Familial Aggregation in Hodgkin’s Lymphoma

SV Shakhtarina, AA Danilenko, NA Falaleeva

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

For correspondence: Svetlana Vasilevna Shakhtarina, MD, PhD, 4 Koroleva str., Obninsk, Kaluga Region, Russian Federation, 249036; Tel.: +7(484)399-31-01; e-mail: shakhtarina@mrrc.obninsk.ru

For citation: Shakhtarina SV, Danilenko AA, Falaleeva NA. Familial Aggregation in Hodgkin’s Lymphoma. Clinical oncohematology. 2021;14(2):193–7. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-193-197


ABSTRACT

Background. Genetic predisposition to Hodgkin’s lymphoma (HL) can be directly evidenced through observing familial HL. The literature data available on the familial aggregation samples of HL are extremely limited.

Aim. To systemize and assess observation data on familial aggregation in patients with classical HL based on the sequence of tumor development in blood relatives.

Materials & Methods. Data on families with HL diagnosed more than in one member were gathered from 4700 HL patients, who received chemotherapy from 1970 to 2019 at the AF Tsyb Medical Radiological Research Centre.

Results. Among the blood relatives 27 HL cases were identified, which amounted to 0.57 % of the total of 4700 patients. The families were arranged into four groups: group I with HL diagnosis in a child born before HL detection and treatment of a parent (15 families); group II with HL diagnosis in a child born after HL treatment of a parent (4 families); group III with HL diagnosis in several children of a family with lymphoma-free parents (6 families); group IV — other categories (2 families). The total number of HL patients was 54. Group I comprised 30 patients (15 children and 15 parents), group II included 8 parents (4 daughters and 4 mothers), group III consisted of 12 patients, and group IV included 4 patients.

Conclusion. The proportion of patients with familial aggregation of HL was 0.57 %. The age of all 54 HL patients enrolled in the study corresponded to the first age peak of HL onset. In the pairs “parent-child” children born before HL treatment of a parent accounted for 78.9 % and children born after HL treatment of a mother accounted for 21.1 % (all of them were girls). There were no HL cases in children born after HL treatment of a father. The data obtained show no effect of a parent’s chemotherapy on the occurrence of HL in a child. This is confirmed by the HL cases of siblings whose parents never received HL treatment as well as by the diagnosis of this malignant tumor first in a grandson and then in his grandmother.

Keywords: Hodgkin’s lymphoma, familial aggregation, children, parents.

Received: October 15, 2020

Accepted: February 1, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Cerhan JR, Slager SL. Familial predisposition and genetic risk factors for lymphoma. Blood. 2015;126(20):2265–73. doi: 10.1182/blood-2015-04-537498.
  2. Kharazmi E, Fallah M, Pukkala E, et al. Risk of familial classical Hodgkin lymphoma by relationship, histology, age and sex: a joint study from five Nordic countries. Blood. 2015;126(17):1990–5. doi: 10.1182/blood-2015-04-639781.
  3. Hemminki K, Li X, Czene K. Familial risk of cancer: data for clinical counseling and cancer genetics. Cancer. 2004;108(1):109–14. doi: 10.1002/ijc.11478.
  4. Paltiel О, Schmit Т, Adler B, et al. The incidence of lymphoma in first-degree relatives of patients with Hodgkin disease and non-Hodgkin lymphoma. Cancer. 2000;88(10):2357–66. doi: 10.1002/(sici)1097-0142(20000515)88:10<2357::aid-cncr21>3.0.co;2-3.
  5. Alteri A, Hemminki K. The familial risk of Hodgkin’s lymphoma ranks among the highest in the Swedish Family-Cancer Database. Leukemia. 2006;20(11):2062–3. doi: 10.1038/sj.leu.2404378.
  6. Звягинцева Д.А. Пути улучшения комбинированного лечения лимфомы Ходжкина у детей и подростков: Дис. … канд. мед. наук. СПб., 2017.
    [Zvyagintseva DA. Puti uluchsheniya kombinirovannogo lecheniya limfomy Khodzhkina u detei i podrostkov. (The ways to improve the combined treatment of Hodgkin’s lymphoma in children and young adults.) [dissertation] Saint Petersburg; 2017. (In Russ)]
  7. Goldin LR, Pfeiffer RM, Gridley G, et al. Familial aggregation of Hodgkin lymphoma and related tumors. Cancer. 2000;100(9):1902–8. doi: 10.1002/cncr.20189.
  8. Mack TM, Cozen W, Shibata DK, et al. Concordance for Hodgkin’s Disease in Identical Twins Suggesting Genetic Susceptibility to the Young-Adult Form of the Disease. N Engl J Med. 1995;332(7):413–8. doi: 10.1056/NEJM199502163320701.
  9. Hemminki K, Czene K. Attributable risks of familial cancer from the Family-Cancer database. Cancer Epidemiol Biomark Prev. 2002;11(12):1638–44.
  10. Goldin LR, McMaster ML, Ter-Minassian, et al. A genome screen of families at high risk for Hodgkin’s lymphoma: evidence for a susceptibility gene of chromosome. J Med Genet. 2005;42(7):595–601. doi: 10.1136/jmg.2004.027433.
  11. Landgren O, Kerstan KF, Gridley G, et al. Familial clustering of Hodgkin’s lymphoma and multiple sclerosis. J Natl Cancer Ins. 2005;97(7):543–4. doi: 10.1093/jnci/dji092.

 

 

Ovary Lesions in Classical Hodgkin’s Lymphoma

SYu Smirnova1, TN Moiseeva1, LS Al-Radi1, AB Fedorov2, SA Makhinya1, NV Volkov1, GA Yatsyk1, IA Shupletsova1, AM Kovrigina1

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

2 MEDSI Clinical Hospital, block 2 с1А Pyatnitskoe sh., 6th km, Moscow Region, Russian Federation, 123464

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

For citation: Smirnova SYu, Moiseeva TN, Al-Radi LS, et al. Ovary Lesions in Classical Hodgkin’s Lymphoma. Clinical oncohematology. 2021;14(2):188–92. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-188-192


ABSTRACT

Hodgkin’s lymphoma is a tumor characterized by predominant lesions in lymph nodes. Primary extranodal lesions are exceedingly rare. In the now available world literature, there are only single observations of classical Hodgkin’s lymphoma with ovary involvements. The present paper reports the diagnosis and treatment of a patient with classical Hodgkin’s lymphoma with ovary involvements confirmed by histological and immunohistochemical analyses.

Keywords: Hodgkin’s lymphoma, ovaries.

Received: September 22, 2020

Accepted: February 16, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Taheri MR, Dighe MK, Kolokythas O, et al. Multifaceted genitourinary lymphoma. Curr Probl Diagn Radiol. 2008;37(2):80–93. doi: 10.1067/j.cpradiol.2007.08.010.
  2. Lin PS, Gershenson DM, Bevers MW, et al. The current status of surgical staging of ovarian serous borderline tumors. Cancer. 1999;85(4):905–11. doi: 10.1002/(sici)1097-0142(19990215)85:4<905::aid-cncr19>3.0.co;2-8.
  3. Matasar MJ, Zelenetz AD. Overview of lymphoma diagnosis and management. Radiol Clin N Am. 2008;46(2):175–98. doi: 10.1016/j.rcl.2008.03.005.
  4. Vang R, Medeiros LJ, Fuller GN, et al. Non-Hodgkin’s lymphoma involving the gynecologic tract. A review of 88 cases. Adv Anat Pathol. 2001;8(4):200–17. doi: 10.1097/00125480-200107000-00002.
  5. Elharroudi T, Ismaili N, Errihani H, A Jalil. Primary lymphoma of the ovary. J Cancer Res Ther. 2008;4(4):195–6. doi: 10.4103/0973-1482.44291.
  6. Vang R, Medeiros LJ, Warnke RA, et al. Ovarian non-Hodgkin’s lymphoma: A clinicopathologic study of eight primary cases. Mod Pathol. 2001;14(11):1093–9. doi: 10.1038/modpathol.3880442.
  7. Мангасарова Я.К., Магомедова А.У., Ковригина А.М. и др. Экстрамедиастинальное поражение у больных первичной медиастинальной В-крупноклеточной лимфомой. Онкогематология. 2018;13(1):21–8.
    [Mangasarova YaK, Magomedova AU, Kovrigina AM, et al. Extramediastinal lesion in patients with primary mediastinal large B-cell Onkogematologiya. 2018;13(1):21–8. (In Russ)]
  8. Jackson H, Parker F. Hodgkin’s disease. II. Pathology. N Eng J Med. 1944;231:35–44.
  9. Heller EL, Palon W. Ovarian involvement in Hodgkin’s disease. Arch Path. 1946;41:282–9.
  10. Bare WW, McCloskey F. Primary Hodgkin’s disease of the ovary (report of a case). Obstet Gynec. 1961;17:477–80.
  11. Chorlton I, Norris HJ, King FM. Malignant reticuloendothelial disease involving the ovary as primary manifestation: a series of 19 lymphomas and 1 granulocytic sarcoma. Cancer. 1974;34(2):397–407. doi: 10.1002/1097-0142(197408)34:2<397::aid-cncr2820340225>3.0.co;2-0.
  12. Bittinger SE, Nazaretian SP, Gook DA, et al. Detection of Hodgkin lymphoma within ovarian tissue. Fertil Steril. 2011;95(2):803e3–803e6. doi: 10.1016/j.fertnstert.2010.07.1068.
  13. Khan MA, Dahill SW, Stewart KS. Primary Hodgkin’s disease of the ovary. Case report. Br J Obstet Gynaecol. 1986;93(12):1300–1. doi: 10.1111/j.1471-0528.1986.tb07870.x.
  14. Swerdlow SH., Campo E, Harris NL, et al. World Health Organization classification of tumors of haematopoietic lymphoid tissues. Revised 4th edition. Lyon: IARC Press; 2017.
  15. Schwab U, Stein H, Gerdes J, et al. Production of a monoclonal antibody specific for Hodgkin and Sternberg-Reed cells of Hodgkin’s disease and a subset of normal lymphoid cells. Nature. 1982;299(5878):65–7. doi: 10.1038/299065a0.
  16. Алгоритмы диагностики и протоколы лечения заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2018. Том 2. С. 41–56.
    [Savchenko VG, ed. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. (Diagnostic algorithms and treatment protocols in hematological diseases.) Moscow: Praktika Publ.; 2018. Vol. 2. pр. 41–56. (In Russ)]
  17. Lassmann S, Gerlach UV, Technau-Ihling K, et al. Application of BIOMED-2 primers in fixed and decalcified bone marrow biopsies: analysis of immunoglobulin H receptor rearrangements in B-cell non-Hodgkin’s lymphomas. J Mol Diagn. 2005;7(5):582–91. doi: 10.1016/S1525-1578(10)60591-0.
  18. Сидорова Ю.В., Рыжикова Н.В., Смирнова С.Ю. и др. Определение В-клеточной клональности при лимфоме Ходжкина. Клиническая онкогематология. 2014;7(1):63–6.
    [Sidorova YuV, Ryzhikova NV, Smirnova SYu. Determination of B­cell clonality in Hodgkin’s lymphoma. Klinicheskaya onkogematologiya. 2014;7(1):63–6. (In Russ)]
  19. Ferrozzi F, Catanese C, Uccelli M, Bassi P. Ovarian lymphoma: Findings with ultrasonography, computerized tomography and magnetic resonance. Radio Med (Torino). 1998;95(5):493–7.
  20. Nelson GA, Dockerty MB, Pratt JH, ReMine WH. Malignant lymphoma involving the ovaries. Am J Obstet Gynecol. 1958;76(4):861–71. doi: 10.1016/0002-9378(58)90022-x.
  21. Skodras G, Fields V, Kragel PJ. Ovarian lymphoma and serous carcinoma of low malignant potential arising in the same ovary: A case report with review of literature of 14 primary ovarian lymphomas. Arch Pathol Lab Med. 1994;118(6):647–50.

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)]

Cytogenetic and Molecular Genetic Abnormalities of CIITA Gene in Patients with Primary Mediastinal (Thymic) Large B-Cell Lymphoma

SA Kuznetsova, VL Surin, YaK Mangasarova, TYu Novikova, LA Grebenyuk, AU Magomedova, SK Kravchenko, OS Pshenichnikova, AM Sergeeva, TN Obukhova

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

For correspondence: Svetlana Aleksandrovna Kuznetsova, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(985)170-61-83; e-mail: svetakuznetsova83@mail.ru

For citation: Kuznetsova SA, Surin VL, Mangasarova YaK, et al. Cytogenetic and Molecular Genetic Abnormalities of CIITA Gene in Patients with Primary Mediastinal (Thymic) Large B-Cell Lymphoma. Clinical oncohematology. 2021;14(2):173–8. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-173-178


ABSTRACT

Background. Primary mediastinal (thymic) large B-cell lymphoma (PMBCL) is an aggressive malignant lymphoproliferative disease which accounts for 2–3 % of all non-Hodgkin’s lymphomas. In 40 % of PMBCL cases rearrangements of the MHC class II activator, i.e. CIITA gene, are observed. CIITA abnormalities lead to decreasing protein expression and surface expression of MHC class II, which results in lack of adaptive cell immunity targeted at tumor cells.

Aim. To assess the rate and spectrum of cytogenetic and molecular genetic abnormalities of CIITA gene in PMBCL patients.

Materials & Methods. The study enrolled 37 patients with diagnosed PMBCL: 10 men and 27 women aged 21–61 years (median of 31 years). Sanger sequencing was performed in 36 patients. In 20 patients CIITA/16p13.13 FISH and in 15 patients standard cytogenetic analysis were carried out.

Results. In 3 (8.3 %) out of 36 patients the sequencing method detected mutations impairing CIITA gene function, as well as microdeletion in exon 1, deletion and nucleotide substitution in a splice donor site. Multiple somatic variations in intron 1 were identified in 21 (58.3 %) patients: in 11 (52.4 %) cases there were deletions and single nucleotide variants (SNV); the other 10 (47.6 %) patients showed only SNVs. In 13 (61.9 %) out of 21 cases the abnormalities of promoter IV and/or alternative exon 1 were observed. In 5 (25 %) out of 20 patients the FISH assay identified CIITA gene translocation. Standard cytogenetic analysis detected complex karyotype in 7 (46.6 %) out of 15 patients. The comparison of data showed hypermutagenesis in 8 out of 10 patients with FISH-detected chromosome aberrations, and in 3 (37.5 %) of them complex karyotype aberrations were found as well.

Conclusion. Molecular genetic methods identified different somatic variations in CIITA gene affecting its functionally important regions, which can be of special interest for further studying the biology of tumors, including PMBCL.

Keywords: primary mediastinal (thymic) B-cell large cell lymphoma, CIITA, FISH, chromosomal aberrations, Sanger sequencing.

Received: December 15, 2020

Accepted: March 11, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Cazals-Hatem D, Lepage E, Brice P, et al. Primary Mediastinal Large B-Cell Lymphoma A Clinicopathologic Study of 141 Cases Compared with 916 Nonmediastinal Large B-cell Lymphomas, a GELA (“Groupe d’Etude des Lymphomes de l’Adulte”) Study. Am J Surg Pathol. 1996;20(7):877–88. doi: 10.1097/00000478-199607000-00012.
  2. Мангасарова Я.К., Магомедова А.У. Нестерова Е.С. и др. Первые результаты терапии первичной медиастинальной В-крупноклеточной лимфомы по программе R-DA-EPOCH-21. Терапевтический архив. 2016;88(7):37–42. doi: 10.17116/terarkh201688737-42.
    [Mangasarova YaK, Magomedova AU, Nesterova ES, et al. Therapy for primary mediastinal large B-cell lymphoma in accordance with the R-DA-EPOCH-21 program: the first results. Terapevticheskii arkhiv. 2016;88(7):37–42. doi: 10.17116/terarkh201688737-42. (In Russ)]
  3. Steidl C, Gascoyne RD. The molecular pathogenesis of primary mediastinal large B-cell lymphoma. Blood. 2011;118(10):2659–69. doi: 10.1182/blood-2011-05-326538.
  4. Masternak K, Muhlethaler-Mottet A, Villard J, et al. CIITA is a transcriptional coactivator that is recruited to MHC class II promoters by multiple synergistic interactions with an enhanceosome complex. Genes Dev. 2000;14(9):1156–66. doi: 10.1101/gad.14.9.1156.
  5. Scholl T, Mahanta SK, Strominger JL. Specific complex formation between the type II bare lymphocyte syndrome-associated transactivators CIITA and RFX5. Proc Natl Acad Sci USA. 1997;94(12):6330–4. doi: 10.1073/pnas.94.12.6330.
  6. Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: Implications for tumor immunotherapy. Cancer Immunol Immunother. 2005;54(4):307–14. doi: 10.1007/s00262-004-0593-x.
  7. Steidl C, Shah SP, Woolcock BW, et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature. 2011;471(7338):377–83. doi: 10.1038/nature09754.
  8. Rimsza LM, Roberts RA, Miller TP, et al. Loss of MHC class II gene and protein expression in diffuse large B-cell lymphoma is related to decreased tumor immunosurveillance and poor patient survival regardless of other prognostic factors: A follow-up study from the Leukemia and Lymphoma Molecular. Blood. 2004;103(11):4251–8. doi: 10.1182/blood-2003-07-2365.
  9. 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.
  10. Jiang Y, Soong TD, Wang L, et al. Genome-wide detection of genes targeted by Non-Ig somatic hypermutation in lymphoma. PLoS One. 2012;7(7):e40332. doi: 10.1371/journal.pone.0040332.
  11. Khodabakhshi AH, Morin RD, Fejes AP, et al. Recurrent targets of aberrant somatic hypermutation in lymphoma. Oncotarget. 2012;3(11):1308–19. doi: 10.18632/oncotarget.653.
  12. Mottok A, Woolcock B, Chan FC, et al. Genomic Alterations in CIITA Are Frequent in Primary Mediastinal Large B Cell Lymphoma and Are Associated with Diminished MHC Class II Expression. Cell Rep. 2015;13(7):1418–31. doi: 10.1016/j.celrep.2015.10.008.
  13. Muhlethaler-Mottet A, Otten LA, Steimle V, et al. Expression of MHC class II molecules in different cellular and functional compartments is controlled by differential usage of multiple promoters of the transactivator CIITA. EMBO J. 1997;16(10):2851–60. doi: 10.1093/emboj/16.10.2851.
  14. Roberts RA, Wright G, Rosenwald AR, et al. Loss of major histocompatibility class II gene and protein expression in primary mediastinal large B-cell lymphoma is highly coordinated and related to poor patient survival. Blood. 2006;108(1):311–8. doi: 10.1182/blood-2005-11-4742.
  15. Balzarotti M, Santoro A. Checkpoint inhibitors in primary mediastinal B-cell lymphoma: a step forward in refractory/relapsing patients? Ann Transl Med. 2020;8(16):1035. doi: 10.21037/atm.2020.04.06.
  16. Rosenwald A, Wright G, Leroy K, et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J Exp Med. 2003;198(6):851–62. doi: 10.1084/jem.20031074.
  17. Miao Y, Medeiros LJ, Li Y, et al. Genetic alterations and their clinical implications in DLBCL. Nat Rev Clin Oncol. 2019;16(10):634–52. doi: 10.1038/s41571-019-0225-1.
  18. Joos S, Otano-Joos M.I, Ziegler S, et al. Primary mediastinal (thymic) B-cell lymphoma is characterized by gains of chromosomal material including 9p and amplification of the REL gene. Blood. 1996;87(4):1571–8.
  19. Feuerhake F, Kutok JL, Monti S, et al. NFκB activity, function, and target-gene signatures in primary mediastinal large B-cell lymphoma and diffuse large B-cell lymphoma subtypes. Blood. 2005;106(4):1392–9. doi: 10.1182/blood-2004-12-4901.
  20. Guiter C, Dusanter-Fourt I, Copie-Bergman C, et al. Constitutive STAT6 activation in primary mediastinal large B-cell lymphoma. Blood. 2004;104(2):543–9. doi: 10.1182/blood-2003-10-3545.
  21. Lees C, Keane C, Gandhi MK, et al. Biology and therapy of primary mediastinal B-cell lymphoma: current status and future directions. Br J Haematol. 2019;185(1):25–41. doi: 10.1111/bjh.15778.

Acute Myeloid Leukemia as Second Tumor in a Patient with Burkitt’s Lymphoma: Literature Review and a Case Report

TT Valiev1, TYu Pavlova1, AM Kovrigina2, IN Serebryakova1

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

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

For correspondence: Timur Teimurazovich Valiev, MD, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: timurvaliev@mail.ru

For citation: Valiev TT, Pavlova TYu, Kovrigina AM, Serebryakova IN. Acute Myeloid Leukemia as Second Tumor in a Patient with Burkitt’s Lymphoma: Literature Review and a Case Report. Clinical oncohematology. 2021;14(2):167–72. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-167-172


ABSTRACT

The application of highly effective tumor treatment protocols in children and increasing number of patients healed resulted in a growing focus on long-term effects of chemotherapy. One of the most dangerous complications of a first malignant neoplasm (MN) is the development of second MNs. Cytostatic drugs of the epipodophyllotoxin group and alkylating agents contribute to secondary acute myeloid leukemias (AML), the rare and prognostically very unfavorable second MNs. The present article provides a review of literature on risks of secondary hematological MNs associated with the therapy of first tumors. It also contains a case report of successful treatment of AML which occurred after Burkitt’s lymphoma therapy.

Keywords: second malignant neoplasms, acute myeloid leukemias, Burkitt’s lymphoma, children.

Received: December 10, 2020

Accepted: March 1, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Altekruse S, Kosary C, Krapcho M, et al. SEER Cancer Statistics Review, 1975–2007. Bethesda: National Cancer Institute; 2007.
  2. Meadows A, Friedman D, Neglia J, et al. Second neoplasms in survivors of childhood cancer: findings from the Childhood Cancer Survivor Study cohort. J Clin Oncol. 2009;27(14):2356–62. doi: 10.1200/JCO.2008.21.1920.
  3. Friedman DL, Whitton J, Leisenring W, et al. Subsequent neoplasms in 5-year survivors of childhood cancer: the Childhood Cancer Survivor Study. J Natl Cancer Inst. 2010;102(14):1083–95. doi: 10.1093/jnci/djq238.
  4. O’Brien MM, Donaldson SS, Balise RR, et al. Second malignant neoplasms in survivors of pediatric Hodgkin’s lymphoma treated with low-dose radiation and chemotherapy. J Clin Oncol. 2010;28(7):1232–9. doi: 10.1200/JCO.2009.24.8062.
  5. Dorffel W, Riepenhausenl M, Luders H, et al. Secondary Malignancies Following Treatment for Hodgkin’s Lymphoma in Childhood and Adolescence. Dtsch Arztebl Int. 2015;112(18):320–7. doi: 10.3238/arztebl.2015.0320.
  6. Bhatia S, Yasui Y, Robison L, et al. High risk of subsequent neoplasms continues with extended follow-up of childhood Hodgkin’s disease: report from the Late Effects Study Group. J Clin Oncol. 2003;21(23):4386–94. doi: 10.1200/JCO.2003.11.059.
  7. Pui C, Behm F, Raimondi S, et al. Secondary acute myeloid leukemia in children treated for acute lymphoid leukemia. N Engl J Med. 1989;321(3):136–42. doi: 10.1056/NEJM198907203210302.
  8. Pui C. Therapy-related myeloid leukaemia. Lancet. 1990;336(8723):1130–1. doi: 10.1016/0140-6736(90)92607-j.
  9. Ratain M, Rowley J. Therapy-related acute myeloid leukemia secondary to inhibitors of topoisomerase II: from the bedside to the target genes. Ann Oncol. 1992;3(2):107–11. doi: 10.1093/oxfordjournals.annonc.a058121.
  10. Tallman MS, Gray R, Bennett JM, et al. Leukemogenic potential of adjuvant chemotherapy for early-stage breast cancer: the Eastern Cooperative Oncology Group experience. J Clin Oncol. 1995;13(7):1557–63. doi: 10.1200/JCO.1995.13.7.1557.
  11. Hijiya N, Ness K, Ribeiro R, Hudson M. Acute leukemia as a secondary malignancy in children and adolescents: current findings and issues. Cancer. 2009;115(1):23–35. doi: 10.1002/cncr.23988.
  12. Swerdlow SH, Campo E, Harris NL, et al. WHO Classification of tumours of hematopoietic and lymphoid tissues. Revised 4th edition. Lion: IARC Press; 2017. p. 581.
  13. Verdeguer A, Ruiz JG, Ferris J, et al. Acute non-lymphoblastic leukemia in children treated for acute lymphoblastic leukemia with an intensive regimen including teniposide. Med Pediatr Oncol. 1992;20(1):48–52. doi: 10.1002/mpo.2950200110.
  14. Pedersen-Bjergaard J, Sigsgaard TC, Nielsen D, et al. Acute monocytic or myelomonocytic leukemia with balanced chromosome translocations to band 11q23 after therapy with 4-epi-doxorubicin and cisplatinum or cyclophosphamide for breast cancer. J Clin Oncol. 1992;10(9):1444–51. doi: 10.1200/JCO.1992.10.9.1444.
  15. Donatini B, Krupp P. Secondary pre-leukemia and etoposide. Lancet. 1991;338(8777):1269. doi: 10.1016/0140-6736(91)92133-M.
  16. Pui CH, Ribeiro RC, Hancock ML, et al. Acute myeloid leukemia in children treated with epipodophyllotoxins for acute lymphoblastic leukemia. N Engl J Med. 1991;325(24):1682–7. doi: 10.1056/NEJM199112123252402.
  17. Ballen KK, Antin JH. Treatment of therapy related acute myelogenous leukemia and myelodysplastic syndromes. Hematol Oncol Clin N Am. 1993;7(2):477–93. doi: 10.1016/s0889-8588(18)30253-3.
  18. Aguilera DG, Vaklavas C, Tsimberidou AM, et al. Pediatric Therapy-related Myelodysplastic Syndrome/Acute Myeloid Leukemia: The MD Anderson Cancer Center Experience. J Pediatr Hematol Oncol. 2009;31(11):803–11. doi: 10.1097/MPH.0b013e3181ba43dc.
  19. De Witte T, Hermans J, van Biezen J, et al. Prognostic variables in bone marrow transplantation for secondary leukemia and myelodysplastic syndrome: a survey of the working party on leukemia. Bone Marrow Transplant. 1991;7(2):40.
  20. Larson RA. Etiology and management of therapy-related myeloid leukemia. Hematology Am Soc Hematol Educ Program. 2007;2007(1):453–9. doi: 10.1182/asheducation-2007.1.453.
  21. Xinan (Holly) Yang, Bin Wang, John M. Cunningham Identification of epigenetic modifications that contribute to pathogenesis in therapy-related AML: Effective integration of genome-wide histone modification with transcriptional profiles. BMC Med Genom. 2015;8(2):S6. doi: 10.1186/1755-8794-8-S2-S6.
  22. Ковригина А.М. Пересмотренная классификация ВОЗ опухолей гемопоэтической и лимфоидной ткани, 2017 г. (4-е издание): миелоидные неоплазии. Архив патологии. 2018;80(6):43–9. doi: 10.17116/patol20188006143.
    [Kovrigina AM. A revised 4 edition WHO Classification of Tumors of Hematopoietic and Lymphoid Tissues, 2017: myeloid neoplasms. Arkhiv patologii. 2018;80(6):43–9. doi: 10.17116/patol20188006143. (In Russ)]
  23. Itzykson R, Kosmider O, Fenaux P. Somatic mutations and epigenetic abnormalities in myelodysplastic syndromes. Best Pract Res Clin Haematol. 2013;26(4):355–64. doi: 10.1016/j.beha.2014.01.001.
  24. Семенова Н.Ю., Бессмельцев С.С., Ругаль В.И. Биология ниши гемопоэтических стволовых клеток. Клиническая онкогематология. 2014;7(4):501–11.
    [Semenova NYu, Bessmeltsev SS, Rugal VI. Biology of Hematopoietic Stem Cell Niche. Klinicheskaya onkogematologiya. 2014;7(4):501–11. (In Russ)]
  25. Rihani R, Bazzeh F, Faqih N, Sultan I. Secondary hematopoietic malignancies in survivors of childhood cancer: an analysis of 111 cases from the Surveillance, Epidemiology, and End Result-9 registry. Cancer. 2010;116(18):4385–94. doi: 10.1002/cncr.25313.
  26. Nottage K, Lanktot J, Li Zh, et al. Long-term risk for subsequent leukemia after treatment for childhood cancer: a report from the Childhood Cancer Survivor Study. 2011;117(23):6315–8. doi: 10.1182/blood-2011-02-335158.
  27. Валиев Т.Т. Лимфома Беркитта у детей: 30 лет терапии. Педиатрия. Журнал им. Г.Н. Сперанского. 2020;99(4):35–41.
    [Valiev TT. Burkitt’s lymphoma in children: 30 years of therapy. Zhurnal im. G.N. Speranskogo. 2020;99(4):35–41. (In Russ)]
  28. Павлова Т.Ю., Валиев Т.Т. Вторые злокачественные опухоли у лиц, перенесших онкологическое заболевание в детстве. Педиатрия. Consilium Medicum. 2020;2:12–6. doi: 10.26442/26586630.2020.2.200234.
    [Pavlova TYu, Valiev TT. Second malignant tumors in pediatric cancer survivors. Consilium Medicum. 2020;2:12–6. doi: 10.26442/26586630.2020.2.200234. (In Russ)]
  29. Lee S-S. Therapy-related Acute Myeloid Leukemia Following Treatment for Burkitt’s Lymphoma. Chonnam Med J. 2017;53(3):229–30. doi: 10.4068/cmj.2017.53.3.229.
  30. Ripperger T, Bielack SS, Borkhardt B, et al. Childhood cancer predisposition syndromes—A concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology. Am J Med Genet Part A. 2017;173(4):1017–37. doi: 10.1002/ajmg.a.38142.

Mastocytosis in Adults: A Retrospective Analysis of the Clinical Course and Treatment of 58 Patients

VG Potapenko1,2, VV Baikov2, IE Belousova3, EA Belyakova4, MV Barabanshchikova2, DV Zaslavsky5, IS Zyuzgin6, AV Klimovich1, YuA Krivolapov4, TG Kulibaba7, EV Lisukova2, EE Leenman4, LA Mazurok8, AM Maksimova3, EV Morozova2, AS Nizamutdinova9, KA Skoryukova1, EA Ukrainchenko9, NV Medvedeva1

1 Municipal Clinical Hospital No. 31, 3 Dinamo pr-t, Saint Petersburg, Russian Federation, 197110

2 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

3 SM Kirov Military Medical Academy, 6 Akademika Lebedeva str., Saint Petersburg, Russian Federation, 194044

4 II Mechnikov North-Western State Medical University, 41 Kirochnaya str., Saint Petersburg, Russian Federation, 191015

5 Saint-Petersburg State Pediatric Medical University, 2 Litovskaya str., Saint Petersburg, Russian Federation, 194100

6 NN Petrov National Medical Cancer Research Center, 68 Leningradskaya str., Pesochnyi settlement, Saint Petersburg, Russian Federation, 197758

7 Saint Petersburg State University, 7/9 Universitetskaya emb., Saint Petersburg, Russian Federation, 199034

8 Kurgan Regional Clinical Hospital, 63 Tomina str., Kurgan, Russian Federation, 640002

9 Aleksandrov Hospital, 4 bld. 3 pr-t Solidarnosti, Saint Petersburg, Russian Federation, 193312

For correspondence: Vsevolod Gennadevich Potapenko, MD, PhD, 3 Dinamo pr-t, Saint Petersburg, Russian Federation, 197110; Tel.: +7(905)284-51-38; e-mail: potapenko.vsevolod@mail.ru

For citation: Potapenko VG, Baikov VV, Belousova IE, et al. Mastocytosis in Adults: A Retrospective Analysis of the Clinical Course and Treatment of 58 Patients. Clinical oncohematology. 2021;14(2):158–66. (In Russ).

DOI: 10.21320/2500-2139-2021-14-2-158-166


ABSTRACT

Background. Mastocytosis is a disease caused by proliferation and accumulation of clonal mast cells in one or more organs. It is often associated with other hematological tumors. Aggressive forms of mastocytosis (AFM) require specific therapy. In non-aggressive forms of mastocytosis (NFM) symptomatic treatment is needed. NFMs prevail, therefore, the disease often goes unrecognized.

Aim. To analyze the clinical course and treatment outcomes in different forms of adult mastocytosis.

Materials & Methods. The retrospective analysis was based on the records of patients who received in-person and distance consultation within the period from 11/2008 to 11/2020. The analysis of complaints in disease onset and over time was carried out using questionnaires. NFM patients received symptomatic treatment with antihistamines. To all AFM patients chemotherapy was administered.

Results. The analysis includes the data of 58 patients: 39 (67.2 %) women and 18 (32.8 %) men. The median age was 40 years (range 18–79 years), the median age on diagnosis was 39 years (range 1–79 years). In all patients skin rashes were reported. The median age of the first skin manifestations was 25 years (range 0.1–70 years). In-person monitoring was conducted in 34 (58.6 %) patients, 24 (41.4 %) patients received distance consultations. Median follow-up was 56.5 months (range 3–564 months). In 8 (13.7 %) patients mastocytosis was diagnosed in childhood with the median of 9 years (range 0–15 years). The diagnosis was morphologically confirmed in 46 (79.3 %) patients. Main complaints included pruritus (67.2 %), edema and erythema response to various irritants (62 %). In 45 (77.5 %) patients NFMs were reported. The regular symptomatic treatment of 78.8 % of NFM patients consisted only of antihistamines (57.9 %), and 2 (4.4 %) patients noted poor disease symptom control. One (2.2 %) patient died of associated chronic myelomonocytic leukemia. None of NFM patients required cytoreductive treatment. AFMs were diagnosed in 13 (22.4 %) patients, 5 (38.4 %) out of them had mast cell leukemia. The indications for starting chemotherapy were cytopenia (n = 3; 23 %), extensive osteolysis (n = 7; 53.8 %), ascitic syndrome with portal hypertension (n = 6; 46,1 %). Overall survival of AFM patients was 84.6 % (n = 11) with median follow-up of 80 months (range 12–131 months).

Conclusion. NFM prognosis is favorable. Antihistamines are effective in relieving complaints of most patients. Cytostatic treatment of AFM in some patients provides long-lasting antitumor response.

Keywords: mastocytosis, tryptase, mast cells, indolent mastocytosis, aggressive mastocytosis, С-KIT, cladribine, imatinib.

Received: December 13, 2020

Accepted: March 3, 2021

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Branford WA. Edward Nettleship (1845–1913) and the description of urticaria pigmentosa. Int J Dermatol. 1994;33(3):214–6. doi: 10.1111/j.1365-4362.1994.tb04957.x.
  2. Unna PG. Beitrage zur Anatomie und Pathogenese der Urticaria simplex und pigmentosa. Monatschr Prakt Dermatol. 1887;3:9.
  3. Brockow K. Epidemiology, prognosis, and risk factors in mastocytosis. Immunol Allergy Clin N Am. 2014;34(2):283–95. doi: 10.1016/j.iac.2014.01.003.
  4. Потапенко В.Г., Скорюкова К.А., Лисукова Е.В. и др. Мастоцитоз у детей. Клиническая и лабораторная характеристика группы 111 пациентов. Педиатрия. 2018;97(4):135–40.
    [Potapenko VG, Skoryukova KA, Lisukova EV, et al. Mastocytosis in children. Clinical and laboratory characteristics of a group of 111 patients. Pediatriya. 2018;97(4):135–40. (In Russ)]
  5. Kristensen T, Vestergaard H, Bindslev-Jensen C, et al. Sensitive KIT D816V mutation analysis of blood as a diagnostic test in mastocytosis. Am J Hematol. 2014;89(5):493–8. doi: 10.1002/ajh.23672.
  6. Akin C, Valent P, Metcalfe DD. Mast cell activation syndrome: proposed diagnostic criteria. J Allergy Clin Immunol. 2010;126(6):1099–104.e4. doi: 10.1016/j.jaci.2010.08.035.
  7. Carter MC, Metcalfe DD. Paediatric mastocytosis. Arch Dis Child. 2002;86(5):315–9. doi: 10.1136/adc.86.5.315.
  8. Van Der Veer E, Van Der Goot W, De Monchy JGR, et al. High prevalence of fractures and osteoporosis in patients with indolent systemic mastocytosis. Allergy Eur J Allergy Clin Immunol. 2012;67(3):431–8. doi: 10.1111/j.1398-9995.2011.02780.x.
  9. Vaughan ST, Jones GN. Systemic mastocytosis presenting as profound cardiovascular collapse during anaesthesia. Anaesthesia. 1998;53(8):804–7. doi: 10.1046/j.1365-2044.1998.00536.x.
  10. Galen BT, Rose MG. Darier’s sign in mastocytosis. Blood. 2014;123(8):1127. doi: 10.1182/blood-2013-11-538355.
  11. Scherber RM, Borate U. How we diagnose and treat systemic mastocytosis in adults. Br J Haematol. 2018;180(1):11–23. doi: 10.1111/bjh.14967.
  12. Heide R, Beishuizen A, De Groot H, et al. Mastocytosis in children: a protocol for management. Pediatr Dermatol. 2008;25(4):493–500. doi: 10.1111/j.1525-1470.2008.00738.x.
  13. Doyle LA, Sepehr GJ, Hamilton MJ, et al. A clinicopathologic study of 24 cases of systemic mastocytosis involving the gastrointestinal tract and assessment of mucosal mast cell density in irritable bowel syndrome and asymptomatic patients. Am J Surg Pathol. 2014;38(6):832–43. doi: 10.1097/PAS.0000000000000190.
  14. Arock M, Valent P. Pathogenesis, classification and treatment of mastocytosis: state of the art in 2010 and future perspectives. Expert Rev Hematol. 2010;3(4):497–516. doi: 10.1586/ehm.10.42.
  15. Valent P, Akin C, Escribano L, et al. Standards and standardization in mastocytosis: consensus statements on diagnostics, treatment recommendations and response criteria. Eur J Clin Invest. 2007;37(6):435–53. doi: 10.1111/j.1365-2362.2007.01807.x.
  16. Sperr WR, Kundi M, Alvarez-Twose I, et al. International prognostic scoring system for mastocytosis (IPSM): a retrospective cohort study. Lancet Haematol. 2019;6(12):e638–e649. doi: 10.1016/S2352-3026(19)30166-8.
  17. Lim KH, Tefferi A, Lasho TL, et al. Systemic mastocytosis in 342 consecutive adults: survival studies and prognostic factors. Blood. 2009;113(23):5727–36. doi: 10.1182/blood-2009-02-205237.
  18. Hartmann K, Escribano L, Grattan C, et al. Cutaneous manifestations in patients with mastocytosis: Consensus report of the European Competence Network on Mastocytosis; the American Academy of Allergy, Asthma &Immunology; and the European Academy of Allergology and Clinical Immunology. J Allergy Clin Immunol. 2016;137(1):35–45. doi: 10.1016/j.jaci.2015.08.034.
  19. Laroche M, Livideanu C, Paul C, et al. Interferon alpha and pamidronate in osteoporosis with fracture secondary to mastocytosis. Am J Med. 2011;124(8):776–8. doi: 10.1016/j.amjmed.2011.02.038.
  20. Rossini M, Zanotti R, Viapiana O, et al. Zoledronic acid in osteoporosis secondary to mastocytosis. Am J Med. 2014;127(11):1127.e1–1127.е4. doi: 10.1016/j.amjmed.2014.06.015.
  21. Wang SA, Hutchinson L, Tang G, et al. Systemic mastocytosis with associated clonal hematological non-mast cell lineage disease: clinical significance and comparison of chomosomal abnormalities in SM and AHNMD components. Am J Hematol. 2013;88(3):219–24. doi: 10.1002/ajh.23380.
  22. Barete S, Lortholary O, Damaj G, et al. Long-term efficacy and safety of cladribine (2-CdA) in adult patients with mastocytosis. Blood. 2015;126(8):1009–16. doi: 10.1182/blood-2014-12-614743.
  23. Gotlib J, Kluin-Nelemans HC, George TI, et al. Efficacy and Safety of Midostaurin in Advanced Systemic Mastocytosis. N Engl J Med. 2016;374(26):2530–41. doi: 10.1056/NEJMoa1513098.
  24. DeAngelo DJ, Quiery AT, Radia D, et al. Clinical activity in a phase 1 study of Blu-285, a potent, highly-selective inhibitor of KIT D816V in advanced systemic mastocytosis (AdvSM). Blood. 2017;130(Suppl 1):2. doi: 10.1182/blood.V130.Suppl_1.2.2.
  25. Vega-Ruiz A, Cortes JE, Sever M, et al. Phase II study of imatinib mesylate as therapy for patients with systemic mastocytosis. Leuk Res. 2009;33(11):1481–4. doi: 10.1016/j.leukres.2008.12.020.
  26. Longley B, Metcalfe DD, Tharp M, et al. Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis. Proc Natl Acad Sci USA. 1999;96(4):1609–14. doi: 1073/pnas.96.4.1609.
  27. Horny HP, Akin C, Arber DA, et al. In: Swerdlow S, Campo E, Harris N, et al. (eds.). WHO classification of tumours of haematopoietic and lymphoid tissues. Revised 4th edition. Lyon: IARC Press; 2017.
  28. Gotlib J, Pardanani A, Akin C, et al. International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) & European Competence Network on Mastocytosis (ECNM) consensus response criteria in advanced systemic mastocytosis. Blood. 2013;121(13):2393–401. doi: 10.1182/blood-2012-09-458521.
  29. Elmaagacli AH, Jehn C, Shikova Y, et al. Advanced systemic mastocytosis with strong expression of signaling lymphocyte activation marker family member 7 (SLAMF7) responsive to therapy with elotuzumab and lenalidomide. Leuk Lymphoma. 2020;61(2):485–7. doi: 10.1080/10428194.2019.1668939.
  30. Meni C, Bruneau J, Georgin-Lavialle S, et al. Paediatric mastocytosis: a systematic review of 1747 cases. Br J Dermatol. 2015;172(3):642–51. doi: 10.1111/bjd.13567.
  31. Middelkamp Hup MA, Heide R, Tank B, et al. Comparison of mastocytosis with onset in children and adults. J Eur Acad Dermatol Venereol. 2002;16(2):115–20. doi: 10.1046/j.1468-3083.2002.00370.x.
  32. Brockow K, Scott LM, Worobec AS, et al. Regression of urticaria pigmentosa in adult patients with systemic mastocytosis: correlation with clinical patterns of disease. Arch Dermatol. 2002;138(6):785–90. doi: 10.1001/archderm.138.6.785.
  33. Wolff K, Komar M, Petzelbauer P. Clinical and histopathological aspects of cutaneous mastocytosis. Leuk Res. 2001;25(7):519–28. doi: 10.1016/s0145-2126(01)00044-3.
  34. Valent P, Oude Elberink JNG, Gorska A, et al. The Data Registry of the European Competence Network on Mastocytosis (ECNM): Set Up, Projects, and Perspectives. J Allergy Clin Immunol Pract. 2019;7(1):81–7. doi: 10.1016/j.jaip.2018.09.024.
  35. Merante S, Ferretti VV, Elena C, et al. The Italian Mastocytosis Registry: 6-year experience from a hospital-based registry. Fut Oncol. 2018;14(26):2713–23. doi: 10.2217/fon-2018-0291.
  36. Gotlib J, Gerds AT, Bose P, et al. Systemic Mastocytosis, Version 2.2019. NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018;16(12):1500–37. doi: 10.6004/jnccn.2018.0088.
  37. Matito A, Morgado JM, Sanchez-Lopez P, et al. Management of Anesthesia in Adult and Pediatric Mastocytosis: A Study of the Spanish Network on Mastocytosis (REMA) Based on 726 Anesthetic Procedures. Int Arch Allergy Immunol. 2015;167(1):47–56. doi: 10.1159/000436969.
  38. Pardanani A, Elliott M, Reeder T, et al. Imatinib for systemic mast cell disease. Lancet. 2003;362(9383):535–6. doi: 10.1016/s0140-6736(03)14115-3.
  39. Droogendijk HJ, Kluin-Nelemans HJ, van Doormaal JJ, et al. Imatinib mesylate in the treatment of systemic mastocytosis: a phase II trial. Cancer. 2006;107(2):345–51. doi: 10.1182/blood.v104.11.1516.1516.
  40. Kluin-Nelemans HC, Oldhoff JM, Van Doormaal JJ, et al. Cladribine therapy for systemic mastocytosis. Blood. 2003;102(13):4270–6. doi: 10.1182/blood-2003-05-1699.

In Memory of Prof. L.A. Makhonova

29 января 2021 г. ушла из жизни основоположник отечественной детской онкогематологии, заслуженный врач РФ, доктор медицинских наук, профессор Лидия Алексеевна Махонова.

Лидия Алексеевна была крупнейшим российским ученым, внесшим значительный вклад в становление и развитие клинической детской онкогематологии.

После окончания в 1949 г. 2-го Московского медицинского института им. Н.И. Пирогова (в настоящее время — Российский национальный исследовательский медицинский университет им. Н.И. Пирогова) работала врачом-педиатром, затем руководила терапевтическим отделением Московской детской больницы № 1. Многие годы клиническая работа Л.А. Махоновой была связана с диагностикой и лечением злокачественных опухолей кроветворной и лимфоидной тканей у детей.

Накопленный опыт и появление первых противоопухолевых препаратов, вызывающих противолейкемический эффект, стали основой кандидатской диссертации на тему: «Современные методы лечения острого лейкоза у детей», которую Лидия Алексеевна защитила в 1963 г. под руководством академика АМН СССР, основателя отечественной гематологии И.А. Кассирского. Важнейшим выводом работы стало положение о том, что при острых лейкозах у детей возможно получение ремиссии с помощью 6-меркаптопурина и преднизолона. Была подтверждена необходимость проведения поддерживающей терапии при острых лейкозах, а также обозначены оптимальные дозы и режимы назначения первых эффективных противоопухолевых препаратов.

Под эгидой кафедры факультетской педиатрии 2-го Московского медицинского института им. Н.И. Пирогова в 1964 г. Лидия Алексеевна организовала первое в нашей стране детское гематологическое отделение на базе Морозовской детской больницы в г. Москве. В новом отделении получали лечение дети с лейкозами, лимфомами, геморрагическим диатезом, анемией. Л.А. Махонова, будучи ярким и талантливым клиницистом, продолжала и развивала традиции отечественной клинико-морфологической гематологической школы. Ее клиническая деятельность не ограничивалась только оценкой особенностей лечения опухолей системы крови у детей. Она принимали активное личное участие в постановке диагноза на основании анализа цитологических препаратов костного мозга и лимфатических узлов. При непосредственном участии Лидии Алексеевны в 1960-е годы по сути были заложены основы новой дисциплины «Детская гематология» и начата профессиональная последипломная подготовка детских гематологов.

В 1960–1970-е годы Л.А. Махонова была главным детским гематологом МЗ РСФСР. Под ее непосредственным руководством заложены основы всей онкогематологической педиатрической службы в РСФСР.

Пристальный интерес у Л.А. Махоновой вызывали работы по поиску новых лекарственных препаратов, эффективных при острых лейкозах. Л.А. Махонова и ее коллеги (С.А. Маякова, И.Е. Гаврилова) в 1970-е годы активно исследовали иммунологические методы терапии. Оценивались противолейкемический эффект аллоиммунизации, роль и место препаратов интерферона в лечении острых лейкозов у детей. В 1973 г. Л.А. Махонова защитила докторскую диссертацию на тему: «Материалы к клинике и лечению (химиотерапия и иммунотерапия) острого лейкоза у детей».

Детская онкогематология еще только формировалась, стандарты лечения острых лейкозов у детей только начинали разрабатываться, а в названии докторской диссертации Л.А. Махоновой в 1973 г. уже звучало новое и перспективное направление противоопухолевого лечения — иммунотерапия. Такой подход с современных позиций можно признать как великий дар научного и клинического предвидения. Ведь сегодня онкогематологию, равно как и онкологию в целом, невозможно представить без достижений иммунотерапии.

А тогда… Как вспоминают соратники Лидии Алексеевны по борьбе с лейкозами, она вынесла тяжелые сражения с коллегами клиники для взрослых, отстаивая перспективность применения интерферонов при злокачественных опухолях системы крови. Смелый и беззаветно преданный лечению детей человек. Напрочь лишенный карьеризма, готовый в любую минуту броситься в бой за свои идеи, пусть даже еще только зарождающиеся, как было с иммунотерапией. Верность этому направлению она пронесла через всю жизнь. И в том, что мы сегодня видим, — настоящий триумф иммунологических подходов в лечении гематологических опухолей, огромная неоценимая заслуга Лидии Алексеевны Махоновой.

В 1976 г. Николай Николаевич Блохин пригласил Лидию Алексеевну возглавить детскую онкогематологическую службу во Всесоюзном онкологическом научном центре АМН СССР (ВОНЦ). Она руководила отделением детской онкогематологии в течение 17 лет, постоянно совершенствуясь в профессии и передавая богатейшие опыт и знания своим ученикам.

В 1980–1990-е годы Л.А. Махонова продолжает изучать и активно внедрять в клиническую практику новые лекарственные препараты и протоколы лечения лейкозов и лимфом у детей. Отделение детской химиотерапии гемобластозов ВОНЦ при непосредственном участии Лидии Алексеевны как руководителя одно из первых в России освоило методики терапии метотрексатом в высоких дозах, разработало принципы сопроводительной терапии и эффективные методы коррекции осложнений противоопухолевого лечения.

Л.А. Махонова, ее коллеги и ученики внесли основополагающий вклад в становление и развитие международного научного сотрудничества благодаря включению отделения химиотерапии гемобластозов НИИ детской онкологии и гематологии Российского онкологического научного центра им. Н.Н. Блохина РАМН (РОНЦ) в международную группу BFM (Berlin-Frankfurt-Munster) по лечению острого лимфобластного лейкоза у детей. До настоящего времени мы используем богатейший клинический и научный опыт Л.А. Махоновой. В отделении используются новые версии наиболее эффективных протоколов лечения острых лейкозов и лимфом у детей: ALL-IC BFM 2002, 2009, 2020; B-NHL-BFM 1995, 2004.

Всю свою профессиональную деятельность Л.А. Махонова особое внимание уделяла проблемам диагностики и лечения гистиоцитарных опухолей у детей и взрослых. Продолжая работу в РОНЦ им. Н.Н. Блохина до 2017 г., Лидия Алексеевна оказывала большую консультативную помощь в двух клинических институтах Центра: НИИ детской онкологии и гематологии и НИИ клинической онкологии им. Н.Н. Трапезникова. Благодаря Лидии Алексеевне в НИИ детской онкологии и гематологии ФГБУ «НМИЦ онкологии им. Н.Н. Блохина» Минздрава России накоплен крупнейший в стране и мире клинический материал, отражающий лечение гистиоцитозов более чем у 1500 детей.

Л.А. Махонова — автор более 300 научных работ, в т. ч. монографий, методических руководств и рекомендаций. Руководитель 32 диссертаций на соискание ученой степени кандидата медицинских наук и 8 — доктора медицинских наук.

Лидия Алексеевна награждена медалью «За заслуги перед Отечеством» (2002), премией Правительства РФ за разработку и применение иммунокорректирующих препаратов (1997), премией РОНЦ им. Н.Н. Блохина за работу «Современная стратегия и результаты лечения лимфоидных опухолей у детей» (2005).

Л.А. Махонова была не только выдающимся врачом, исследователем, педагогом, но и мудрым наставником. Жизнь Л.А. Махоновой — пример беззаветного служения великому делу лечения тяжело больных детей с онкогематологическими заболеваниями.

 

Коллеги по работе и коллектив редакции


Read in PDF

CAR-T Technology and New Opportunities for Tumor Treatment

VYu Pavlova1, ES Livadnyi2

1 SV Belyaev Kemerovo Regional Clinical Hospital, 22 bld. 2 Oktyabrskii pr-t, Kemerovo, Russian Federation, 650066

2 Kemerovo State Medical University, 22a Voroshilova str., Kemerovo, Russian Federation, 650066

For correspondence: Vera Yurevna Pavlova, MD, PhD, 22 bld. 2 Oktyabrskii pr-t, Kemerovo, Russian Federation, 650066; Tel.: +7(951)570-57-86; e-mail: vera.4447.kem@mail.ru

For citation: Pavlova VYu, Livadnyi ES. CAR-T Technology and New Opportunities for Tumor Treatment. Clinical oncohematology. 2021;14(1):149–56. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-149-156


ABSTRACT

As a cause of death malignant neoplasms come in at the second place after cardiovascular disorders. CAR-T (chimeric antigen receptor of T-cells) therapy is an advanced malignant tumor treatment method. The use of CAR-T lymphocytes refers to adoptive immunotherapy. CAR-T technology is based on “extracting” immune cells (T-lymphocytes) and their genetic modification aimed at acquiring antitumor properties and followed by reinfusion. The advantage of CAR-T therapy in comparison to other treatment methods is that for target cell recognition T-lymphocytes are not dependent on major histocompatibility complex class 1 (MHC-I) molecules. The literature data we collected and analyzed show that this is a fundamentally new and effective treatment method of oncohematological diseases including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin’s lymphomas. Clinical trials proved the advantage of CAR-T therapy in comparison to other treatment methods applied in this field. The analysis of literature showed that CAR-T therapy can be reasonably regarded as one of the advanced opportunities for malignant tumor treatment.

Keywords: adoptive immunotherapy, CAR-T lymphocytes, chimeric antigen receptor.

Received: September 20, 2020

Accepted: December 1, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–Е386. doi: 10.1002/ijc.29210.
  2. Stewart BW, Wilde CP (eds). World cancer report 2014. Lyon: IARC Press, 2014. 619 p.
  3. Plummer M, de Martel C, Vignat J, et al. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob 2016;4(9):e609–e616. doi: 10.1016/S2214-109X(16)30143-7.
  4. Состояние онкологической помощи населению России в 2018 году. Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Петровой. М.: МНИОИ им. П.А. Герцена — филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2019. 236 с.
    [Caprin AD, Starinskii VV, Petrova GV, eds. Sostoyanie onkologicheskoi pomoshchi naseleniyu Rossii v 2018 godu. (The state of cancer care in Russia in 2018.) Moscow: MNIOI im. P.A. Gertsena — a branch of FGBU “NMITs radiologii” Minzdrava Rossii Publ.; 2019. 236 p. (In Russ)]
  5. Здравоохранение в России. Статистический сборник. М.: Росстат, 2011. 326 с.
    [Zdravookhranenie v Rossii. Statisticheskii sbornik. (Health care in Russia. Statistics digest.) Мoscow: Rosstat Publ.; 2011. 326 p. (In Russ)]
  6. Halaleh K, Gale RP. Cancer care in the Palestinian territoried. Lancet Oncol. 2018;19(7):e359–е364. doi: 10.1016/S1470-2045(18)30323-1.
  7. Чикилева И.О., Шубина И.Ж., Киселевский М.В. Влияние регуляторных Т-клеток на функциональную активность натуральных киллеров при иммунотерапии злокачественных опухолей. Вестник РАМН. 2012;67(4):60–4.
    [Chikileva IO, Shubina IZh, Kiselevskii MV. Influence of regulatory T-cells on the functioning of natural killer cells during cancer immunotherapy. Vestnik RAMN. 2012;67(4):60–4. (In Russ)]
  8. Титов К.С., Демидов Л.В., Шубина И.Ж. и др. Технологии клеточной иммунотерапии в лечении больных со злокачественными новообразованиями. Вестник Российского государственного медицинского университета. 2014;1:42–7.
    [Titov KS, Demidov LV, Shubina IZh, et al. Technologies of cell immunotherapy in treatment of cancer patients. Vestnik Rossiiskogo gosudarstvennogo meditsinskogo universiteta; 2014;1:42–7. (In Russ)]
  9. Wahlang B, Falkner KC, Cave MC, et al. Role of Cytochrome P450 Monooxygenase in Carcinogen and Chemotherapeutic Drug Metabolism. Adv Pharmacol. 2015;74:1–33. doi: 10.1016/bs.apha.2015.04.004.
  10. Janeway CA Jr, Travers P, Walport M, et al. Immunobiology: The Immune System in Health and Disease. 5th edition. New York: Garland Science, 2001. 884 p.
  11. Croce CM. Oncogenes and cancer. N Engl J Med. 2008;358(5):502–11. doi: 10.1056/NEJMra072367.
  12. Vicente-Duenas C, Romero-Camarero I, Cobaleda C, et al. Function of oncogenes in cancer development: a changing paradigm. EMBO J. 2013;32(11):1502–13. doi: 10.1038/emboj.2013.97.
  13. Den Haan JM, Arens R, Van Zelm MC. The activation of the adaptive immune system: cross-talk between antigen-presenting cells, T cells and B cells. Immunol Lett. 2014;162(2 Pt B):103–12. doi: 10.1016/j.imlet.2014.10.011.
  14. Emtage PC, Lo AS, Gomes EM, et al. Second-generation anti-carcinoembryonic antigen designer T cells resist activation-induced cell death, proliferate on tumor contact, secrete cytokines, and exhibit superior antitumor activity in vivo: a preclinical evaluation. Clin Cancer Res. 2008;14(24):8112–22. doi: 10.1158/1078-0432.
  15. Maher Immunotherapy of Malignant Disease Using Chimeric Antigen Receptor Engrafted T Cells. ISRN Oncol. 2012;2012:278093. doi: 10.5402/2012/278093.
  16. BonifantL, Jackson HJ, Brentjens RJ, et al. Toxicity and management in CAR T-cell therapy. Mol Ther. 2016;3:16011. doi: 10.1038/mto.2016.11.
  17. Xu J, Wang Q, Xu H, et al. Anti-BCMA CAR-T cells for treatment of plasma cell dyscrasia: case report on POEMS syndrome and multiple myeloma. J Hematol Oncol. 2018;11(1):128. doi: 10.1186/s13045-018-0672-7.
  18. Wang J, Chen S, Xiao W, et al. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol. 2018;11(1):7. doi: 10.1186/s13045-017-0553-5.
  19. Wei J, Han X, Bo J, et al. Target selection for CAR-T therapy. J Hematol Oncol. 2019;12(1):62. doi: 10.1186/s13045-019-0758-x.
  20. Si W, Li C, Wei P. Synthetic immunology: T-cell engineering and adoptive immunotherapy. Synth Syst Biotechnol. 2018;3(3):179–85. doi: 10.1016/j.synbio.2018.08.001.
  21. Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. J Cell Immunother. 2016;2(2):59–68. doi: 10.1016/j.jocit.2016.08.001.
  22. Chmielewski M, Abken H. Trucks: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):1145–54. doi: 10.1517/14712598.2015.1046430.
  23. Zhao Z, Chen Y, Francisco NM, et al. The application of CAR-T cell therapy in hematological malignancies: advantages and challenges. Acta Pharm Sin B. 2018;8(4):539–51. doi: 10.1016/j.apsb.2018.03.001.
  24. Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev. 2019;34:45–55. doi: 10.1016/j.blre.2018.11.002.
  25. Brudno JN, Maric I, Hartman SD, et al. T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor prognosis relapsed multiple myeloma. J Clin Oncol. 2018;36(22):2267–80. doi: 10.1200/JCO.2018.77.8084.
  26. Maude SL, Frey N, Shaw PA, et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222.
  27. Fernandez A Cure for Cancer? How CAR T-Cell Therapy is Revolutionizing Oncology. Available from: https://www.labiotech.eu/features/car-t-therapy-cancer-review/ (accessed 25.11.2020).
  28. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi: 10.1126/scitranslmed.aac5415.
  29. Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377(26):2545–54. doi: 10.1056/NEJMoa1708566.
  30. Pule MA, Savoldo B, Myers GD, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14(11):1264–70. doi: 10.1038/nm.1882.
  31. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  32. Porter DL, Levine BL, Kalos M, et al. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33. doi: 10.1056/NEJMoa1103849.
  33. Riches JC, Gribben JG. Understanding the immunodeficiency in chronic lymphocytic leukemia: potential clinical implications. Hematol Oncol Clin North Am. 2013;27(2):207–35. doi: 10.1016/j.hoc.2013.01.003.
  34. Ellard R, Stewart O. The EBMT Guidelines for practice. A framework for managing Patient Care, CRS and Neurotoxicity. 1st European CAR T cell meeting, 14–16 February 2019, Paris, France.

CAR T-Cell Therapy with NKG2D Chimeric Antigen Receptor in Relapsed/Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome

KA Levchuk1, EV Belotserkovskaya1,2, DYu Pozdnyakov1, LL Girshova1, AYu Zaritskey1, AV Petukhov1,2,3

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

2 Institute of Cytology, 4 Tikhoretskii pr-t, Saint Petersburg, Russian Federation, 194064

3 Sirius University of Science and Technology, 1 Olimpiiskii pr-t, Sochi, Russian Federation, 354340

For correspondence: Kseniya Aleksandrovna Levchuk, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; e-mail: levchuk_ka@almazovcentre.ru

For citation: Levchuk KA, Belotserkovskaya EV, Pozdnyakov DYu, et al. CAR T-Cell Therapy with NKG2D Chimeric Antigen Receptor in Relapsed/Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome. Clinical oncohematology. 2021;14(1):138–48. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-138-148


ABSTRACT

NK-cells as innate immunity elements manifest key reactions of antitumor immune response. NKG2D is an activating transmembrane receptor of NK-cells which is responsible for cytotoxicity initiation in response to the binding of specific ligands of genetically modified cells. Selective expression of NKG2D ligands provides a unique perspective on the therapy of wide variety of tumors. Acute myeloid leukemias (AML) are malignant hematological tumors with a high relapse risk. Due to the complexity of AML treatment strategy it is necessary to develop new approaches to tumor elimination using novel genetic constructs. Currently available CAR T-cell drugs with NKG2D receptor are successfully subjected to clinical studies in AML patients and prove their high therapeutic potential.

Keywords: acute myeloid leukemias, chimeric antigen receptor, adoptive therapy, NKG2D, NK-cells.

Received: August 22, 2020

Accepted: December 5, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Arber D, Orazi A, Hasserjian R. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391–405. doi: 10.1182/blood-2016-03-643544.
  2. Bullinger L, Dohner K, Dohner H. Genomics of Acute Myeloid Leukemia Diagnosis and Pathways. J Clin Oncol. 2017;35(9):934–46. doi: 10.1200/JCO.2016.71.2208.
  3. The Leukemia & Lymphoma Society Updated data on blood cancers. Facts 2018–2019. Available from: https://www.lls.org/facts-and-statistics/facts-and-statistics-overview/facts-and-statistics (accessed 30.11.2020).
  4. Dohner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. 2017;129(4):424–47. doi: 10.1182/blood-2016-08-733196.
  5. Herold T, Rothenberg-Thurley M, Grunwald VV, et al. Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia Leukemia. [published online ahead of print, 2020 Mar 30] doi: 10.1038/s41375-020-0806-0.
  6. Estey EH, Schrier SL. Prognosis of the myelodysplastic syndromes in adults. UpToDate. 2017. Available from: https://www.uptodate.com/contents/prognosis-of-the-myelodysplastic-syndromes-in-adults (accessed 28.11.2020).
  7. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. 2005;106(4):1154–63. doi: 10.1182/blood-2005-01-0178.
  8. Burnett AK, Milligan D, Goldstone A, et al. The impact of dose escalation and resistance modulation in older patients with acute myeloid leukemia and high risk myelodysplastic syndrome: the results of the LRF AML14 trial. Br J Haematol. 2009;145(3):318–32. doi: 10.1111/j.1365-2141.2009.07604.x.
  9. Lowenberg G. Strategies in the treatment of acute myeloid leukemia. Haematologica. 2004;89(9):1029–32.
  10. Burnett AK. Acute myeloid leukemia: Treatment of adults under 60 years. Rev Clin Exp Hematol. 2002;6(1):26–45. doi: 10.1046/j.1468-0734.2002.00058.x.
  11. Estey EH. Treatment of relapsed and refractory acute myelogenous leukemia. 2000;14(3):476–9. doi: 10.1038/sj.leu.2401568.
  12. Giles F, O’Brien S, Cortes J, et al. Outcome of patients with acute myelogenous leukemia after second salvage therapy. 2005;104(3):547–54. doi: 10.1002/cncr.21187.
  13. Leopold LH, Willemze R. The treatment of acute myeloid leukemia in first relapse: A comprehensive review of the literature. Leuk Lymphoma. 2002;43(9):1715–27. doi: 10.1080/1042819021000006529.
  14. Lee S, Tallman MS, Oken MM, et al. Duration of second complete remission compared with first complete remission in patients with acute myeloid leukemia. 2000;14(8):1345–8. doi: 10.1038/sj.leu.2401853.
  15. Patel SA, Gerber JM. A User’s Guide to Novel Therapies for Acute Myeloid Leukemia. Clin Lymphoma Myel Leuk. 2020;20(5):277–88. doi: 10.1016/j.clml.2020.01.011.
  16. Kucukyurt S, Eskazan AE. New drugs approved for acute myeloid leukemia in 2018. Br J Clin Pharmacol. 2018;85(12):2689–93. doi: 10.1111/bcp.14105.
  17. Spear P, Wu MR, Sentman ML, Sentman CL. NKG2D ligands as therapeutic targets. Cancer Immun. 2013;13:8.
  18. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. 2012;120(12):2454–65. doi: 10.1016/s0145-2126(13)70009-2.
  19. Blum WG. Hypomethylating agents in myelodysplastic syndromes. Clin Adv Hematol Oncol. 2011;9(2):123–8.
  20. Семочкин С.В., Толстых Т.Н., Иванова В.Л. и др. Азацитидин в лечении миелодиспластических синдромов: клиническое наблюдение и обзор литературы. Клиническая онкогематология. 2012;5(3):233–8.
    [Semochkin SV, Tolstykh TN, Ivanova VL, et al. Azacitidine in the treatment of myelodysplastic syndromes: case report and literature review. Klinicheskaya onkogematologiya. 2012;5(3):233–8. (In Russ)]
  21. Ширин А.Д., Баранова О.Ю. Гипометилирующие препараты в онкогематологии. Клиническая онкогематология. 2016;9(4):369–82. doi: 10.21320/2500-2139-2016-9-4-369-382.
    [Shirin AD, Baranova OYu. Hypomethylating Agents in Oncohematology. Clinical oncohematology. 2016;9(4):369–82. doi: 10.21320/2500-2139-2016-9-4-369-382. (In Russ)]
  22. Richard-Carpentier G, DeZern AE, Takahashi K, et al. Preliminary Results from the Phase II Study of the IDH2-Inhibitor Enasidenib in Patients with High-Risk IDH2-Mutated Myelodysplastic Syndromes (MDS). 2019;134(1):678. doi: 10.1182/blood-2019-130501.
  23. Foran JM, DiNardo CD, Watts JM, et al. Ivosidenib (AG-120) in Patients with IDH1-Mutant Relapsed/Refractory Myelodysplastic Syndrome: Updated Enrollment of a Phase 1 Dose Escalation and Expansion Study. 2019;134(1):4254. doi: 10.1182/blood-2019-123946.
  24. Garcia JS. Prospects for Venetoclax in Myelodysplastic Syndromes. Hematol Oncol Clin N Am. 2020;34(2):441–8. doi: 10.1016/j.hoc.2019.10.005.
  25. Germing U, Schroeder T, Kaivers J, et al. Novel therapies in low- and high-risk myelodysplastic syndrome. Exp Rev Hematol. 2019;12(10):893–908. doi: 10.1080/17474086.2019.1647778.
  26. Platzbecker U. Treatment of MDS. 2019;133(10):1096–107. doi: 10.1182/blood-2018-10-844696.
  27. Swoboda DM, Sallman DA. Mutation-Driven Therapy in MDS. Curr Hematol Malig Rep. 2019;14(6):550–60. doi: 10.1007/s11899-019-00554-4.
  28. Миелодиспластические синдромы. Интервью с С.В. Грицаевым. Клиническая онкогематология. 2018;11(2):125–37.
    [Myelodysplastic syndromes. Interview with SV Gritsaev. Clinical oncohematology. 2018;11(2):125–37. (In Russ)]
  29. Manley PW, Weisberg E, Sattler M, et al. Midostaurin, a Natural Product-Derived Kinase Inhibitor Recently Approved for the Treatment of Hematological Malignancies. 2018;57(5):477–8. doi: 10.1021/acs.biochem.7b01126.
  30. Liu X, Gong Y. Isocitrate dehydrogenase inhibitors in acute myeloid leukemia. Biomark Res. 2019;7(1):22. doi: 10.1186/s40364-019-0173-z.
  31. Kim ES. Enasidenib: First Global Approval. 2017;77(15):1705–11. doi: 10.1007/s40265-017-0813-2.
  32. Garcia-Aranda M, Perez-Ruiz E, Redondo M. Bcl-2 Inhibition to Overcome Resistance to Chemo- and Immunotherapy. Int J Mol Sci. 2018;19(12):3950. doi: 10.3390/ijms19123950.
  33. Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for patients with relapse after allogeneic transplantation. Leukemia and Lymphoma Society Blood Cancer Research Partnership. N Engl J Med. 2016;375(2):143–53. doi: 10.1056/NEJMoa1601202.
  34. Li F, Sutherland MK, Yu C, et al. Characterization of SGN-CD123A, A Potent CD123-Directed Antibody-Drug Conjugate for Acute Myeloid Leukemia. Mol Cancer Ther. 2018;17(2):554–64. doi: 10.1158/1535-7163.MCT-17-0742.
  35. Mawad R, Gooley TA, Rajendran JG, et al. Radiolabeled AntiCD45 Antibody with Reduced-Intensity Conditioning and Allogeneic Transplantation for Younger Patients with Advanced Acute Myeloid Leukemia or Myelodysplastic Syndrome. Biol Blood Marrow Transplant. 2014;20(9):1363–8. doi: 10.1016/j.bbmt.2014.05.014.
  36. Guy DG, Uy GL. Bispecific Antibodies for the Treatment of Acute Myeloid Leukemia. Curr Hematol Malig Rep. 2018;13(6):417– doi: 10.1007/s11899-018-0472-8.
  37. Di Stasi A, Jimenez AM, Minagawa K, et al. Review of the Results of WT1 Peptide Vaccination Strategies for Myelodysplastic Syndromes and Acute Myeloid Leukemia from Nine Different Studies. Front Immunol. 2015;6:36. doi: 10.3389/fimmu.2015.00036.
  38. Van Acker HH, Versteven M, Lichtenegger FS, et al. Dendritic Cell-Based Immunotherapy of Acute Myeloid Leukemia. J Clin Med. 2019;8(5):579. doi: 10.3390/jcm8050579.
  39. Yabe T, McSherry C, Bach FH, et al. A multigene family on human chromosome 12 encodes natural killer-cell lectins. 1993;37(6):455–60. doi: 10.1007/bf00222470.
  40. Houchins JP, Yabe T, McSherry C, Bach FH. DNA sequence analysis of NKG2, a family of related cDNA clones encoding type II integral membrane proteins on human natural killer cells. J Exp Med. 1991;173(4):1017–20. doi: 10.1084/jem.173.4.1017.
  41. Upshaw JL, Arneson LN, Schoon RA, et al. NKG2D-mediated signaling requires a DAP10-bound Grb2-Vav1 intermediate and phosphatidylinositol-3-kinase in human natural killer cells. Nat Immunol. 2006;7(5):524–32. doi: 10.1038/ni1325.
  42. Diefenbach A, Tomasello E, Lucas M, et al. Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat Immunol. 2002;3(12):1142–9. doi: 10.1038/ni858.
  43. Duan S, Guo W, Xu Z, et al. Natural killer group 2D receptor and its ligands in cancer immune escape. Mol Cancer. 2019;18(1):29. doi: 10.1186/s12943-019-0956-8.
  44. Wu J, Song Y, Bakker AB, et al. An activating immunoreceptor complex formed by NKG2D and DAP10. 1999;285(5428):730–2. doi: 10.1126/science.285.5428.730.
  45. Ogasawara K, Lanier LL. NKG2D in NK and T cell-mediated immunity. J Clin Immunol. 2005;25(6):534–40. doi: 10.1007/s10875-005-8786-4.
  46. Gilfillan S, Ho EL, Cella M, et al. NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol. 2002;3(12):1150–5. doi: 10.1038/ni857.
  47. Groh V, Rhinehart R, Randolph-Habecker J, et al. Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol. 2001;2(3):255–60. doi: 10.1038/85321.
  48. Jamieson AM, Diefenbach A, McMahon CW, et al. The role of the NKG2D immunoreceptor in immune cell activation and natural killing. 2002;17(1):19–29. doi: 10.1016/s1074-7613(02)00333-3.
  49. Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol. 2003;3(10):781–90. doi: 10.1038/nri1199.
  50. Roberts AI, Lee L, Schwarz E, et al. NKG2D receptors induced by IL-15 costimulate CD28-negative effector CTL in the tissue microenvironment. J Immunol. 2001;167(10):5527–30. doi: 10.4049/jimmunol.167.10.5527.
  51. Lanier LL. NK cell recognition. Annu Rev Immunol. 2005;23(1):225–74. doi: 10.1146/annurev.immunol.23.021704.115526.
  52. Raulet DH, Gasser S, Gowen BG, et al. Regulation of ligands for the NKG2D activating receptor. Annu Rev Immunol. 2013;31(1):413–41. doi: 10.1146/annurev-immunol-032712-095951.
  53. Stephens HA. MICA and MICB genes: can the enigma of their polymorphism be resolved?. Trends Immunol. 2001;22(7):378–85. doi: 10.1016/s1471-4906(01)01960-3.
  54. Carapito R, Bahram S. Genetics, genomics, and evolutionary biology of NKG2D ligands. Immunol Rev. 2015;267(1):88–116. doi: 10.1111/imr.12328.
  55. Bartkova J, Horejsi Z, Koed K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. 2005;434(7035):864–70. doi: 10.1038/nature03482.
  56. Gorgoulis VG, Vassiliou LV, Karakaidos P, et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. 2005;434(7035):907–13. doi: 10.1038/nature03485.
  57. Maeda T, Towatari M, Kosugi H, Saito H. Up-regulation of costimulatory/adhesion molecules by histone deacetylase inhibitors in acute myeloid leukemia cells. Blood. 2000;96(12):3847–56. doi: 1182/blood.v96.12.3847.
  58. Diermayr S, Himmelreich H, Durovic B, et al. NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. 2008;111(3):1428–36. doi: 10.1182/blood-2007-07-101311.
  59. Chang YH, Connolly J, Shimasaki N, et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73(6):1777–86. doi: 10.1158/0008-5472.CAN-12-3558.
  60. Hamerman JA, Ogasawara K, Lanier LL. Cutting edge: Toll-like receptor signaling in macrophages induces ligands for the NKG2D receptor. J Immunol. 2004;172(4):2001–5. doi: 10.4049/jimmunol.172.4.2001.
  61. Carlsten M, Bjorkstrom NK, Norell H, et al. DNAX accessory molecule-1 mediated recognition of freshly isolated ovarian carcinoma by resting natural killer cells. Cancer Res. 2007;67(3):1317–25. doi: 10.1158/0008-5472.CAN-06-2264.
  62. McGilvray RW, Eagle RA, Rolland P, et al. ULBP2 and RAET1E NKG2D ligands are independent predictors of poor prognosis in ovarian cancer patients. Int J Cancer. 2010;127(6):1412–20. doi: 10.1002/ijc.25156.
  63. Cathro HP, Smolkin ME, Theodorescu D, et al. Relationship between HLA class I antigen processing machinery component expression and the clinicopathologic characteristics of bladder carcinomas. Cancer Immunol Immunother. 2010;59(3):465–72. doi: 10.1007/s00262-009-0765-9.
  64. Seitz S, Hohla F, Schally AV, et al. Inhibition of estrogen receptor positive and negative breast cancer cell lines with a growth hormone-releasing hormone antagonist. Oncol Rep. 2008;20(5):1289–94.
  65. Mamessier E, Sylvain A, Thibult ML, et al. Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 2011;121(9):3609–22. doi: 10.1172/JCI45816.
  66. Busche A, Goldmann T, Naumann U, et al. Natural killer cell-mediated rejection of experimental human lung cancer by genetic overexpression of major histocompatibility complex class I chain-related gene A. Hum Gene Ther. 2006;17(2):135–46. doi: 10.1089/hum.2006.17.135.
  67. Platonova S, Cherfils-Vicini J, Damotte D, et al. Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 2011;71(16):5412–22. doi: 10.1158/0008-5472.CAN-10-4179.
  68. Jinushi M, Takehara T, Tatsumi T, et al. Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int J Cancer. 2003;104(3):354–61. doi: 10.1002/ijc.10966.
  69. Watson NF, Spendlove I, Madjd Z, et al. Expression of the stress-related MHC class I chain-related protein MICA is an indicator of good prognosis in colorectal cancer patients. Int J Cancer. 2006;118(6):1445–52. doi: 10.1002/ijc.21510.
  70. Sconocchia G, Spagnoli GC, Del Principe D, et al. Defective infiltration of natural killer cells in MICA/B-positive renal cell carcinoma involves beta(2)-integrin-mediated interaction. 2009;11(7):662–71. doi: 10.1593/neo.09296.
  71. Wu JD, Higgins LM, Steinle A, et al. Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest. 2004;114(4):560–8. doi: 10.1172/JCI22206.
  72. Salih HR, Antropius H, Gieseke F, et al. Functional expression and release of ligands for the activating immunoreceptor NKG2D in leukemia. 2003;102(4):1389–96. doi: 10.1182/blood-2003-01-0019.
  73. Diermayr S, Himmelreich H, Durovic B, et al. NKG2D ligand expression in AML increases in response to HDAC inhibitor valproic acid and contributes to allorecognition by NK-cell lines with single KIR-HLA class I specificities. 2008;111(3):1428–36. doi: 10.1182/blood-2007-07-101311.
  74. Sconocchia G, Lau M, Provenzano M, et al. The antileukemia effect of HLA-matched NK and NK-T cells in chronic myelogenous leukemia involves NKG2D-target-cell interactions. 2005;106(10):3666–72. doi: 10.1182/blood-2005-02-0479.
  75. Nuckel H, Switala M, Sellmann L, et al. The prognostic significance of soluble NKG2D ligands in B-cell chronic lymphocytic leukemia. 2010;24(6):1152–9. doi: 10.1038/leu.2010.74.
  76. Zhang B, Kracker S, Yasuda T, et al. Immune surveillance and therapy of lymphomas driven by Epstein-Barr virus protein LMP1 in a mouse model. 2012;148(4):739–51. doi: 10.1016/j.cell.2011.12.031.
  77. Girlanda S, Fortis C, Belloni D, et al. MICA expressed by multiple myeloma and monoclonal gammopathy of undetermined significance plasma cells costimulates pamidronate-activated gammadelta lymphocytes. Cancer Res. 2005;65(16):7502–8. doi: 10.1158/0008-5472.CAN-05-0731.
  78. Paschen A, Sucker A, Hill B, et al. Differential clinical significance of individual NKG2D ligands in melanoma: soluble ULBP2 as an indicator of poor prognosis superior to S100B. Clin Cancer Res. 2009;15(16):5208–15. doi: 10.1158/1078-0432.CCR-09-0886.
  79. Verhoeven DH, de Hooge AS, Mooiman EC, et al. NK cells recognize and lyse Ewing sarcoma cells through NKG2D and DNAM-1 receptor dependent pathways. Mol Immunol. 2008;45(15):3917–25. doi: 10.1016/j.molimm.2008.06.016.
  80. Friese MA, Platten M, Lutz SZ, et al. MICA/NKG2D-mediated immunogene therapy of experimental gliomas. Cancer Res. 2003;63(24):8996–9006.
  81. Raffaghello L, Prigione I, Airoldi I, et al. Downregulation and/or release of NKG2D ligands as immune evasion strategy of human neuroblastoma. 2004;6(5):558–68. doi: 10.1593/neo.04316.
  82. Chitadze G, Lettau M, Bhat J, et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int J Cancer. 2013;133(7):1557–66. doi: 10.1002/ijc.28174.
  83. Zhang T, Lemoi BA, Sentman CL. Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy. 2005;106(5):1544–51. doi: 10.1182/blood-2004-11-4365.
  84. Zhang T, Barber A, Sentman CL. Generation of antitumor responses by genetic modification of primary human T cells with a chimeric NKG2D receptor. Cancer Res. 2006;66(11):5927–33. doi: 10.1158/0008-5472.CAN-06-0130.
  85. Barber A, Zhang T, DeMars LR, et al. Chimeric NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. Cancer Res. 2007;67(10):5003–8. doi: 10.1158/0008-5472.CAN-06-4047.
  86. Barber A, Zhang T, Megli CJ, et al. Chimeric NKG2D receptor-expressing T cells as an immunotherapy for multiple myeloma. Exp Hematol. 2008;36(10):1318–28. doi: 10.1016/j.exphem.2008.04.010.
  87. Barber A, Meehan KR, Sentman CL. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D receptor-expressing T cells. Gene Ther. 2011;18(5):509–16. doi: 10.1038/gt.2010.174.
  88. Barber A, Rynda A, Sentman CL. Chimeric NKG2D expressing T cells eliminate immunosuppression and activate immunity within the ovarian tumor microenvironment. J Immunol. 2009;183(11):6939–47. doi: 10.4049/jimmunol.0902000.
  89. Zhang T, Sentman CL. Cancer immunotherapy using a bispecific NK receptor fusion protein that engages both T cells and tumor cells. Cancer Res. 2011;71(6):2066–76. doi: 10.1158/0008-5472.CAN-10-3200.
  90. Zhang T, Sentman CL. Mouse tumor vasculature expresses NKG2D ligands and can be targeted by chimeric NKG2D-modified T cells. J Immunol. 2013;190(5):2455–63. doi: 10.4049/jimmunol.1201314.
  91. Lehner M, Gotz G, Proff J, et al. Redirecting T cells to Ewing’s sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS One. 2012;7(2):e31210. doi: 10.1371/journal.pone.0031210.
  92. Song DG, Ye Q, Santoro S, et al. Chimeric NKG2D CAR-expressing T cell-mediated attack of human ovarian cancer is enhanced by histone deacetylase inhibition. Hum Gene Ther. 2013;24(3):295–305. doi: 10.1089/hum.2012.143.
  93. Kalos M, Levine BL, Porter DL, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842.
  94. Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra38. doi: 10.1126/scitranslmed.3005930.
  95. Meehan KR, Talebian L, Tosteson TD, et al. Adoptive cellular therapy using cells enriched for NKG2D+CD3+CD8+ T cells after autologous transplantation for myeloma. Biol Blood Marrow Transplant. 2013;19(1):129–37. doi: 10.1016/j.bbmt.2012.08.018.
  96. Nakajima J, Murakawa T, Fukami T, et al. A phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous gammadelta T cells. Eur J Cardiothorac Surg. 2010;37(5):1191–7. doi: 10.1016/j.ejcts.2009.11.051.
  97. Abe Y, Muto M, Nieda M, et al. Clinical and immunological evaluation of zoledronate-activated Vgamma9gammadelta T-cell-based immunotherapy for patients with multiple myeloma. Exp Hematol. 2009;37(8):956–68. doi: 10.1016/j.exphem.2009.04.008.
  98. Gattinoni L, Powell DJ Jr, Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6(5):383–93. doi: 10.1038/nri1842.
  99. June CH. Principles of adoptive T cell cancer therapy. J Clin Invest. 2007;117(5):1204–12. doi: 10.1172/JCI31446.
  100. Morgan RA, Chinnasamy N, Abate-Daga D, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36(2):133–51. doi: 10.1097/CJI.0b013e3182829903.
  101. Morgan RA, Yang JC, Kitano M, et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843–51. doi: 10.1038/mt.2010.24.
  102. Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. 2005;105(8):3051–7. doi: 10.1182/blood-2004-07-2974.
  103. Sentman CL, Meehan KR. NKG2D CARs as cell therapy for cancer. Cancer J. 2014;20(2):156–9. doi: 10.1097/PPO.0000000000000029.
  104. Lonez C, Hendlisz A, Shaza L, et al. Celyad’s novel CAR T-cell therapy for solid malignancies. Curr Res Transl Med. 2018;66(2):53–6. doi: 10.1016/j.retram.2018.03.001.
  105. Baumeister SH, Murad J, Werner L, et al. Phase I Trial of Autologous CAR T Cells Targeting NKG2D Ligands in Patients with AML/MDS and Multiple Myeloma. Cancer Immunol Res. 2019;7(1):100–12. doi: 10.1158/2326-6066.CIR-18-0307.
  106. Al-Homsi S, Purev E, Lewalle P, et al. Interim Results from the Phase I Deplethink Trial Evaluating the Infusion of a NKG2D CAR T-Cell Therapy Post a Non-Myeloablative Conditioning in Relapse or Refractory Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. 2019;134(Suppl_1):3844. doi: 10.1182/blood-2019-128267.
  107. Liu H, Wang S, Xin J, et al. Role of NKG2D and its ligands in cancer immunotherapy. Am J Cancer Res. 2019;9(10):2064–78.

Current View on Diagnosis and Treatment of Classical Ph-Negative Myeloproliferative Neoplasms

AL Melikyan1, IN Subortseva1, VA Shuvaev2,3, EG Lomaia4, EV Morozova5, LA Kuzmina1, OYu Vinogradova6,7,8, AYu Zaritskey4

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

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

3 VV Veresaev Municipal Clinical Hospital, 10 Lobnenskaya str., Moscow, Russian Federation, 127644

4 VA Almazov National Medical Research Center, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341

5 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

6 SP Botkin Municipal Clinical Hospital, 5 2-i Botkinskii pr-d, Moscow, Russian Federation, 125284

7 Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, 1 Samory Mashela str., Moscow, Russian Federation, 117997

8 NI Pirogov Russian National Research Medical University, 1 Ostrovityanova str., Moscow, Russian Federation, 117997

For correspondence: Anait Levonovna Melikyan, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: anoblood@mail.ru

For citation: Melikyan AL, Subortseva IN, Shuvaev VA, et al. Current View on Diagnosis and Treatment of Classical Ph-Negative Myeloproliferative Neoplasms. Clinical oncohematology. 2021;14(1):129–37. (In Russ).

DOI: 10.21320/2500-2139-2021-14-1-129-137


ABSTRACT

Classical Ph-negative myeloproliferative neoplasms (MPN) constitute a group of diseases including polycythemia vera, essential thrombocythemia, and primary myelofibrosis. Over the past decade, the approaches to understanding of MPN pathogenesis and therapy have considerably changed. At the same time, etiological factors and pathophysiological mechanisms of disease progress are being thoroughly studied. The improvement of diagnosis methods and new approaches to therapy can reduce complications and mortality risks. The review outlines the current diagnosis methods, such as the molecular genetic one, and provides prognostic scores. Different methods of conservative therapy are assessed. Special attention is paid to quality of life measurement and targeted treatment of patients.

Keywords: myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia, primary myelofibrosis, JAK2V617F, CALR, MPL, prognosis, constitutional symptoms, MPN10, ruxolitinib.

Received: September 1, 2020

Accepted: December 10, 2020

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Меликян А.Л., Туркина А.Г., Абдулкадыров К.М. и др. Клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз). Гематология и трансфузиология. 2014;59:31–56.
    [Melikyan AL, Turkina AG, Abdulkadyrov KM, et al. Clinical guidelines on diagnosis and therapy of Ph-negative myeloproliferative neoplasms (polycythemia vera, essential thrombocythemia, primary myelofibrosis). Gematologiya i transfuziologiya. 2014;59:31–56. (In Russ)]
  2. Меликян А.Л., Ковригина А.М., Суборцева И.Н. и др. Национальные клинические рекомендации по диагностике и терапии Ph-негативных миелопролиферативных заболеваний (истинная полицитемия, эссенциальная тромбоцитемия, первичный миелофиброз) (редакция 2018 г.) Гематология и трансфузиология. 2018;63(3):275–315.
    [Melikyan AL, Kovrigina AM, Subortseva IN, et al. National clinical guidelines on diagnosis and therapy of Ph-negative myeloproliferative neoplasms (polycythemia vera, essential thrombocythemia, primary myelofibrosis) (edition 2018). Gematologiya i transfuziologiya. 2018;63(3):275–315. (In Russ)]
  3. Абрамова А.В., Абдуллаев А.О., Азимова М.Х. и др. Алгоритмы диагностики и протоколы лечения заболеваний системы крови. В 2 томах. М.: Практика, 2018. Том 2.
    [Abramova AV, Abdullaev AO, Azimova MKh, et al. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. V 2 tomakh. (Diagnostic algorithms and treatment protocols in hematological diseases. 2 volumes.) Moscow: Praktika Publ.; 2018. 2. (In Russ)]
  4. Меликян А.Л., Суборцева И.Н., Галстян Г.М. Протокол дифференцированного посиндромного лечения больных первичным миелофиброзом. В кн.: Алгоритмы диагностики и протоколы лечения заболеваний системы крови. По ред. А.В. Абрамовой, А.О. Абдуллаева и др. В 2 томах. М.: Практика, 2018. Том 2. С. 777–802.
    [Melikyan AL, Subortseva IN, Galstyan GM. Protocol of differentiated syndromic treatment of patients with primary myelofibrosis. In: Abramova AV, Abdullaev AO, et al., eds. Algoritmy diagnostiki i protokoly lecheniya zabolevanii sistemy krovi. V 2 tomakh. (Diagnostic algorithms and treatment protocols in hematological diseases. 2 volumes.) Moscow: Praktika Publ.; 2018. Vol. 2. pр. 777–802. (In Russ)]
  5. Geyer H, Scherber R, Kosiorek H, et al. Symptomatic Profiles of Patients With Polycythemia Vera: Implications of Inadequately Controlled Disease. J Clin Oncol. 2016. 34(2):151–9. doi: 10.1200/JCO.2015.62.9337.
  6. Ионова Т.И., Анчукова Л.В., Виноградова О.Ю. и др. Качество жизни и спектр симптомов у больных миелофиброзом на фоне терапии: данные клинической практики. Гематология и трансфузиология. 2016;61(1):17–25. doi: 10.18821/0234-5730-2016-61-1-17-25.
    [Ionova TI, Anchukova LV, Vinogradova OYu, et al. Quality of life and symptom profile in patients with myelofibrosis undergoing treatment: Data of clinical practice. Gematologiya i transfuziologiya. 2016;61(1):17–25. doi: 10.18821/0234-5730-2016-61-1-17-25. (In Russ)]
  7. Xiao Z, Chang C-S, Morozova E, et al. Impact of myeloproliferative neoplasms (MPNS) and perceptions of treatment goals amongst physicians and patients in 6 countries: an expansion of the MPN landmark survey. 2019;3(s1):294–5. doi: 10.1097/01.hs9.0000561008.75001.e7.
  8. Cervantes F, Dupriez B, Pereira A, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. 2009;113(13):2895–901. doi: 10.1182/blood-2008-07-170449.
  9. Passamonti F, Cervantes F, Vannucchi AM, et al. Dynamic International Prognostic Scoring System (DIPSS) predicts progression to acute myeloid leukemia in primary myelofibrosis. 2010;116(15):2857–8. doi: 10.1182/blood-2010-06-293415.
  10. Gangat N, Caramazza D, Vaidya R, et al. DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol. 2011;29(4):392–7. doi: 10.1200/JCO.2010.32.2446.
  11. Vannucchi AM, Guglielmelli P, Rotunno G, et al. Mutation-Enhanced International Prognostic Scoring System (MIPSS) for primary myelofibrosis: an AGIMM & IWG-MRT project. 2014;124(21):405. doi: 10.1182/blood.v124.21.405.405.
  12. Guglielmelli P, Lasho TL, Rotunno G, et al. MIPSS70: Mutation-Enhanced International Prognostic Score System for transplantation-age patients with primary myelofibrosis. J Clin Oncol. 2018;36(4):310–8. doi: 10.1200/JCO.2017.76.4886.
  13. Passamonti F, Giorgino T, Mora B, et al. A clinical-molecular prognostic model to predict survival in patients with post polycythemia vera and post essential thrombocythemia myelofibrosis. 2017;31(12):2726–31. doi: 10.1038/leu.2017.169.
  14. Robin M, de Wreede LC, Wolschke C, et al. Long-term outcome after allogeneic hematopoietic cell transplantation for myelofibrosis. 2019;104(9):1782–8. doi: 10.3324/haematol.2018.205211.
  15. Барабанщикова М.В., Морозова Е.В., Байков В.В. и др. Аллогенная трансплантация гемопоэтических стволовых клеток при миелофиброзе. Клиническая онкогематология. 2016;9(3):279–86. doi: 10.21320/2500-2139-2016-9-3-279-286.
    [Barabanshchikova MV, Morozova EV, Baykov VV, et al. Allogeneic Hematopoietic Stem Cell Transplantation in Myelofibrosis. Clinical oncohematology. 2016;9(3):279–86. doi: 10.21320/2500-2139-2016-9-3-279-286. (In Russ)]
  16. Виноградова О.Ю., Шуваев В.А., Мартынкевич И.С. и др. Таргетная терапия миелофиброза. Клиническая онкогематология. 2017;10(4):471–8. doi: 10.21320/2500-2139-2017-10-4-471-478.
    [Vinogradova OYu, Shuvaev VA, Martynkevich IS, et al. Targeted Therapy of Myelofibrosis. Clinical oncohematology. 2017;10(4):471–8. doi: 10.21320/2500-2139-2017-10-4-471-478. (In Russ)]
  17. Руксолитиниб (инструкция по медицинскому применению). Доступно по: https://www.vidal.ru/drugs/molecule/2304. Ссылка активна на 22.10.2020.
    [Ruxolitinib (package insert). Available from: https://www.vidal.ru/drugs/molecule/2304. (accessed 22.10.2020) (In Russ)]
  18. Tefferi A, Cervantes F, Mesa R, et al. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European Leukemia Net (ELN) consensus report. 2013;122(8):1395–8. doi: 10.1182/blood-2013-03-488098.
  19. Ломаиа Е.Г., Сиордия Н.Т., Сендерова О.М. и др. Ранний ответ и отдаленные результаты терапии миелофиброза руксолитинибом: многоцентровое ретроспективное исследование в 10 центрах Российской Федерации. Клиническая онкогематология. 2020;13(3):335–45. doi: 10.21320/2500-2139-2020-13-3-335-345.
    [Lomaia EG, Siordiya NT, Senderova OM, et al. Early Response and Long-Term Outcomes of Ruxolitinib Therapy in Myelofibrosis: Multicenter Retrospective Study in 10 Centers of the Russian Federation. Clinical oncohematology. 2020;13(3):335–45. doi: 10.21320/2500-2139-2020-13-3-335-345. (In Russ)]
  20. Lomaia E, Siordiya N, Dimov G, et al. Early spleen response is a good prognostic factor of ruxolinib outcome in patients with myelofibrosis. 2019;3(S1):989. doi: 10.1097/01.hs9.0000567308.09016.52.