103(8):2794–9. doi: 10.1073/pnas.0510423103.
  60. Luetkens T, Schafhausen P, Uhlich F, et al. Expression, epigenetic regulation, and humoral immunogenicity of cancer-testis antigens in chronic myeloid leukemia. Leuk Res. 2010;34(12):1647–55. doi: 10.1016/j.leukres.2010.03.039.
  61. Hughes A, Clarson J, Tang C, et al. CML patients with deep molecular responses to TKI have restored immune effectors and decreased PD-1 and immune suppressors. Blood. 2017;129(9):1166–76. doi: 10.1182/blood-2016-10-745992.
  62. Khateeb EE, Morgan D. Preferentially Expressed Antigen of Melanoma (PRAME) and Wilms’ Tumor 1 (WT 1) Genes Expression in Childhood Acute Lymphoblastic Leukemia, Prognostic Role and Correlation with Survival. Open Access Maced J Med Sci. 2015;3(1):57–62. doi: 10.3889/oamjms.2015.001.
  63. Zhang YH, Lu AD, Yang L, et al. PRAME overexpression predicted good outcome in pediatric B-cell acute lymphoblastic leukemia patients receiving chemotherapy. Leuk Res. 2017;52):43–9. doi: 10.1016/j.leukres.2016.11.005.
  64. McElwaine S, Mulligan C, Groet J, et al. Microarray transcript profiling distinguishes the transient from the acute type of megakaryoblastic leukaemia (M7) in Down’s syndrome, revealing PRAME as a specific discriminating marker. Br J Haematol. 2004;125(6):729–42. doi: 10.1111/j.1365-2141.2004.04982.x.
  65. Tanaka N, Wang YH, Shiseki M, et al. Inhibition of PRAME expression causes cell cycle arrest and apoptosis in leukemic cells. Leuk Res. 2011;35(9):1219–25. doi: 10.1016/j.leukres.2011.04.005.
  66. De Carvalho D.D, Binato R, Pereira W.O, et al. BCR-ABL-mediated upregulation of PRAME is responsible for knocking down TRAIL in CML patients. Oncogene. 2011;30(2):223–33. doi: 10.1038/onc.2010.409.
  67. Tajeddine N, Gala JL, Louis M, et al. Tumor-associated antigen preferentially expressed antigen of melanoma (PRAME) induces caspase-independent cell death in vitro and reduces tumorigenicity in vivo. Cancer Res. 2005;65(16):7348–55. doi: 10.1158/0008-5472.CAN-04-4011.
  68. Yan H, Zhao RM, Wang ZJ, et al. Knockdown of PRAME enhances adriamycin-induced apoptosis in chronic myeloid leukemia cells. Eur Rev Med Pharmacol Sci. 2015;19(24):4827–34. doi: 10.18632/oncotarget.9977.
  69. Xu Y, Yue Q, Wei H, Pan G. PRAME induces apoptosis and inhibits proliferation of leukemic cells in vitro and in vivo. Int J Clin Exp Pathol. 2015;8(11):14549–55.
  70. Xu Y, Rong LJ, Meng SL, et al. PRAME promotes in vitro leukemia cells death by regulating S100A4/p53 signaling. Eur Rev Med Pharmacol Sci. 2016;20(6):1057–63.
  71. Bullinger L, Schlenk RF, Gotz M, et al. PRAME-Induced Inhibition of Retinoic Acid Receptor Signaling-Mediated Differentiation – Possible Target for ATRA Response in AML without t(15;17). Clin Cancer Res. 2013;19(9):2562–71. doi: 10.1158/1078-0432.CCR-11-2524.

New Possibilities of Treatment for Relapsed/Refractory Multiple Myeloma: A Literature Review

OM Votyakova

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

For correspondence: Ol’ga Mikhailovna Votyakova, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: 8(499)324-92-09; e-mail: omvtk@yandex.ru 24

For citation: Votyakova OM. New Possibilities of Treatment for Relapsed/Refractory Multiple Myeloma: A Literature Review. Clinical oncohematology. 2017;10(4):425–34 (In Russ).

DOI: 10.21320/2500-2139-2017-10-4-425-434


ABSTRACT

Despite improvements in the treatment of patients with newly diagnosed multiple myeloma (MM) through the introduction of new drugs and high-dose chemotherapy with autologous hematopoietic stem cell transplantation, relapses are still inevitable in all patients. The use of immunomodulatory agents (thalidomide, lenalidomide) and proteasome inhibitor bortezomib has improved the treatment of relapses. However, the disease progression lead to repeated relapses and eventually refractory MM. For these patients new therapeutic strategies are needed, including the development of more effective drugs within the existing classes and the study of new combinations, as well as searching new targets for the treatment of MM. We present the key clinical data on the efficacy and safety of the most promising proteasome inhibitors (carfilzomib, ixazomib), new generation immunomodulatory drug pomalidomide, and monoclonal antibodies (daratumumab and elotuzumab).

Keywords: proteasome inhibitors, immunomodulatory medication, monoclonal antibodies, multiple myeloma.

Received: March 14, 2017

Accepted: June 20, 2017

Read in PDF


REFERENCES

  1. Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued improvement in survival in multiple myeloma: changes in early mortality and outcomes in older patients. Leukemia. 2014;28(5):1122–5. doi: 10.1038/leu.2013.313.
  2. Ludwig H, Sonneveld P, Davies F, et al. European Perspective on Multiple Myeloma Treatment Strategies in 2014. Oncologist. 2014;19(8):829–44. doi: 10.1634/theoncologist.2014-0042.
  3. Rajkumar SV, Harousseau J-L, Durie B, et al. Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1. Blood. 2011;117(18):4691–5. doi: 10.1182/blood-2010-10-299487.
  4. Richardson PG, Sonnefeld P, Schuster M, et al. Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood. 2007;110(11):3557–60. doi: 10.1182/blood-2006-08-036947.
  5. Dimopoulos MA, Spencer A, Attal M, et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med. 2007;357(21):2123–32. doi: 10.1056/NEJMoa070594.
  6. Weber DM, Chen C, Niesvizky R, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med. 2007;357(21):2133–42. doi: 10.1056/NEJMoa070596.
  7. Weber D, Knight R, Chen C, et al. Prolonged overall survival with lenalidomide plus dexamethasone in patients with relapsed or refractory multiple myeloma. Blood. 2007;110(11): Abstract 412.
  8. Kumar SK, Therneau TM, Gertz MA, et al. Clinical course of patients with relapsed multiple myeloma. Mayo Clin Proc. 2004;79(7):867–74.
  9. Usmani S, Ahmadi T, Ng Y, et al. Analysis of Real-World Data on Overall Survival in Multiple Myeloma Patients With ≥ 3 Prior Lines of Therapy Including a Proteasome Inhibitor (PI) and an Immunomodulatory Drug (IMiD), or Double Refractory to a PI and an IMiD. Oncologist. 2016;21:1355–61. doi: 10.1634/theoncologist.2016-0104.
  10. Kumar SK, Lee JH, Lahuerta JJ, et al. Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: a multicenter International Myeloma Working Group study. Leukemia. 2012;26(1):149–57. doi: 10.1038/leu.2011.196.
  11. Bolli N, Avert-Loiseau H, Wedge DC, et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun. 2014;5:2997. doi: 10.1038/ncomms3997.
  12. Lohr JG, Stojanov P, Carter SL, et al. Multiple Myeloma Research Consortium. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell. 2014;25(1):91–101. doi: 10.1016/j.ccr.2013.12.015.
  13. Egan JB, Shi CH, Tembe W, et al. Whole-genome sequencing of multiple myeloma from diagnosis to plasma cell leukemia reveals genomic initiating events, evolution, and clonal tides. Blood. 2012;120(5):1060–6. doi: 10.1182/blood-2012-01-405977.
  14. Yee AJ, Raje NS. Sequencing of nontransplant treatments in multiple myeloma patients with active disease. Hematology Am Soc Hematol Educ Program. 2016(1):495–503. doi: 10.1182/asheducation-2016.1.495.
  15. 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.
  16. 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.
  17. 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.
  18. Dimopoulos MA, Oriol A, Nahi H, et al. Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med. 2016;375(14):1319–31. doi: 10.1056/NEJMoa1607751.
  19. Kuhn DJ, Orlowski RZ, Bjorklund CC. Second generation proteasome inhibitors: carfilzomib and immunoproteasome-specific inhibitors (IPSIs). Curr Cancer Drug Targets. 2011;11(3):285–95. doi: 10.2174/156800911794519725.
  20. Siegel DS, Martin T, Wang M, et al. A phase 2 study of single agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma. Blood. 2012;120(14):2817–25. doi: 10.1182/blood-2012-05-425934.
  21. Dimopoulos MA, Moreau P, Palumbo A, et al. Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study. Lancet Oncol. 2015;17(1):27–38. doi: 10.1016/S1470-2045(15)00464-7.
  22. Avet-Loiseau H, Fonseca R, Siegel D, et al. Efficacy and Safety of Carfilzomib, Lenalidomide, and Dexamethasone Vs Lenalidomide and Dexamethasone in Patients with Relapsed Multiple Myeloma Based on Cytogenetic Risk Status: Subgroup Analysis from the Phase 3 Study Aspire (NCT01080391). Blood. 2015;126(23):731.
  23. Kupperman E, Lee EC, Cao Y, et al. Evaluation of the proteasome inhibitor MLN9708 in preclinical models of human cancer. Cancer Res. 2010;70(5):1970–80. doi: 10.1158/0008-5472.CAN-09-2766.
  24. Lee EC, Fitzgerald M, Bannerman B, et al. Antitumor activity of the investigational proteasome inhibitor MLN9708 in mouse models of B-cell and plasma cell malignancies. Clin Cancer Res. 2011;17(23):7313–23. doi: 10.1158/1078-0432.CCR-11-0636.
  25. San Miguel J, Weisel K, Moreau P, et al. Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, Phase III trial. Lancet Oncol. 2013;14(11):1055–66. doi: 10.1016/S1470-2045(13)70380-2.
  26. Dimopoulos MA, Weisel KC, Song KW, et al. Cytogenetics and long-term survival of patients with refractory or relapsed and refractory multiple myeloma treated with pomalidomide and low-dose dexamethasone. Haematologica. 2015;100(10):1327–33. doi: 10.3324/haematol.2014.117077.
  27. Dimopoulos MA, Leleu X, Palumbo A, et al. Expert panel consensus statement on the optimal use of pomalidomide in relapsed and refractory multiple myeloma. Leukemia. 2014;28(8):1573–85. doi: 10.1038/leu.2014.60.
  28. ИМНОВИД® (IMNOVID®) инструкция по применению [электронный документ]. Доступно по: https://www.vidal.ru/drugs/imnovid__44356. Ссылка активна на 31.07.2017.[IMNOVID® instruction for medical use [Internet]. Available from: https://www.vidal.ru/drugs/imnovid__44356. (accessed 31.07.17) (In Russ)]
  29. Baz RC, Martin TG, Lin H-Y, et al. Randomized multicenter phase 2 study of pomalidomide, cyclophosphamide, and dexamethasone in relapsed refractory myeloma. Blood. 2016;127(21):2561–8. doi: 10.1182/blood-2015-11-682518.
  30. Lacy MQ, LaPlant BR, Laumann KM, et al. Pomalidomide, Bortezomib and Dexamethasone (PVD) for Patients with Relapsed Lenalidomide Refractory Multiple Myeloma (MM). Blood. 2014;124(21):304.
  31. Collins SM, Bakan CE, Swartzel GD, et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma via CS1 ligation: evidence for augmented NK cell function complementing ADCC. Cancer Immunol Immunother. 2013;62(12):1841–9. doi: 10.1007/s00262-013-1493-8.
  32. Veillette A, Guo H. CS1, a SLAM family receptor involved in immune regulation, is a therapeutic target in multiple myeloma. Crit Rev Oncol Hematol. 2013;88(1):168–77. doi: 10.1016/j.critrevonc.2013.04.003.
  33. Lonial S, Richardson P, Mateos M-V, et al. ELOQUENT-2 update: Phase III study of elotuzumab plus lenalidomide/dexamethasone (ELd) vs Ld in relapsed/refractory multiple myeloma (RRMM)—Identifying responders by subset analysis. 2016 ASCO Annual Meeting. Poster 8037. Available from: http://meetinglibrary.asco.org/record/126339/abstract (accessed 31.07.17).
  34. Lin P, Owens R, Tricot G, Wilson CS. Flow cytometric immunophenotypic analysis of 306 cases of multiple myeloma. Am J Clin Pathol. 2004;121(4):482–8. doi: 10.1309/74r4-tb90-buwh-27jx.
  35. de Weers M, Tai YT, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J. Immunol. 2011;186(3):1840–8. doi: 10.4049/jimmunol.1003032.
  36. Lammerts van Bueren J, Jakobs D, Kaldenhoven N, et al. Direct in vitro comparison of daratumumab with surrogate analogs of CD38 antibodies MOR03087, SAR650984 and Ab79. Blood. 2014;124(21):3474.
  37. Overdijk MB, Verploegen S, Bogels M, et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. mAbs. 2015;7(2):311–21. doi: 10.1080/19420862.2015.1007813.
  38. Jansen JHM, Boross P, Overdijk MB, et al. Daratumumab, a human CD38 antibody induces apoptosis of myeloma tumor cells via Fc receptor-mediated crosslinking. Blood. 2012;120(21): Abstract 2974.
  39. Krejcik J, Casneuf T, Nijhof I, et al. Immunomodulatory effects and adaptive immune response to daratumumab in multiple myeloma. Blood. 2015:126(23): Abstract 3037.
  40. Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373(13):1207–19. doi: 10.1056/NEJMoa1506348.
  41. Lonial S, Weiss BM, Usmani SZ, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet. 2016;387(10027):1551–60. doi: 10.1016/S0140-6736(15)01120-4.
  42. Usmani SZ, Weiss BM, Plesner T, et al. Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. Blood. 2016;128(1):37–44. doi: 10.1182/blood-2016-03-705210.
  43. Palumbo A, Chanan-Khan A, Weisel K, et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N Engl J Med. 2016;375:754–66. doi: 10.1056/NEJMoa1606038.