Stable Chronology of Granulopoiesis under R(G)-DHAP Immunochemotherapy-Induced Cytotoxic Stress in Non-Hodgkin’s Lymphomas

In memory of Academician A.I. Vorob’ev,
Russian Academy of Medical Sciences and Russian Academy of Sciences

KA Sychevskaya, SK Kravchenko, FE Babaeva, AE Misyurina, AM Kremenetskaya, AI Vorob’ev

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

For correspondence: Kseniya Andreevna Sychevskaya, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(910)409-79-44; e-mail: sychevskaya-ka@yandex.ru

For citation: Sychevskaya KA, Kravchenko SK, Babaeva FE, et al. Stable Chronology of Granulopoiesis under R(G)-DHAP Immunochemotherapy-Induced Cytotoxic Stress in Non-Hodgkin’s Lymphomas. Clinical oncohematology. 2021;14(2):204–19. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-204-219


ABSTRACT

Background. Chronology of granulopoiesis based on periodic hematopoiesis model has been thoroughly studied. However, the pattern of influence of chemotherapy- and immunotherapy-induced cytotoxic stress on the development rhythm of a stem cell requires further investigation. The interaction of antitumor drugs with normal hematopoietic cells is relevant for assessing the intensity of chemotherapy adverse events. Besides, there is a demand for studying hematopoiesis under cytotoxic stress to predict immunological reactivity as a condition for efficacy of immunotherapeutic agents, the effect of which is based on cell immunity.

Aim. To study the chronological pattern of leukocyte count dynamics after R(G)-DHAP immunochemotherapy in non-Hodgkin’s lymphomas.

Materials & Methods. The dynamics of leukocyte count changes after R(G)-DHAP immunochemotherapy was analyzed using the data of 39 treatment courses in 19 non-Hodgkin’s lymphomas patients. After 18 out of 39 cycles of treatment granulocyte colony-stimulating factor (G-CSF) was administered to prevent granulocytopenia, in other cases the previously planned hematopoietic stem cell mobilization was performed according to the accepted protocol.

Results. Time to activation of spontaneous granulopoiesis depends neither on G-CSF stimulation, nor on the total dose of growth-stimulating factor and corresponds on average to Day 10 or Day 11 of the break from the last day of immunochemotherapy. The tendency of shorter agranulocytosis duration on prophylactic use of G-CSF is associated with transient hyperleukocytosis at an early stage after completing immunochemotherapy. Regimens with platinum-based drugs, like R(G)-DHAP, are suggested to be combined with immunochemotherapeutic agents in patients with the failure of first-line chemotherapy. The time interval preceding myelopoiesis activation within the first days of the break between the courses is likely to contribute to the initiation of treatment with immunotherapeutic drugs after second-line chemotherapy.

Conclusion. The determination of granulopoiesis dynamics under R(G)-DHAP immunochemotherapy-induced cytotoxic stress enables to plan the optimum G-CSF regimen and to predict the optimum timing of immune antitumor effect combined with chemotherapy.

Keywords: periodic hematopoiesis, mathematical hematopoiesis model, non-Hodgkin’s lymphomas, chemotherapy, immunotherapy, G-CSF, antitumor immunity, R(G)-DHAP.

Received: November 15, 2020

Accepted: February 25, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Foley C, Mackey MC. Dynamic hematological disease: a review. J Math Biol. 2009;58(1–2):285–322. doi: 10.1007/s00285-008-0165-3.
  2. Morley AA. A neutrophil cycle in healthy individuals. Lancet. 1966;2(7475):1220–2. doi: 10.1016/s0140-6736(66)92303-8.
  3. Mackey MC, Glass L. Oscillation and chaos in physiological control systems. Science. 1977;197(4300):287–9. doi: 10.1126/science.267326.
  4. Mackey Cell kinetic status of haematopoietic stem cells. Cell Prolif. 2001;34(2):71–83. doi: 10.1046/j.1365-2184.2001.00195.x.
  5. Pujo-Menjouet L, Mackey MC. Contribution to the study of periodic chronic myelogenous leukemia. Compt Rend Biol. 2004;327(3):235–44. doi: 10.1016/j.crvi.2003.05.004.
  6. Schirm S, Engel C, Loeffler M, Scholz M. Modelling chemotherapy effects on granulopoiesis. BMC Syst Biol. 2014;8(1):138. doi: 10.1186/s12918-014-0138-7.
  7. Dale DC, Bolyard AA, Aprikyan A. Cyclic neutropenia. Semin Hematol. 2002;39(2):89–94. doi: 10.1053/shem.2002.31917.
  8. Levy EJ, Schetman D. Cyclic neutropenia. Arch Dermatol. 1961;84(3):429–33. doi: 10.1001/archderm.1961.01580150075012.
  9. Colijn C, Mackey MC. A mathematical model of hematopoiesis: II. Cyclical neutropenia. J Theor Biol. 2005;237(2):133–46. doi: 10.1016/j.jtbi.2005.03.034.
  10. Horwitz M, Benson KF, Person RE, et al. Mutations in ELA2, encoding neutrophil elastase, define a 21-day biological clock in cyclic haematopoiesis. Nat Genet. 1999;23(4):433–6. doi: 10.1038/70544.
  11. Aprikyan AA, Liles WC, Rodger E, et al. Impaired survival of bone marrow hematopoietic progenitor cells in cyclic neutropenia. Blood. 2001;97(1):147–53. doi: 10.1182/blood.v97.1.147.
  12. Horwitz MS, Corey SJ, Grimes HL, Tidwell T. ELANE mutations in cyclic and severe congenital neutropenia: genetics and pathophysiology. Hematol Oncol Clin N Am. 2013;27(1):19-vii. doi: 10.1016/j.hoc.2012.10.004.
  13. Welte K, Zeidler C, Dale DC. Severe congenital neutropenia. Semin Hematol. 2006;43(3):189–95. doi: 10.1053/j.seminhematol.2006.04.004.
  14. Haurie C, Dale DC, Rudnicki R, Mackey MC. Modeling complex neutrophil dynamics in the grey collie. J Theor Biol. 2000;204(4):505–19. doi: 10.1006/jtbi.2000.2034.
  15. Horwitz MS, Duan Z, Korkmaz B, et al. Neutrophil elastase in cyclic and severe congenital neutropenia. Blood. 2007;109(5):1817–24. doi: 10.1182/blood-2006-08-019166.
  16. Go RS. Idiopathic cyclic thrombocytopenia. Blood Rev. 2005;19(1):53–9. doi: 10.1016/j.blre.2004.05.001.
  17. Zhuge C, Mackey MC, Lei J. Origins of oscillation patterns in cyclical thrombocytopenia. J Theor Biol. 2019;462:432–45. doi: 10.1016/j.jtbi.2018.11.024.
  18. Apostu R, Mackey MC. Understanding cyclical thrombocytopenia: a mathematical modeling approach. J Theor Biol. 2008;251(2):297–316. doi: 10.1016/j.jtbi.2007.11.029.
  19. Colijn C, Mackey MC. A mathematical model of hematopoiesis–I. Periodic chronic myelogenous leukemia. J Theor Biol. 2005;237(2):117–32. doi: 10.1016/j.jtbi.2005.03.033.
  20. Fortin P, Mackey MC. Periodic chronic myelogenous leukaemia: spectral analysis of blood cell counts and aetiological implications. Br J Haematol. 1999;104(2):336–45. doi: 10.1046/j.1365-2141.1999.01168.x.
  21. Morley A, Stohlman F Jr. Cyclophosphamide-induced cyclical neutropenia. An animal model of a human periodic disease. N Engl J Med. 1970;282(12):643–6. doi: 10.1056/NEJM197003192821202.
  22. Kennedy Cyclic leukocyte oscillations in chronic myelogenous leukemia during hydroxyurea therapy. Blood. 1970;35(6):751–60. doi: 10.1182/blood.v35.6.751.751.
  23. Zhuge C, Lei J, Mackey MC. Neutrophil dynamics in response to chemotherapy and G-CSF. J Theor Biol. 2012;293:111–20. doi: 10.1016/j.jtbi.2011.10.017.
  24. Price TH, Chatta GS, Dale DC. Effect of recombinant granulocyte colony-stimulating factor on neutrophil kinetics in normal young and elderly humans. Blood. 1996;88(1):335–40. doi: 10.1182/blood.V88.1.335.335.
  25. Chatta GS, Price TH, Allen RC, Dale DC. Effects of in vivo recombinant methionyl human granulocyte colony-stimulating factor on the neutrophil response and peripheral blood colony-forming cells in healthy young and elderly adult volunteers. Blood. 1994;84(9):2923–9. doi: 10.1182/blood.V84.9.2923.2923.
  26. Dancey JT, Deubelbeiss KA, Harker LA, Finch CA. Neutrophil kinetics in man. J Clin Invest. 1976;58(3):705–15. doi: 10.1172/JCI108517.
  27. Kerrigan DP, Castillo A, Foucar K, et al. Peripheral blood morphologic changes after high-dose antineoplastic chemotherapy and recombinant human granulocyte colony-stimulating factor administration. Am J Clin Pathol. 1989;92(3):280–5. doi: 10.1093/ajcp/92.3.280.
  28. Hakansson L, Hoglund M, Jonsson UB, et al. Effects of in vivo administration of G-CSF on neutrophil and eosinophil adhesion. Br J Haematol. 1997;98(3):603–11. doi: 10.1046/j.1365-2141.1997.2723093.x.
  29. Ohsaka A, Saionji K, Sato N, et al. Granulocyte colony-stimulating factor down-regulates the surface expression of the human leucocyte adhesion molecule-1 on human neutrophils in vitro and in vivo. Br J Haematol. 1993;84(4):574–80. doi: 10.1111/j.1365-2141.1993.tb03130.x.
  30. Mehta HM, Malandra M, Corey SJ. G-CSF and GM-CSF in Neutropenia. J Immunol. 2015;195(4):1341–9. doi: 10.4049/jimmunol.1500861.
  31. Dale DC, Bonilla MA, Davis MW, et al. A randomized controlled phase III trial of recombinant human granulocyte colony-stimulating factor (filgrastim) for treatment of severe chronic neutropenia. Blood. 1993;81(10):2496–502. doi: 10.1182/blood.V81.10.2496.2496.
  32. Shinjo K, Takeshita A, Ohnishi K, Ohno R. Granulocyte colony-stimulating factor receptor at various differentiation stages of normal and leukemic hematopoietic cells. Leuk Lymphoma. 1997;25(1–2):37–46. doi: 10.3109/10428199709042494.
  33. Clark OA, Lyman GH, Castro AA, et al. Colony-stimulating factors for chemotherapy-induced febrile neutropenia: a meta-analysis of randomized controlled trials. J Clin Oncol. 2005;23(18):4198–214. doi: 10.1200/JCO.2005.05.645.
  34. Garcia-Carbonero R, Mayordomo JI, Tornamira MV, et al. Granulocyte colony-stimulating factor in the treatment of high-risk febrile neutropenia: a multicenter randomized trial. J Natl Cancer Inst. 2001;93(1):31–8. doi: 10.1093/jnci/93.1.31.
  35. Maher DW, Lieschke GJ, Green M, et al. Filgrastim in patients with chemotherapy-induced febrile neutropenia. A double-blind, placebo-controlled trial. Ann Intern Med. 1994;121(7):492–501. doi: 10.7326/0003-4819-121-7-199410010-00004.
  36. Mitchell PL, Morland B, Stevens MC, et al. Granulocyte colony-stimulating factor in established febrile neutropenia: a randomized study of pediatric patients. J Clin Oncol. 1997;15(3):1163–70. doi: 10.1200/JCO.1997.15.3.1163.
  37. Trillet-Lenoir V, Green J, Manegold C, et al. Recombinant granulocyte colony stimulating factor reduces the infectious complications of cytotoxic chemotherapy. Eur J Cancer. 1993;29A(3):319–24. doi: 10.1016/0959-8049(93)90376-q.
  38. Crawford J, Ozer H, Stoller R, et al. Reduction by granulocyte colony-stimulating factor of fever and neutropenia induced by chemotherapy in patients with small-cell lung cancer. N Engl J Med. 1991;325(3):164–70. doi: 10.1056/NEJM199107183250305.
  39. Crawford J, Becker PS, Armitage JO, et al. Myeloid Growth Factors, Version 2.2017. NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2017;15(12):1520–41. doi: 10.6004/jnccn.2017.0175.
  40. Aapro MS, Bohlius J, Cameron DA, et al. 2010 update of EORTC guidelines for the use of granulocyte-colony stimulating factor to reduce the incidence of chemotherapy-induced febrile neutropenia in adult patients with lymphoproliferative disorders and solid tumours. Eur J Cancer. 2011;47(1):8–32. doi: 10.1016/j.ejca.2010.10.013.
  41. Crawford J, Caserta C, Roila F, ESMO Guidelines Working Group. Hematopoietic growth factors: ESMO Clinical Practice Guidelines for the applications. Ann Oncol. 2010;21(Suppl 5):v248–v251. doi: 10.1093/annonc/mdq195.
  42. Lawrence SM, Corriden R, Nizet V. The Ontogeny of a Neutrophil: Mechanisms of Granulopoiesis and Homeostasis. Microbiol Mol Biol Rev. 2018;82(1):e00057–17. doi: 10.1128/MMBR.00057-17.
  43. Murphy P. The Neutrophil. Boston: Springer; 1976. pp. 33–67.
  44. Lord BI, Bronchud MH, Owens S, et al. The kinetics of human granulopoiesis following treatment with granulocyte colony-stimulating factor in vivo. Proc Natl Acad Sci USA. 1989;86(23):9499–503. doi: 10.1073/pnas.86.23.9499.
  45. Lie AK, Hui CH, Rawling T, et al. Granulocyte colony-stimulating factor (G-CSF) dose-dependent efficacy in peripheral blood stem cell mobilization in patients who had failed initial mobilization with chemotherapy and G-CSF. Bone Marrow Transplant. 1998;22(9):853–7. doi: 10.1038/sj.bmt.1701463.
  46. van Der Auwera P, Platzer E, Xu ZX, et al. Pharmacodynamics and pharmacokinetics of single doses of subcutaneous pegylated human G-CSF mutant (Ro 25-8315) in healthy volunteers: comparison with single and multiple daily doses of filgrastim. Am J Hematol. 2001;66(4):245–51. doi: 10.1002/ajh.1052.
  47. Morstyn G, Campbell L, Souza LM, et al. Effect of granulocyte colony stimulating factor on neutropenia induced by cytotoxic chemotherapy. Lancet. 1988;1(8587):667–72. doi: 10.1016/s0140-6736(88)91475-4.
  48. Shochat E, Rom-Kedar V, Segel LA. G-CSF control of neutrophils dynamics in the blood. Bull Math Biol. 2007;69(7):2299–338. doi: 10.1007/s11538-007-9221-1.
  49. Shochat E, Rom-Kedar V. Novel strategies for granulocyte colony-stimulating factor treatment of severe prolonged neutropenia suggested by mathematical modeling. Clin Cancer Res. 2008;14(20):6354–63. doi: 10.1158/1078-0432.CCR-08-0807.
  50. Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9(1):181–218. doi: 10.1146/annurev-pathol-020712-164023.
  51. Hayes MP, Enterline JC, Gerrard TL, Zoon KC. Regulation of interferon production by human monocytes: requirements for priming for lipopolysaccharide-induced production. J Leuk Biol. 1991;50(2):176–81. doi: 10.1002/jlb.50.2.176.
  52. Boneberg EM, Hareng L, Gantner F, et al. Human monocytes express functional receptors for granulocyte colony-stimulating factor that mediate suppression of monokines and interferon-γ. Blood. 2000;95(1):270–6. doi: 10.1182/blood.V95.1.270.
  53. de Kleijn S, Langereis JD, Leentjens J, et al. IFN-γ-stimulated neutrophils suppress lymphocyte proliferation through expression of PD-L1. PLoS One. 2013;8(8):e72249. doi: 10.1371/journal.pone.0072249.
  54. Rutella S, Zavala F, Danese S, et al. Granulocyte colony-stimulating factor: a novel mediator of T cell tolerance. J Immunol. 2005;175(11):7085– doi: 10.4049/jimmunol.175.11.7085.
  55. Ali N. Chimeric antigen T cell receptor treatment in hematological malignancies. Blood Res. 2019;54(2):81– doi: 10.5045/br.2019.54.2.81.
  56. Bais S, Bartee E, Rahman MM, et al. Oncolytic virotherapy for hematological malignancies. Adv Virol. 2012;2012:1–8. doi: 10.1155/2012/186512.
  57. Calton CM, Kelly KR, Anwer F, et al. Oncolytic Viruses for Multiple Myeloma Therapy. Cancers (Basel). 2018;10(6):198. doi: 10.3390/cancers10060198.
  58. Matveeva OV, Chumakov PM. Defects in interferon pathways as potential biomarkers of sensitivity to oncolytic viruses. Rev Med Virol. 2018;28(6):e2008. doi: 10.1002/rmv.2008.

Nivolumab in a Primary Refractory Hodgkin’s Lymphoma Patient with Absolute Lymphopenia Prior to Chemotherapy: Literature Review and a Case Report

TI Bogatyreva, AO Afanasov, NA Falaleeva, LYu Grivtsova, AYu Terekhova

AF Tsyb Medical Radiological Research Centre, branch of the NMRC of Radiology, 4 Koroleva str., Obninsk, Kaluga Region, Russian Federation, 249036

For correspondence: Tatyana Ivanovna Bogatyreva, MD, PhD, 4 Koroleva str., Obninsk, Kaluga Region, Russian Federation, 249036; e-mail: bogatyreva@mrrc.obninsk.ru

For citation: Bogatyreva TI, Afanasov AO, Falaleeva NA, et al. Nivolumab in a Primary Refractory Hodgkin’s Lymphoma Patient with Absolute Lymphopenia Prior to Chemotherapy: Literature Review and a Case Report. Clinical oncohematology. 2021;14(2):179–87. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-179-187


ABSTRACT

The paper presents a case report of PET-adapted therapy of primary refractory classical Hodgkin’s lymphoma, stage IIАХ, in a female patient with absolute lymphopenia prior to chemotherapy. It also provides literature review on the choice of clinical management for similar categories of patients. Nivolumab was prescribed to the patient in February 2019 due to Hodgkin’s lymphoma progression after the failure of 4 chemotherapy lines including brentuximab vedotin. A bulk of mediastinal lymph nodes was exposed to radiation. Complete metabolic response was retained 18 months after nivolumab therapy start and 6 months after its discontinuation. The initial lymphopenia in this patient with primary refractory Hodgkin’s lymphoma did not interfere with the realization of full clinical effect of nivolumab.

Keywords: classical Hodgkin’s lymphoma, absolute lymphopenia, chemotherapy-refractory disease, immunotherapy, salvage therapy.

Received: September 9, 2020

Accepted: February 18, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Brockelmann PJ, Sasse S, Engert A. Balancing risk and benefit in early-stage classical Hodgkin lymphoma. Blood. 2018;131(15):1666–78. doi: 10.1182/blood-2017-10-772665.
  2. Богатырева Т.И., Павлов В.В. Лечение лимфомы Ходжкина. В кн.: Терапевтическая радиология: национальное руководство. Под ред. А.Д. Каприна, Ю.С. Мардынского. М.: ГЭОТАР-Медиа, 2018. С. 525–46.
    [Bogatyreva TI, Pavlov VV. Treatment of Hodgkin’s lymphoma. In: Kaprin AD, Mardynskii YuS, eds. Terapevticheskaya radiologiya: natsional’noe rukovodstvo. (Therapeutic radiology: national guidelines.) Moscow: GEOTAR-Media Publ.; 2018. pp. 525–46. (In Russ)]
  3. Sieber M, Engert A, Diehl V. Treatment of Hodgkin’s disease: results and current concepts of the German Hodgkin’s Lymphoma Study Group. Ann Oncol. 2000;11(Suppl 1):81–5. doi: 10.1093/annonc/11.suppl_1.s81.
  4. Spinner MA, Advani RH, Connors JM, et al. New Treatment Algorithms in Hodgkin Lymphoma: Too Much or Too Little? Am Soc Clin Oncol Educ Book. 2018;38:626–36. doi: 10.1200/EDBK_200679.
  5. Eichenauer DA, Aleman BMP, Andre M, et al. on behalf of the ESMO Guidelines Committee. Hodgkin’s lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2018;29(Suppl 4):iv18–iv29. doi: 10.1093/annonc/mdy080.
  6. von Tresckow B, Plutschow A, Fuchs M, et al. Dose-intensification in early unfavorable Hodgkin’s lymphoma: final analysis of the German Hodgkin Study Group HD14 trial. J Clin Oncol. 2012;30(9):907–13. doi: 10.1200/JCO.2011.38.5807.
  7. Демина Е.А. Лимфома Ходжкина. В кн.: Российские клинические рекомендации по диагностике и лечению злокачественных лимфопролиферативных заболеваний. Под ред. И.В. Поддубной, В.Г. Савченко. М., 2018. С. 28–43.
    [Demina EA. Hodgkin lymphoma. In: Poddubnaya IV, Savchenko VG, eds. Rossiiskie klinicheskie rekomendatsii po diagnostike i lecheniyu zlokachestvennykh limfoproliferativnykh zabolevanii. (Russian clinical guidelines on diagnosis and treatment of malignant lymphoproliferative diseases.) Moscow; 2018. pp. 28–43. (In Russ)]
  8. Богатырева Т.И., Терехова А.Ю., Афанасов А.О. и др. Влияние исходного дефицита СD4+ Т-лимфоцитов периферической крови на результаты химиолучевого лечения больных лимфомой Ходжкина. Гематология и трансфузиология. 2019;64(3):317–30. doi: 10.35754/0234-5730-2019-64-3-317-330.
    [Bogatyreva TI, Terekhova AYu, Afanasov AO, et al. Impact of the pre-treatment CD4+ T-lymphocyte deficiency in the peripheral blood on the results of chemoradiotherapy in patients with Hodgkin’s lymphoma. Gematologiya i transfuziologiya. 2019;64(3):317–30. doi: 10.35754/0234-5730-2019-64-3-317-330. (In Russ)]
  9. Andre MPE, Girinsky T, Federico M, et al. Early positron emission tomography response-adapted treatment in stage I and II Hodgkin lymphoma: final results of the randomized EORTC/LYSA/FIL H10 trial. J Clin Oncol. 2017;35(16):1786–94. doi: 10.1200/JCO.2016.68.6394.
  10. Sureda A, Constans M, Iriondo A. 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.
  11. Chen R, Gopal AK, Smith SE, et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood. 2016;128(12):1562–6. doi: 10.1182/blood-2016-02-699850.
  12. Шкляев С.С., Фалалеева Н.А., Богатырева Т.И. и др. Бендамустин в лечении пациентов с рецидивами и рефрактерным течением лимфомы Ходжкина (обзор литературы и собственные данные). Клиническая онкогематология. 2020;13(2):136–49. doi: 10.21320/2500-2139-2020-13-2-136-149.
    [Shklyaev SS, Falaleeva NA, Bogatyreva TI, et al. Bendamustine in the Treatment of Relapsed/Refractory Hodgkin’s Lymphoma: Literature Review and Clinical Experience. Clinical oncohematology. 2020;13(2):136–49. doi: 10.21320/2500-2139-2020-13-2-136-149. (In Russ)]
  13. Bogatyreva TI, Terekhova AY, Shklyaev SS, et al. Long-term treatment outcome of patients with refractory or relapsed Hodgkin’s lymphoma in the anthracycline era: a single-center intention-to-treat analysis. Ann Oncol. 2018;29(Suppl 8):viii364. doi: 10.1093/annonc/mdy286.016.
  14. Cheah CY, Chihara D, Horowitz S, et al. Patients with classical Hodgkin lymphoma experiencing disease progression after treatment with brentuximab vedotin have poor outcomes. Ann Oncol. 2016;27(7):1317–23. doi: 10.1093/annonc/mdw169.
  15. Лепик К.В., Михайлова Н.Б., Кондакова Е.В. и др. Эффективность и безопасность ниволумаба в лечении рецидивирующей и рефрактерной классической лимфомы Ходжкина: опыт ПСПбГМУ им. акад. И.П. Павлова. Онкогематология. 2018;13(4):17–26. doi: 10.17650/1818-8346-2019-13-4-17-26.
    [Lepik KV, Mikhailova NV, Kondakova EV, et al. Efficacy and safety of nivolumab in the treatment of relapsed/refractory classical Hodgkin’s lymphoma: Pavlov First Saint Petersburg State Medical University experience. Oncohematology. 2018;13(4):17–26. doi: 10.17650/1818-8346-2019-13-4-17-26. (In Russ)]
  16. 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.
  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. Yamamoto R, Nishikori M, Kitawaki T, et al. PD-1-PD-1 ligand interaction contributes to immunosuppressive microenvironment of Hodgkin lymphoma. Blood. 2008;111(6):3220–4. doi: 10.1182/blood-2007-05-085159.
  19. Опдиво® (инструкция по медицинскому применению). Принстон, США: Bristol-Myers Squibb Company. Доступно по: https://www.vidal.ru/drugs/opdivo. Ссылка активна на 18.02.2021.
    [Opdivo® (package insert). Princeton, USA: Bristol-Myers Squibb Company. Available from: https://www.vidal.ru/drugs/opdivo. Accessed 18.02.2021. (In Russ)]
  20. Armand P, Engert A, Younes A, et al. Nivolumab for relapsed/refractory classic Hodgkin’s 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.
  21. Hude I, Sasse S, Brockelmann PJ. Leucocyte and eosinophil counts predict progression-free survival in relapsed or refractory classical Hodgkin Lymphoma patients treated with PD1 inhibition. Br J Haematol. 2018;181(6):837–40. doi: 10.1111/bjh.14705.
  22. Hasenclever D, Diehl V, Armitage JO, et al. A Prognostic Score for Advanced Hodgkin’s Disease. N Engl J Med. 1998;339(21):1506–14. doi: 10.1056/NEJM199811193392104.
  23. Демина Е.А., Леонтьева А.А., Тумян Г.С. и др. Оптимизация терапии первой линии у пациентов с распространенными стадиями лимфомы Ходжкина: эффективность и токсичность интенсивной схемы ЕАСОРР-14 (опыт ФГБУ «НМИЦ онкологии им. Н.Н. Блохина» Минздрава России). Клиническая онкогематология. 2017;10(4):443–52. doi: 10.21320/2500-2139-2017-10-4-443-452.
    [Demina EA, Leont’eva AA, Tumyan GS, et al. First-Line Therapy for Patients with Advanced Hodgkin’s Lymphoma: Efficacy and Toxicity of Intensive ЕАСОРР-14 Program (NN Blokhin National Medical Cancer Research Center Data). Clinical oncohematology. 2017;10(4):443–52. doi: 10.21320/2500-2139-2017-10-4-443-452. (In Russ)]
  24. Gallamini A, Tarella C, Viviani S, et al. Early chemotherapy intensification with escalated BEACOPP in patients with advanced-stage Hodgkin lymphoma with a positive interim positron emission tomography/computed tomography scan after two ABVD cycles: long-term results of the GITIL/FIL HD 0607 trial. J Clin Oncol. 2018;36(5):454–622. doi: 10.1200/JCO.2017.75.2543.
  25. Bari A, Marcheselli R, Sacchi S, et al. The classic prognostic factors in advanced Hodgkin’s lymphoma patients are losing their meaning at the time of PET-guided treatments. Ann Hematol. 2020;99(2):277–82. doi: 10.1007/s00277-019-03893-7.
  26. Kumar A, Casulo C, Yahalom J. Brentuximab vedotin and AVD followed by involved-site radiotherapy in early stage, unfavorable risk Hodgkin lymphoma. Blood. 2016;128(11):1458–64. doi: 10.1182/blood-2016-03-703470.
  27. Brockelmann PJ, Goergen H, Keller U, et al. Efficacy of Nivolumab and AVD in Early-Stage Unfavorable Classic Hodgkin Lymphoma: The Randomized Phase 2 German Hodgkin Study Group NIVAHL Trial. JAMA Oncol. 2020;6(6):872. doi: 10.1001/jamaoncol.2020.0750.
  28. Богатырева Т.И., Терехова А.Ю., Шкляев С.С. и др. Исходы лечения больных лимфомой Ходжкина с рефрактерным и рецидивирующим течением: анализ 142 последовательных случаев. Евразийский онкологический журнал. 2020;8(приложение 2):229.
    [Bogatyreva TI, Terekhova AYu, Shklyaev SS, et al. Treatment outcomes in patients with relapsed/refractory Hodgkin’s lymphoma: analysis of 142 successive cases. Evraziiskii onkologicheskii zhurnal. 2020;8(Suppl 2):229. (In Russ)]

Checkpoint Inhibitors and Classical Hodgkin’s Lymphoma: Efficacy and Safety of Pembrolizumab in Relapsed/Refractory Tumor (Experience at the NI Pirogov Russian National Medical Center of Surgery)

VO Sarzhevskii, EA Demina, NE Mochkin, AA Spornik, AA Mamedova, EG Smirnova, AE Bannikova, AA Samoilova, VS Bogatyrev, VYa Melnichenko

NI Pirogov Russian National Medical Center of Surgery, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203

For correspondence: Prof. Vladislav Olegovich Sarzhevskii, MD, PhD, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203; Tel.: +7(495)603-72-17; e-mail: vladsar100@gmail.com

For citation: Sarzhevskii VO, Demina EA, Mochkin NE, et al. Checkpoint Inhibitors and Classical Hodgkin’s Lymphoma: Efficacy and Safety of Pembrolizumab in Relapsed/Refractory Tumor (Experience at the NI Pirogov Russian National Medical Center of Surgery). Clinical oncohematology. 2021;14(1):53–62. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-53-62


ABSTRACT

Background. Checkpoint inhibitors contribute to improving the treatment outcomes in patients with relapsed/refractory classical Hodgkin’s lymphoma (cHL). The paper describes the first generalized experience with pembrolizumab-inducing cHL immunotherapy in Russia. The hallmark of the study is a long follow-up period.

Aim. To retrospectively assess efficacy and safety of pembrolizumab-inducing immunotherapy of relapsed/refractory cHL.

Materials & Methods. The study enrolled 14 cHL patients: 3 men and 11 women aged 24–57 years (median 33 years). Pembrolizumab 200 mg or 2 mg/kg was intravenously administered every 3 weeks. Median pembrolizumab administration number was 27 (max. 52 administrations), median follow-up after immunotherapy onset was 31 months.

Results. Complete response (as best response) was achieved in 8 (57 %) patients, 3 (21 %) patients showed partial response (as best response). Overall objective response was 78 %. Median number of pembrolizumab administrations resulting in better responses to immunotherapy was 4, which corresponded to 3 months of treatment. Maximum number of pembrolizumab administrations before achieving best response was 32. Best response duration (the period from achieving it to disease progression/relapse or to the end-point of data collection in case of sustained response) varied from 3 to 56 months (median 15 months). Most common severe adverse events of grade 3–4 were pulmonary complications. Overall survival for 12, 24, and 36 months was 92.9 %, 85.7 %, and 85.7 %, respectively, and progression-free survival was 76.9 %, 59.3 %, and 37.1 %, respectively; median time before progression was 27.7 months.

Conclusion. The experience with pembrolizumab-inducing immunotherapy of relapsed/refractory cHL in Russia proves the efficacy and relative safety of this treatment approach. Due to long follow-up period a series of crucial practical immunotherapy-related issues were raised, which will need to be dealt with in future studies.

Keywords: сheckpoint inhibitors, immunotherapy, classical Hodgkin’s lymphoma, pembrolizumab.

Received: September 7, 2020

Accepted: December 2, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Ansell S, Lesokhin A, 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.
  2. 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.
  3. 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.
  4. Chen R, Zinzani P, Fanale M, 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.
  5. Zinzani P, Lee H, Armand P, et al. Three-Year Follow-up of Keynote-087: Pembrolizumab Monotherapy in Relapsed/Refractory Classic Hodgkin Lymphoma. 2019;134(Suppl_1):240. doi: 10.1182/blood-2019-127280.
  6. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059–68. doi: 10.1200/JCO.2013.54.8800.
  7. Cheson BD, Ansell S, Schwartz L, et al. Refinement of the Lugano Classification lymphoma response criteria in the era of immunomodulatory therapy. Blood. 2016;128(21):2489–96. doi: 10.1182/blood-2016-05-718528.
  8. Younes A, Hilden P, Coiffier B, et al. International Working Group consensus response evaluation criteria in lymphoma (RECIL 2017). Ann 2017;28(7):1436–47. doi: 10.1093/annonc/mdx097.
  9. Лепик К.В. Эффективность и безопасность PD-1 ингибитора (ниволумаба) в лечении резистентной и рецидивирующей лимфомы Ходжкина: Автореф. дис. … канд. мед. наук. СПб., 2019.
    [Lepik KV. Effektivnost i bezopasnost PD-1 ingibitora (nivolumaba) v lechenii rezistentnoi i retsidiviruyushchei limfomy Khodzhkina. (Efficacy and safety of PD-1 inhibitor (nivolumab) in the treatment of relapsed/refractory Hodgkin’s lymphoma.) [dissertation] Saint Petersburg; 2019. (In Russ)]
  10. Mokrane F-Z, Chen A, Schwartz LH, et al. Performance of CT Compared with 18F-FDG PET in Predicting the Efficacy of Nivolumab in Relapsed or Refractory Hodgkin Lymphoma. Radiology. 2020;295(3):651–61. doi: 10.1148/radiol.2020192056.
  11. Ansell S, Armand Р, Timmerman J, et al. Nivolumab re-treatment in patients with relapsed/refractory Hodgkin lymphoma: safety and efficacy outcomes from a phase 1 clinical trial. Poster presentation at the 10th International Symposium on Hodgkin Lymphoma (ISHL); October 22–25, 2016; Cologne, Germany. Abstract 583/P090.
  12. Chen R, Zinzani PL, Lee HJ, et al. Pembrolizumab in relapsed or refractory Hodgkin lymphoma: Two-year follow-up of KEYNOTE-087. Blood. 2019;134(14):114–53. doi: 10.1182/blood.2019000324.
  13. Manson G, Brice P, Herbaux C, et al. Efficacy of anti-PD1 Re-Treatment in Patients With Hodgkin Lymphoma Who Relapsed After anti-PD1 Discontinuation. Haematologica. 2020;105. [Epub ahead of print] doi: 10.3324/haematol.2019.242529.
  14. Armand P, Kuruvilla J, Michot J-M, et al. KEYNOTE-013 4-year follow-up of pembrolizumab in classical Hodgkin lymphoma after brentuximab vedotin failure. Blood Adv. 2020;4(12):2617–22. doi: 10.1182/bloodadvances.2019001367.
  15. Domingo-Domenech E, Sureda А. Treatment of Hodgkin Lymphoma Relapsed after Autologous Stem Cell Transplantation. J Clin Med. 2020;9(5):1384. doi: 10.3390/jcm9051384.

The Use of Checkpoint Inhibitors in Classical Hodgkin’s Lymphoma during the COVID-19 Pandemic (Pirogov Medical Center’s Experience)

VO Sarzhevskii, EA Demina, NE Mochkin, AA Spornik, AA Mamedova, EG Smirnova, AE Bannikova, AA Samoilova, VS Bogatyrev, OYu Bronov, YuA Abovich, VYa Melnichenko

NI Pirogov Russian National Medical Center of Surgery, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203

For correspondence: Vladislav Olegovich Sarzhevskii, MD, PhD, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203; Tel.: +7(495)603-72-17; e-mail: vladsar100@gmail.com

For citation: Sarzhevskii VO, Demina EA, Mochkin NE, et al. The Use of Checkpoint Inhibitors in Classical Hodgkin’s Lymphoma during the COVID-19 Pandemic (Pirogov Medical Center’s Experience). Clinical oncohematology. 2020;13(3):307–15 (In Russ).

DOI: 10.21320/2500-2139-2020-13-3-307-315


ABSTRACT

Background. Currently, there are neither systematic data nor clinical guidelines for checkpoint inhibitor immunotherapy in cancer patients in the context of the COVID-19 pandemic. In this respect classical Hodgkin’s lymphoma (cHL) is no exception. The article deals with the experience of Pirogov Medical Center (NI Pirogov Russian National Medical Center of Surgery) in PD-1-inhibitor immunotherapy in relapsed/refractory cHL in the context of the COVID-19 pandemic. The authors endeavour to cover matters of current interest concerning immunotherapy and differential diagnosis of pulmonary adverse events emerging in the context of new realities in providing medical care to cancer patients.

Aim. To assess feasibility and safety of checkpoint inhibitor immunotherapy in relapsed/refractory cHL in the context of the COVID-19 pandemic.

Materials & Methods. This is a retrospective analysis of adverse events of therapy and COVID-19 mortality, and incidence in 50 cHL patients who received immunotherapy at the Pirogov Medical Center in the period of spring COVID-19 pandemic in 2020.

Results. During the reported period (from March 11, 2020, when the COVID-19 pandemic was declared, to May 25, 2020) the group of 50 cHL patients showed relatively low overall incidence rate of newly diagnosed immune-mediated adverse events (14 %; n = 7). Severe adverse events were identified in 2 (4 %) patients. Bacterial infection incidence was 6 % (n = 3). Clinical signs of corona virus confirmed by subsequent laboratory COVID-19 tests were observed in 2 (4 %) patients. One patient died due to the non-COVID-19-associated reason. The main issue the center’s staff was faced with was the need for differential diagnosis between drug-induced (as well as immune-mediated) pulmonitis and COVID-19-associated pneumonia.

Conclusion. The retrospective analysis reveals that PD-1-inhibitor immunotherapy in cHL patients during the COVID-19 pandemic is a feasible method of therapy, but it requires high awareness. Special focus should be given to clinical and radiological similarities of COVID-19-associated pneumonia and pulmonitis as a complication of immunotherapy.

Keywords: classical Hodgkin’s lymphoma, immunotherapy, PD-1-inhibitors, COVID-19 pandemic.

Received: May 29, 2020

Accepted: June 28, 2020

Read in PDF


REFERENCES

  1. Coronavirus W.H.O. WHO; 2020. COVID-19. [Internet] Available from: https://who.sprinklr.com. (accessed 28.05.2020).

  2. Стопкороновирус.рф. [электронный документ] Доступно по: https://стопкоронавирус.рф. Ссылка активна на 28.05.2020.[Stopcoronavirus.rf. [Internet] Available from: https://стопкоронавирус.рф (accessed 28.05.2020) (In Russ)]

  3. Liang W, Guan W, Chen R, et al. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. 2020;21(3):335–7. doi: 10.1016/S1470-2045(20)30096-6.

  4. Zhang L, Zhu F, Xie L, et al. Clinical characteristics of COVID-19-infected cancer patients: a retrospective case study in three hospitals within Wuhan, China. Ann Oncol. 2000 (in press). doi: 10.1016/j.annonc.2020.03.296.

  5. Petrelli F, Ardito R, Borgonovo K, et al. Haematological toxicities with immunotherapy in patients with cancer: a systematic review and meta-analysis. Eur J Cancer. 2018;103:7–16. doi: 10.1016/j.ejca.2018.07.129.

  6. Finkel I, Sternschuss M, Wollner M, et al. Immune-related neutropenia following treatment with immune checkpoint inhibitors. J Immunother. 2020;43(2):67–74. doi: 10.1097/CJI.0000000000000293.

  7. Choi J, Lee SY. Clinical characteristics and treatment of immune-related adverse events of immune checkpoint inhibitors. Immune Netw. 2020;20(1):e9. doi: 10.4110/in.2020.20.e9.

  8. Stroud CR, Hegde A, Cherry C, et al. Tocilizumab for the management of immune mediated adverse events secondary to PD-1 blockade. J Oncol Pharm Pract. 2019;25(3):551–7. doi: 10.1177/1078155217745144.

  9. Xu X, Han M, Li T, et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc Nat Acad Sci. 2020;117(20):10970–5. doi: 10.1073/pnas.2005615117.

  10. Ansell S, Lesokhin A, 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.

  11. 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.

  12. Chen R, Zinzani P, Fanale M, 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.

  13. D’Souza A, Jaiyesimi I, Trainor L, et al. Granulocyte Colony-Stimulating Factor Administration: Adverse Events. Transfus Med Rev. 2008;22(4):280–90. doi: 10.1016/j.tmrv.2008.05.005.

  14. Rochefoucauld J, Noel N, Lambotte O. Management of Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitors in Cancer Patients: A Patient-Centred Approach. Intern Emerg Med. 2020. doi: 10.1007/s11739-020-02295-2.

  15. Diamantopoulos P, Gaggadi M, Kassi E, et al. Late-onset Nivolumab-Mediated Pneumonitis in a Patient With Melanoma and Multiple Immune-Related Adverse Events. Melanoma Res. 2017;27(4):391–5. doi: 10.1097/CMR.0000000000000355.

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)]