Tyrosine Kinase Inhibitor Resistance in Patients with Chronic Myeloid Leukemia: A 10-Year Study of BCR-ABL Gene Mutation Profile in Russia (2006–2016)

VV Tikhonova1,2, MA Isakov3, VA Misyurin1, YuP Finashutina1,2, LA Kesaeva1,2, NA Lyzhko 1, IN Soldatova2, NN Kasatkina1, EN Misyurina4, AV Misyurin1,2

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

2 GenoTekhnologiya, 104 Profsoyuznaya str., Moscow, Russian Federation, 117485

3 Aston Consulting, 31g Shabolovka str., Moscow, Russian Federation, 115162

4 Municipal Clinical Hospital No. 52, 3 Pekhotnaya str., Moscow, Russian Federation, 123182

For correspondence: Vera Vyacheslavovna Tikhonova, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(967)008-02-84; e-mail: brilfor@mail.ru

For citation: Tikhonova VV, Isakov MA, Misyurin VA, et al. Tyrosine Kinase Inhibitor Resistance in Patients with Chronic Myeloid Leukemia: A 10-Year Study of BCR-ABL Gene Mutation Profile in Russia (2006–2016). Clinical oncohematology. 2018;11(3):227–33.

DOI: 10.21320/2500-2139-2018-11-3-227-233


ABSTRACT

Background. Kinase domain mutations of BCR-ABL gene is the most common cause of tyrosine kinase inhibitor resistance.

Aim. To present the data on prognostic value of BCR-ABL mutation burden in Russian patients over the last 10 years.

Materials & Methods. The study included 1885 chronic myeloid leukemia (CML) patients with tyrosine kinase inhibitor resistance who were followed up from 2006 to 2016. BCR-ABL point mutations in mRNA samples were analyzed by means of polymerase chain reaction and subsequent Sanger sequencing.

Results. In 1257 CML patients with signs of tyrosine kinase inhibitor resistance BCR-ABL expression level was > 1 %. BCRABL mutations were detected in 31.8 % of patients. Total mutation count was 467 (70 mutation types). Total count of patients with mutation-associated tyrosine kinase inhibitor resistance decreased from 36.6 % (2006–2008) to 24.95 % (2013–2016) and to marked decrease of 23.12 % in 2014. Detection rate of imatinib-resistant mutations and F359V mutation was shown to decrease within the period from 2010–2011 to 2014–2015. F317L level, which is responsible for dasatinib resistance, considerably increased in 2015. T315I frequency was the highest in 2014, afterwards it was gradually decreasing. Mutation-associated resistance rates varied by region of the Russian Federation.

Conclusion. The analysis of trends of mutation incidence in patients with CML can be of extreme significance in long-term prognosis of resistance development and in improvement of treatment planning.

Keywords: chronic myeloid leukemia, kinase domain mutations of BCR-ABL gene, targeted therapy, resistance.

Received: January 22, 2018

Accepted: April 16, 2018

Read in PDF 


REFERENCES

  1. Soverini S, Colarossi S, Gnani A, et al. Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: by the GIMEMA Working Party on Chronic Myeloid Leukemia. Clin Cancer Res. 2006;12(24):7374–9. doi: 10.1158/1078-0432.ccr-06-1516.
  2. Baccarani M, Cortes J, Pane F, et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European Leukemia Net. J Clin Oncol. 2009;27(35):6041–51. doi: 10.1200/JCO.2009.25.0779.
  3. Soverini S, Rosti G, Iacobucci I, et al. Choosing the best second-line tyrosine kinase inhibitor in imatinib-resistant chronic myeloid leukemia patients harboring Bcr-Abl kinase domain mutations: how reliable is the IC50? Oncologist. 2011;16(6):868–76. doi: 10.1634/theoncologist.2010-0388.
  4. Овсянникова Е.Г., Капланов К.Д., Клиточенко Т.Ю. и др. Мутационный статус резистентных к иматинибу больных хроническим миелолейкозом. Онкогематология. 2012;4:16–24.[Ovsyannikova EG, Kaplanov KD, Klitochenko TYu, et al. Mutation status of chronic myeloid leukemia patients with imatinib resistance. Onkogematologiya. 2012;4:16–24. (In Russ)]
  5. Patkar N, Ghodke K, Joshi S, et al. Characteristics of BCR-ABL kinase domain mutations in chronic myeloid leukemia from India: not just missense mutations but insertions and deletions are also associated with TKI resistance. Leuk Lymphoma. 2016;57(11):2653–60. doi: 10.3109/10428194.2016.1157868.
  6. Elnahass YH, Mahmoud HK, Ali FT, et al. Abl Kinase Domain Mutations in Imatinib-treated Egyptian Patients with Chronic Myeloid Leukemia. J Leuk. 2013;1(1):106. doi: 10.4172/2329-6917.1000106.
  7. Awidi A, Ababneh N, Magablah A, et al. ABL Kinase Domain Mutations in Patients with Chronic Myeloid Leukemia in Jordan. Genet Test Mol Biomark. 2012;16(11):1317–21. doi: 10.1089/gtmb.2012.0147.
  8. Elias MH, Baba AA, Husin A, et al. Contribution of BCR-ABL kinase domain mutations to imatinib mesylate resistance in Philadelphia chromosome positive Malaysian chronic myeloid leukemia patients. Hematol Rep. 2012;4(4):e23. doi: 10.4081/hr.2012.e23.
  9. Vaidya S, Vundinti BR, Shanmukhaiah C, et al. Evolution of BCR/ABL Gene Mutation in CML Is Time Dependent and Dependent on the Pressure Exerted by Tyrosine Kinase Inhibitor. PLoS One. 2015;10(1):e0114828. doi: 10.1371/journal.pone.0114828.
  10. Челышева Е.Ю., Шухов О.А., Лазарева О.В., Туркина А.Г. Мутации киназного домена гена BCR-ABL при хроническом миелолейкозе. Клиническая онкогематология. 2012;5(1):13–21.[Chelysheva EYu, Shukhov OA, Lazareva OV, Turkina AG. Kinase domain mutations of BCR-ABL gene in patients with chronic myeloid leukemia. Klinicheskaya onkogematologiya. 2012;5(1):13–21. (In Russ)]
  11. Kimura S, Ando T, Kojima K. BCR-ABL Point Mutations and TKI Treatment in CML Patients. J Hematol Transfus. 2014;2(3):1022.
  12. Soverini S, de Benedittis C, Mancini M, Martinelli G. Mutations in the BCR-ABL1 Kinase Domain and Elsewhere in Chronic Myeloid Leukemia. Clin Lymph Myel Leuk. 2015;15(Suppl):S120–8. doi: 10.1016/j.clml.2015.02.035.
  13. Soverini S, De Benedittis C, Papayannidis C, et al. Drug resistance and BCR-ABL kinase domain mutations in Philadelphia chromosome-positive acute lymphoblastic leukemia from the imatinib to the second-generation tyrosine kinase inhibitor era: The main changes are in the type of mutations, but not in the frequency of mutation involvement. 2014;120(7):1002–9. doi: 10.1002/cncr.28522.
  14. Мисюрин А.В., Мисюрина Е.Н., Тихонова В.В. и др. Частота встречаемости мутаций киназного домена гена BCR-ABL у больных хроническим миелолейкозом, резистентных к терапии иматинибом. Российский биотерапевтический журнал. 2016;15(4):102–9. doi: 10.17650/1726-9784-2016-15-4-102-109.[Misyurin AV, Misyurina EN, Tikhonova VV, et al. BCR-ABL gene kinase domain mutation frequency in imatinib resistant chronic myeloid leukemia patients. Rossiiskii bioterapevticheskii zhurnal. 2016;15(4):102–9. doi: 10.17650/1726-9784-2016-15-4-102-109. (In Russ)]
  15. Hughes TP, Saglio G, Quintas-Cardama A, et al. BCR-ABL1 mutation development during first-line treatment with dasatinib or imatinib for chronic myeloid leukemia in chronic phase. Leukemia. 2015;29(9):1832–8. doi: 10.1038/leu.2015.168.
  16. Абдулкадыров К.М., Шуваев В.А., Фоминых М.С. Дженерики иматиниба: мифы и реальность (обзор литературы и собственные данные). Клиническая онкогематология. 2014;7(3):311–6.[Abdulkadyrov KM, Shuvaev VA, Fominykh MS. Imatinib Generics: Myths and Reality (Literature Review and Our Experience). Klinicheskaya onkogematologiya. 2014;7(3):311–6. (In Russ)]
  17. Валиев Т.Т., Левашов А.С., Сенжапова Э.Р. Таргетные препараты в детской онкологии. Онкопедиатрия. 2016;3(1):8–15. doi: 10.15690/onco.v3i1.1524.[Valiev TT, Levashov AS, Senzhapova ER. Targeted Drugs in Pediatric Oncology. Onkopediatriya. 2016;3(1):8–15. doi: 10.15690/onco.v3i1.1524. (In Russ)]

PD-1 Blockade with Nivolumab as a New Immunotherapy for Classical Hodgkin’s Lymphoma

EA Demina

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

For correspondence: Prof. Elena Andreevna Demina, MD, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: drdemina@yandex.ru

For citation: Demina EA. PD-1 Blockade with Nivolumab as a New Immunotherapy for Classical Hodgkin’s Lymphoma. Clinical oncohematology. 2018;11(3):213–19.

DOI: 10.21320/2500-2139-2018-11-3-213-219


ABSTRACT

During the last two decades individualization of programmed treatment combined with intensified chemotherapy has proven to be effective treatment for the majority of classical Hodgkin’s lymphoma (cHL) patients. However, in 10–30 % of cases relapses and resistance to therapy still occur. Further intensification of therapy induces toxicity that leads to decrease in overall survival and quality of life. The standard second-line treatment with high-dose chemotherapy (HDCT) and autologous hematopoietic stem cell transplantation (auto-HSCT) allows for the achievement of long-term 5-year progression-free survival only in 50–60 % of patients with relapsed disease and not more than 40–45 % of patients with refractory disease. Approximately 50 % of patients relapse after HDCT and auto-HSCT. The median overall survival of relapsed patients does not exceed 2 years. Allogeneic HSCT improves treatment results to some extent, but is not an optimal strategy in all patients. A search for new treatment options has been made to improve effectiveness of relapsed and refractory cHL treatment and to reduce toxicity of highly effective programs. А new CD30-targeted conjugate brentuximab vedotin was developed to use anti-CD30 monoclonal antibodies against a specific marker of tumor Reed-Sternberg cells allowing for the transfer of the highly effective antitumor compound of monomethyl auristatin E directly to tumor cells. This drug showed high effectiveness, although failed to provide a complete solution to the problem. The development of anti-PD1 antibody nivolumab opened up new opportunities for cHL treatment. This paper reviews literature information on pharmacological data and antitumor mechanisms of the drug as well as the results of significant international randomised studies.

Keywords: nivolumab, Hodgkin’s lymphoma, relapse, resistance, treatment.

Received: February 5, 2018

Accepted: April 30, 2018

Read in PDF 


REFERENCES

  1. Engert A, Jounes A, Hematologic malignancies: Hodgkin lymphoma. A Comprehensive Overview. 2nd edition. Berlin, Heidelberg: Springer; 2015. pp. 437. doi: 10.1007/978-3-319-12505-3.
  2. Skoetz N, Trelle S, Rancea M, et al. Effect of initial treatment strategy on survival of patients with advanced-stage Hodgkin’s lymphoma: a systematic review and network meta-analysis. Lancet Oncol. 2013;14(10):943–52. doi:1016/s1470-2045(13)70341-3.
  3. Czyz J, Szydlo R, Knopinska-Posluszny W, et al. Treatment for primary refractory Hodgkin’s disease: a comparison of high-dose chemotherapy followed by ASCT with conventional therapy. Bone Marrow Transplant. 2004;33(12):1225–9. doi: 10.1038/sj.bmt.1704508.
  4. Gerrie AS, Power MM, Shepherd JD, et al. Chemoresistance can be overcome with high-dose chemotherapy and autologous stem-cell transplantation for relapsed and refractory Hodgkin lymphoma. Ann Oncol. 2014;25(11):2218–23. doi: 10.1093/annonc/mdu387.
  5. Sureda A, Constans M, Iriondo A, et al. Prognostic factors affecting long-term outcome after stem cell transplantation in Hodgkin’s lymphoma autografted after a first relapse. Ann Oncol. 2005;16(4):625–33. doi: 10.1093/annonc/mdi119.
  6. Brice P, Bouabdallah R, Moreau P, et al. Prognostic factors for survival after high-dose therapy and autologous stem cell transplantation for patients with relapsing Hodgkin’s disease: analysis of 280 patients from the French registry. Societe Francaise de Greffe de Moelle. Bone Marrow Transplant. 1997;20(1):21–6. doi: 10.1038/sj.bmt.1700838.
  7. Crump M. Management of Hodgkin lymphoma in relapse after autologous stem cell transplant. Hematology Am Soc Hematol Educ Program. 2008;2008(1):326–33. doi: 10.1182/asheducation-2008.1.326.
  8. Francisco JA, Cerveny CG, Meyer DL, et al. cAC10-vcMMAE, an antiCD30–monomethyl auristatin E conjugate with potent and selective antitumor activity. Blood. 2003;102(4):1458–65. doi: 10.1182/blood-2003-01-0039.
  9. Sutherland MSK, Sanderson RJ, Gordon KA, et al. Lysosomal Trafficking and Cysteine Protease Metabolism Confer Target-specific Cytotoxicity by Peptide-linked Anti-CD30-Auristatin Conjugates. J Biol Chem. 2006;281(15):10540–7. doi: 10.1074/jbc.M510026200.
  10. Gopal AK, Chen R, Smith SE, et al. Durable remissions in a pivotal phase 2 study of brentuximab vedotin in relapsed or refractory Hodgkin lymphoma. Blood. 2015;125(8):1236–43. doi: 10.1182/blood-2014-08-595801.
  11. Arai S, Fanale M, DeVos S, et al. Defining a Hodgkin lymphoma population for novel therapeutics after relapse from autologous hematopoietic cell transplant. Leuk Lymphoma. 2013;54(11):2531–3. doi: 10.3109/10428194.2013.798868.
  12. Moskowitz CH, Nademanee A, Masszi T, et Brentuximab vedotin as consolidation therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2015;385(9980):1853–62. doi: 10.1016/S0140-6736(15)60165-9.
  13. Boussiotis VA. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N Engl J Med. 2016;375(18):1767–78. doi: 10.1056/NEJMra1514296.
  14. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372(21):2006–17. doi: 10.1056/NEJMoa1414428.
  15. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N Engl J Med. 2016;375(19):1823–33. doi: 10.1056/NEJMoa1606774
  16. Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med. 2015;373(19):1803–13. doi: 10.1056/NEJMoa1510665.
  17. Kuppers R. The biology of Hodgkin’s lymphoma. Nat Rev Cancer. 2009;9(1):15–27. doi: 10.1038/nrc2542.
  18. Yamamoto R, Nishikori M, Kitawaki T, et al. PD-1-PD-1 ligand interaction contributes to immunosuppressive microenvironment of Hodgkin lymphoma. Blood. 2008;111(6):3220–4. doi: 1182/blood-2007-05-085159.
  19. 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.
  20. 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.
  21. Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activity and EBV infection induce PD-L1 in Hodgkin lymphomas and posttransplant lymphoproliferative disorders: implications for targeted therapy. Clin Cancer Res. 2012;18(6):1611–8. doi: 1158/1078-0432.ccr-11-1942.
  22. 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.
  23. Merryman R, Armand Ph. Hodgkin lymphoma and PD-1 blockade: an unfinished story. Ann Lymphoma. 2017;1:4. doi: 10.21037/aol.2017.08.03.
  24. Ansell SM. Nivolumab in the Treatment of Hodgkin Lymphoma. Clin Cancer Res. 2017;23(7):1623–6. doi: 10.1158/1078-0432.CCR-16-1387.
  25. Ferris RL, Blumenschein G, Fayette J, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med. 2016;375(19):1856–67. doi: 10.1056/NEJMoa1602252.
  26. Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single arm, phase 2 trial. Lancet Oncol. 2017;18(3):312–22. doi: 10.1016/S1470-2045(17)30065-7.
  27. Borghaei H, Paz‑Ares L, Horn L, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non–Small-Cell Lung Cancer. N Engl J Med. 2015;373(17):1627–39. doi: 10.1056/NEJMoa1507643.
  28. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non–Small-Cell Lung Cancer. N Engl J Med. 2015;373(2):123–35. doi: 10.1056/NEJMoa1504627.
  29. Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med. 2015;373(19):1803–13. doi: 10.1056/NEJMoa1510665.
  30. Wolchok JD, Rollin L, Larkin J. Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2017;377(25):2503–4. doi: 10.1056/NEJMc1714339.
  31. 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: 10.1056/NEJMoa1411087.
  32. Armand P. Immune checkpoint blockade in hematologic malignancies. Blood. 2015;125(22):3393–400. doi: 10.1182/blood-2015-02-567453.
  33. Kasamon YL, De Carlo RA, Wang Y, et al. FDA Approval Summary: Nivolumab for the Treatment of Relapsed or Progressive Classical Hodgkin Lymphoma. 2017;22(5):585–91. doi: 10.1634/theoncologist.2017-0004.
  34. Cheson BD, Pfistner B, Juweid ME, et al. Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579–86. doi: 10.1200/JCO.2006.09.2403.
  35. Fanale M, Engert A, Younes A. Nivolumab for relapsed/refractory classical Hodgkin lymphoma after autologous transplant: full results after extended follow-up of the phase 2 CheckMate 205 trail. Hematol Oncol. 2017;35:135–6. doi: 10.1002/hon.2437_124.
  36. Majhail NS, Weisdorf DJ, Defor TE, et al. Long-Term Results of Autologous Stem Cell Transplantation for Primary Refractory or Relapsed Hodgkin’s Lymphoma. Biol Blood Marrow 2006;12(10):1065–72. doi: 10.1016/j.bbmt.2006.06.006
  37. Merryman RW, Kim HT, Zinzani PL, et al. Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood. 2017;129(10):1380–8. doi: 10.1182/blood-2016-09-738385.
  38. Saha A, Aoyama K, Taylor PA, et al. Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. Blood. 2013;122(17):3062–73. doi: 10.1182/blood-2013-05-500801.
  39. Ciurea SO, Zhang MJ, Bacigalupo AA, et al. Haploidentical transplant with posttransplant cyclophosphamidevs matched unrelated donor transplant for acute myeloid leukemia. Blood. 2015;126(8):1033–40. doi: 10.1182/blood-2015-04-639831.
  40. Опдиво® [инструкция по медицинскому применению]. Принстон, США: Bristol-Myers Squibb Company. Доступно по: https://www.vidal.ru/drugs/opdivo. Ссылка активна на 30.03.2018.
    [Opdivo® [package insert]. Princeton, NJ, USA: Bristol-Myers Squibb Company. Available from: https://www.vidal.ru/drugs/opdivo. (accessed 30.03.2018) (In Russ)]
  41. Hoppe RT, Advani RH, Ai WZ, et al. NCCN Clinical Practice Guidelines in Oncology. Hodgkin Lymphoma. Version 1.2018. Available from: https://www.nccn.org/professionals/physician_gls/pdf/hodgkins.pdf (accessed 05.04.2018).
  42. Herrera AF, Moskowitz AJ, Bartlett NL, et al. Interim results from a phase 1/2 study of brentuximab vedotin in combination with relapsed or refractory Hodgkin lymphoma. Hematol Oncol. 2017;35:85–6. doi: 10.1002/hon.2437_73.
  43. Ramchandren R, Fanale MA, Rueda A, et al. Nivolumab for Newly Diagnosed Advanced-Stage Classical Hodgkin Lymphoma (cHL): Results from the Phase 2 CheckMate 205 Study. ASH Annual Meeting Abstracts. 2017: Abstract 651.
  44. Mikhailova N, Lepik K, Kondakova E, et al. Regaining the Tumor Control in Relapsed/Refractory Hodgkin Lymphoma after Nivolumab Failure with Addition of Another Antineoplastic Agent. ASH Annual Meeting Abstracts. 2017: Abstract