Effectiveness of the Initial Escalation of Immunochemotherapy in Patients with High Risk MALT-Lymphoma: Pilot Study Results

AK Smol’yaninova, NG Gabeeva, SA Tatarnikova, AV Belyaeva, AM Kovrigina, EG Gemdzhyan, EE Zvonkov

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

For correspondence: Anna Konstantinovna Smol’yaninova, MD, PhD, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; Tel.: +7(495)612-48-10; e-mail: annmo8@mail.ru.

For citation: Smol’yaninova AK, Gabeeva NG, Tatarnikova SA, et al. Effectiveness of the Initial Escalation of Immunochemotherapy in Patients with High Risk MALT-Lymphoma: Pilot Study Results. Clinical oncohematology. 2018;11(4):338–48.

DOI: 10.21320/2500-2139-2018-11-4-338-348


ABSTRACT

Background. MALT-lymphoma is usually characterized with an indolent course. The factors underlying the effectiveness of the standard chemotherapy in patients with MALT-lymphomas include MALT-IPI risk group and a high SUVmax according to the results of positron emission tomography (PET). All well-known MALT-lymphoma risk factors indirectly indicate a high risk of transformation to large cell lymphoma. The search for an effective chemotherapy continues.

Aim. To evaluate the effectiveness of the R-EPOCH/R-BAC escalated immunochemotherapy for MALT-lymphoma patients with poor prognosis factors.

Materials & Methods. In the period of 2016–2017 the study included 5 female MALT-lymphoma patients (the mean age of 41 years), of which 1 patient had an early relapse after surgery and 4 patients were newly diagnosed. Prior to therapy 4 patients were evaluated with PET. The mean SUVmax was 10.04. According to MALT-IPI 2 patients belonged to a high-risk group and 3 belonged to a middle-risk group. All the patients received R-EPOCH/R-BAC regimen therapy. A month after completing the treatment all the patients were again evaluated with PET.

Results. In 4 patients with 10–24 months follow-up complete remission was reported, which was confirmed by the results of histology and PET. The treatment of 1 patient was not completed. The immunotherapy was well tolerated by the patients. Hematological toxicity grade 3–4 occurred only after completing R-BAC treatment regimens. No severe infectious complications were reported.

Conclusion. MALT-lymphoma patients need to be evaluated in terms of all prognostic factors to identify the high-risk patients for whom escalated therapy is to be used already in the first line treatment. This pilot study of the use of R-EPOCH/R-BAC for treatment of MALT-lymphoma patients with poor prognosis factors yielded positive results and showed its acceptable tolerance.

Keywords: MALT-lymphoma, immunochemotherapy, positron emission tomography, prognosis factors, rituximab, ribomustin, cytarabine.

Received: April 10, 2018

Accepted: August 3, 2018

Read in PDF 


REFERENCES

  1. Rosand СB, Valla K, Flowers CR, et al. Effective management strategies for patients with marginal zone lymphoma. Fut Oncol. 2018;14(12):1213–22. doi: 10.2217/fon-2017-0480.

  2. Salar A, Domingo-Domenech E, Panizo C, et al. First-line response-adapted treatment with the combination of bendamustine and rituximab in patients with mucosa-associated lymphoid tissue lymphoma (MALT2008-01): a multi-centre, single-arm, phase 2 trial. Lancet Haematol. 2014;1(3):e104–е11. doi: 10.1016/s2352-3026(14)00021-0.

  3. Морозова А.К., Звонков Е.Е., Кравченко С.К. Лечение взрослых больных диффузной B-крупноклеточной лимфомой с первичным поражением костей по модифицированной программе NHL-BFM-90. В кн.: Программное лечение заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2012. Т. 2. С. 679–99.

    [Morozova AK, Zvonkov EE, Kravchenko SK. Treatment of adult patients with diffuse large B-cell lymphoma and primary bone lesions using the modified NHL-BFM-90 program. In: Savchenko VG, ed. Programmnoe lechenie zabolevanii sistemy krovi. (Programmed treatment of blood diseases.) Moscow: Praktika Publ.; 2012. Vol. 2. pp. 679–99. (In Russ)]

  4. Cheson BD, Horning SJ, Coiffier B, et al. Report of an international workshop to standardize response criteria for non-Hodgkin’s lymphomas. NCI sponsored International Working Group. J Clin Oncol. 1999;17(4):1244. doi: 10.1200/jco.1999.17.4.1244.

  5. Common Terminology Criteria for Adverse Events, version 3.0 (CTCAE). Published August 9, 2006. Available from: http://ctep.cancer.gov/protocolDevelopment/electronic_applications/docs/ctcaev3.pdf. (accessed05.2018).

  6. Thieblemont C, Cascione L, Conconi A, et al. A MALT lymphoma prognostic index. 2017;130(12):1409–17. doi: 10.1182/blood-2017-03-771915.

  7. Ekstrom SK, Vajdic CM, Falster M, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. Blood. 2008;111(8):4029–38. doi: 0.1182/blood-2007-10-119974.

  8. Thieblemont C, Berger F, Dumontet C, et al. Mucosa-associated lymphoid tissue lymphoma is a disseminated disease in one third of 158 patients analyzed. Blood. 2000;95(3):802–6.

  9. Yoon RG, Kim MY, Songb JW, et al. Primary Endobronchial Marginal Zone B-Cell Lymphoma of Bronchus-Associated Lymphoid Tissue: CT Findings in 7 Patients. Korean J Radiol. 2013;14(2):366–74. doi: 10.3348/kjr.2013.14.2.366.

  10. Zinzani PL, Pellegrini C, Gandolfi L, et al. Extranodal marginal zone B-cell lymphoma of the lung: experience with fludarabine and mitoxantrone-containing regimens. Hematol Oncol. 2012;31(4):183–8. doi: 10.1002/hon.2039.

  11. Borie R, Wislez M, Thabut G, et al. Clinical characteristics and prognostic factors of pulmonary MALT lymphoma. Eur Respir J. 2009;34(6):1408–16. doi: 10.1183/09031936.00039309.

  12. Rummel MJ, Kaiser U, Balser C. Bendamustine Plus Rituximab Versus Fludarabine Plus Rituximab In Patients with Relapsed Follicular, Indolent and Mantle Cell Lymphomas – Final Results of the Randomized Phase III Study NHL 2-2003 on Behalf of the StiL (Study Group Indolent Lymphomas, Germany). ASH Annual Meeting Abstracts. 2010;116:856.

  13. Rummel MJ, Niederle N, Maschmeyer G, et al. Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial. Lancet. 2013;381(9873):1203–10. doi: 10.1016/S0140-6736(12)61763-2.

  14. Zucca E, Conconi A, Martinelli G, et al. Final Results of the IELSG-19 Randomized Trial of Mucosa-Associated Lymphoid Tissue Lymphoma: Improved Event-Free and Progression-Free Survival With Rituximab Plus Chlorambucil Versus Either Chlorambucil or Rituximab Monotherapy. J Clin Oncol. 2017;35(17):1905–12. doi: 10.1200/jco.2016.70.6994.

  15. Zinzani PL, Stefoni V, Musuraca G, et al. Fludarabine-Containing Chemotherapy as Frontline Treatment of Nongastrointestinal Mucosa-Associated Lymphoid Tissue Lymphoma. Cancer. 2004;100(10):2190–4. doi: 10.1002/cncr.20237.

  16. Brown JR, Friedberg JW, Feng Y, et al. A phase 2 study of concurrent fludarabine and rituximab for the treatment of marginal zone lymphomas. Br J Haematol. 2009;145(6):741–8. doi: 10.1111/j.1365-2141.2009.07677.x.

  17. Prabhash K, Vikram GS, Nair R, et al. Fludarabine in lymphoproliferative malignancies: a single-centre experience. Natl Med J India. 2008;21(4):171–4.

  18. Cencini E, Fabbri A, Lauria F, et al. Long-term efficacy and toxicity of rituximab plus fludarabine and mitoxantrone (R-FM) for gastric marginal zone lymphoma: a single-center experience and literature review. Ann Hematol. 2018;97(5):821–9. doi: 10.1007/s00277-018-3243-7.

  19. Salar A, Domingo-Domenech E, Panizo C, et al. Long-term results of a phase II study of rituximab and bendamustine for mucosa-associated lymphoid tissue lymphoma. Blood. 2017;130(15):1772–4. doi: 10.1182/blood-2017-07-795302.

  20. Flinn I, van der Jagt R, Chang J, et al. First-line treatment of iNHL or MCL patients with BR or R-CHOP/R-CVP: results of the BRIGHT 5-year follow-up study. Hematol Oncol. 2017;35:140–1. doi: 10.1002/hon.2437_130.

  21. Treglia G, Zucca E, Sadeghi R, et al. Detection rate of fluorine-18-fluorodeoxyglucose positron emission tomography in patients with marginal zone lymphoma of MALT type: a meta-analysis. Hematol Oncol. 2015;33(3):113–24. doi: 10.1002/hon.2152.

  22. Carrillo-Cruz E, Marin-Oyaga V, de la Cruz VF, et al. Role of 18F-FDG-PET/CT in the management of marginal zone B cell lymphoma. Hematol Oncol. 2015;33(4):151–8. doi: 10.1002/hon.2181.

  23. Hwang JP, Lim I, Byun BH, et al. Prognostic value of SUVmax measured by pretreatment 18F-FDG PET/CT in patients with primary gastric lymphoma. Nucl Med Commun. 2016;37(12):1267–72. doi: 10.1097/mnm.0000000000000579.

  24. Noy A, Schoder H, Gonen M, et al. The majority of transformed lymphomas have high standardized uptake values (SUVs) on positron emission tomography (PET) scanning similar to diffuse large B-cell lymphoma (DLBCL). Ann Oncol.2009;20(3):508–12. doi: 10.1093/annonc/mdn657.

  25. Castegnaro S, Visco C, Chieregato K, et al. Cytosine arabinoside potentiates the apoptotic effect of bendamustine on several B- and T-cell leukemia/lymphoma cells and cell lines. Leuk Lymphoma. 2012;53(11):2262–8. doi: 10.3109/10428194.2012.688200.

  26. Visco C, Chiappella A, Nassi L, et al. Rituximab, bendamustine, and low-dose cytarabine as induction therapy in elderly patients with mantle cell lymphoma: a multicentre, phase 2 trial from Fondazione Italiana Linfomi. Lancet Haematol. 2017;4(1):e15–e23. doi: 10.1016/S2352-3026(16)30185-5.

 

EBV-Positive Lymphoproliferative Diseases: A New Concept and Differential Diagnosis (Literature Review and Case Reports)

АM Kovrigina

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

For correspondence: Professor Alla Mikhailovna Kovrigina, PhD in Biology, 4 Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: kovrigina.alla@gmail.com

For citation: Kovrigina AM. EBV-Positive Lymphoproliferative Diseases: A New Concept and Differential Diagnosis (Literature Review and Case Reports). Clinical oncohematology. 2018;11(4):326–37.

DOI: 10.21320/2500-2139-2018-11-4-326-337


ABSTRACT

In recent years increasing attention focuses on the concept of EBV-positive lymphoproliferative diseases related to primary or secondary immunodeficiency resulting from immunosuppressive therapy and persistent infections. Due to the progress of treatment methods in oncohematology and oncology this pathology also occurs as a delayed event when new surgical and therapeutic approaches are applied. The paper presents proof for the pathogenetic significance of Epstein-Barr virus (EBV) in the pathology under consideration with its various clinical manifestations and describes the evolution of knowledge on posttransplant lymphoproliferative disorders with their morphological classification underlying EBV+ lymphoproliferative diseases. The WHO Classification of Tumours of Hematopoietic and Lymphoid Tissues revised in 2017 includes new forms of EBV+ lymphoproliferative diseases (mucocutaneous ulcer, T- and NK-cell childhood lymphoproliferative disorders including cutaneous and systemic forms of chronic active EBV infection) and EBV+ large B-cell lymphomas (unspecified and fibrin-associated diffuse large B-cell lymphomas). The paper summarizes major characteristics of these diseases and exemplifies them by the author’s own experience.

Keywords: B-, T-, NK-cell lymphoproliferative diseases, Epstein-Barr virus (EBV), immunodeficiency, immune imbalance, immunosuppression, morphology, diagnosis.

Received: July 20, 2018

Accepted: September 25, 2018

Read in PDF 


REFERENCES

  1. Ibrahim HA, Naresh KN. Posttransplant lymphoproliferative disorders. Adv Hematol. 2012;2012:230173. doi: 10.1155/2012/230173.

  2. Swerdlow SH, Campo E, Harris NL, et al. (eds) WHO classification of tumours of haematopoietic and lymphoid tissues. 4th edition. Lyon: IARC Press; 2008.

  3. Swerdlow SH, Campo E, Harris NL, et al. (eds) WHO classification of tumours of haematopoietic and lymphoid tissues. Revised 4th edition. Lyon: IARC Press; 2017.

  4. Gratzinger D, Jaffe ES, Chadburn A. Primary/Congenital Immunodeficiency: 2015 SH/EAHP Workshop Report—Part 5. Am J Clin Pathol. 2017;147(2):204–16. doi: 10.1093/AJCP/AQW215.

  5. Epstein MA, Achong BG, Barr YM. Virus particles in cultured lymphoblasts from Burkitt’s lymphoma. Lancet. 1964;1:702–3.

  6. Гурцевич В.Э. Роль вируса Эпштейна—Барр в онкогематологических заболеваниях человека. Клиническая онкогематология. 2010;3(3):222–32.

    [Gurtsevich VE. Role of Epstein-Barr virus in human hematological malignancies. Klinicheskaya onkogematologiya. 2010;3(3):222–32. (In Russ)]

  7. Tse E, Kwong Y-L. Epstein Barr virus-associated lymphoproliferative diseases: the virus as a therapeutic target. Exp Mol Med. 2015;47(1):e136. doi: 10.1038/emm.2014.102.

  8. Roschewski M, Wilson WH. EBV-associated lymphomas in adults. Best Pract Res Clin Haematol. 2012;25(1):75–89. doi: 10.1016/j.beha.2012.01.005.

  9. Shannon-Lowe C, Rickinson AB, Bell AI. Epstein–Barr virus-associated lymphomas. Philos Trans R Soc Lond B Biol Sci. 2017;372(1732):20160271. doi: 10.1098/rstb.2016.0271.

  10. Natkunam Y, Goodlad JR, Chadburn A, et al. EBV-Positive B-Cell Proliferations of Varied Malignant Potential: 2015 SH/EAHP Workshop Report—Part 1. Am J Clin Pathol. 2017;147(2):129–52. doi: 10.1093/AJCP/AQW214.

  11. Гаврилина О.А., Троицкая В.В., Звонков Е.Е. и др. Лимфопролиферативное EBV-позитивное заболевание с поражением центральной нервной системы, ассоциированное с иммуносупрессией после органной трансплантации: длительная ремиссия без химиотерапевтического лечения. Терапевтический архив. 2017;89(7):69–75. doi: 10.17116/terarkh201789769-75.

    [Gavrilina OA, Troitskaya VV, Zvonkov EE, et al. EBV-positive central nervous system lymphoproliferative disease associated with immunosuppression after organ transplantation: long-term remission without chemotherapy. Terapevticheskii arkhiv. 2017;89(7):69–75. doi: 10.17116/terarkh201789769-75. (In Russ)]

  12. Boyer DF, McKelvie PA, de Leval L, et al. Fibrin-associated EBV-positive large B-cell lymphoma: an indolent neoplasm with features distinct from diffuse large B-cell lymphoma associated with chronic inflammation. Am J Surg Pathol. 2017;41(3):299–312. doi: 10.1097/PAS.0000000000000775.

  13. Cohen M, De Matteo E, Narbaitz M, et al. Epstein-Barr virus presence in pediatric diffuse large B-cell lymphoma reveals a particular association and latency patterns: Analysis of viral role in tumor microenvironment. Int J Cancer. 2013;132(7):1572–80. doi: 10.1002/ijc.27845.

  14. Uccini S, Al-Jadiry MF, Scarpino S, et al. Epstein-Barr virus-positive diffuse large B-cell lymphoma in children: A disease reminiscent of Epstein-Barr virus-positive diffuse large B-cell lymphoma of the elderly. Hum Pathol. 2015;46(5):716–24. doi: 10.1016/j.humpath.2015.01.011.

  15. Nicolae A, Pittaluga S, Abdullah S, et al. EBV-positive large B-cell lymphomas in young patients: A nodal lymphoma with evidence for a tolerogenic immune environment. Blood. 2015;126(7):863–72. doi: 10.1182/blood-2015-02-630632.

  16. Kunitomi A, Hasegawa Y, Asano N, et al. EBV-positive reactive hyperplasia progressed into EBV-positive diffuse large B-cell lymphoma of the elderly over a 6-year period. Intern Med. 2018;57(9):1287–90. doi: 10.2169/internalmedicine.9112-17.

  17. de la Hera Magallanes AI, Montes-Moreno S, Hernandez SG, et al. Early phase of Epstein-Barr virus (EBV)-positive diffuse large B cell lymphoma of the elderly mimicking EBV-positive reactive follicular hyperplasia. Histopathology. 2011;59(3):571–5. doi: 10.1111/j.1365-2559.2011.03950.x.

  18. Roberts TK, Chen X, Liao JJ. Diagnostic and therapeutic challenges of EBV-positive mucocutaneous ulcer: a case report and systematic review of the literature. Exp Hematol Oncol. 2016;5(1):13. doi: 10.1186/s40164-016-0042-5.

  19. Dojcinov SD, Venkataraman G, Raffeld M, et al. EBV positive mucocutaneous ulcer–a study of 26 cases associated with various sources of immunosuppression. Am J Surg Pathol. 2010;34(3):405–17. doi: 10.1097/PAS.0b013e3181cf8622.

  20. Docinov SD, Venkataraman G, Pittaluga S, et al. Age-related EBV-associated lymphoproliferative disorders in the Western population: a spectrum of reactive lymphoid hyperplasia and lymphoma. Blood. 2011;117(8):4726–35. doi: 10.1182/blood-2010-12-323238.

  21. Gratzinger D, Jong D, Jaffe ES, et al. T- and NK-Cell Lymphomas and Systemic Lymphoproliferative Disorders and the Immunodeficiency Setting: 2015 SH/EAHP Workshop Report—Part 4. Am J Clin Pathol. 2017;147(2):188–203. doi: 10.1093/AJCP/AQW213.

Primary Mediastinal (Thymic) Large B-Cell Lymphoma: Diagnostics of Extramediastinal Lesions and Treatment Opportunities

YaK Mangasarova, AU Magomedova, AM Kovrigina, IE Kostina, ES Nesterova, LG Gorenkova, AE Misyurina, OV Margolin, SK Kravchenko

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

For correspondence: Yana Konstantinovna Mangasarova, MD, PhD, 4a Novyi Zykovskii pr-d, Moscow, Russian Federation, 125167; e-mail: v.k.jana@mail.ru.

For citation: Mangasarova YaK, Magomedova AU, Kovrigina AM, et al. Primary Mediastinal (Thymic) Large B-Cell Lymphoma: Diagnostics of Extramediastinal Lesions and Treatment Opportunities. Clinical oncohematology. 2018;11(3):220–26.

DOI: 10.21320/2500-2139-2018-11-3-220-226


ABSTRACT

Background. Current diagnostic methods and the introduction of molecular investigations into clinical practice allow to improve the understanding of classical primary mediastinal (thymic) large B-cell lymphoma (PMBCL).

Aim. To investigate clinical characteristics of PMBCL patients with extramediastinal lesions.

Materials & Methods. The study was performed from 2007 to 2017 in the National Medical Hematology Research Center and included 157 PMBCL patients. The data of 16 (10.2 %; 4 men and 12 women) patients with extramediastinal lesions were analyzed; the median age was 27 years (range 23–69).

Results. The extramediastinal lesions were found in pancreas (6; 37.5 %), kidneys (5; 31.2 %), ovaries (3; 18.7 %), liver (3; 18.7 %), bone marrow (3; 18.7 %), and breasts (2; 12 %); the lesions in stomach, bones, soft tissues, spleen, adrenals, and small pelvis were observed each in a single case. In 15 of 16 cases extramediastinal lesions were accompanied by involvement of superior mediastinum, and only 1 patient had an isolated lesion in thoracic soft tissues without mediastinal involvement. The samples of 8 out of 16 patients were analyzed using PCR. In all samples overexpression of 2 or more genes (JAK2, TRAF1, MAL, PDL1, PDL2) was determined which allowed to confirm, and in some cases to revise the diagnosis of PMBCL. Overall 5-year survival (93 %) of patients with classical PMBCL with thoracic involvement was similar to the cohort with extramediastinal lesions. All unfavourable events (progression/relapse) were identified at an early stage, i.e. within a year after the completion of therapy.

Conclusion. PMBCL patients can have not only superior mediastinum involvement, but extramediastinal lesions as well, including bone marrow. The spreading of the disease beyond superior mediastinum should be differentiated from diffuse large B-cell lymphoma using standard evaluation methods, and molecular analysis in some cases.

Keywords: primary mediastinal (thymic) large B-cell lymphoma, extramediastinal lesions, bone marrow involvement.

Received: January 22, 2018

Accepted: April 15, 2018

Read in PDF 


REFERENCES

  1. Lichtenstein AK, Levine A, Taylor CR, et al. Primary mediastinal lymphoma in adults. Am J Med. 1980;68(4):509–14. doi: 10.1016/0002-9343(80)90294-6.
  2. 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.
  3. Rosenwald A, Wright G, Leroy K, et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diff use large B cell lymphoma related to Hodgkin lymphoma. J Exp Med. 2003;198(6):851–62. doi: 10.1084/jem.20031074.
  4. Zhang B, Wang Z, Li T, et al. NF-kappaB2 mutation targets TRAF1 to induce lymphomagenesis. Blood. 2007;110(2):743–51. doi: 10.1182/blood-2006-11-058446.
  5. Meier C, Hoeller S, Bourgau C, et al. Recurrent numerical aberrations of JAK2 and deregulation of the JAK2-STAT cascade in lymphomas. Mod Pathol. 2009;22(3):476–87. doi: 10.1038/modpathol.2008.207.
  6. 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.
  7. Joos S, Granzow M, Holtgreve-Grez H, et al. Hodgkin’s lymphoma cell lines are characterized by frequent aberrations on chromosomes 2p and 9p including REL and JAK2. Int J Cancer. 2003;103(4):489–95. doi: 10.1002/ijc.10845.
  8. Мангасарова Я.К., Мисюрин А.В., Магомедова А.У. и др. Молекулярная диагностика первичной медиастинальной В-клеточной лимфомы и диффузной В-крупноклеточной лимфомы с первичным вовлечением лимфатических узлов средостения. Клиническая онкогематология. 2011;4(2):142–5.
    [Mangasarova YaK, Misyurin AV, Magomedova AU, et al. Molecular diagnostics of primary mediastinal B-cell lymphoma and diffuse large B-cell lymphoma with primary involvement of mediastinal lymph nodes. Klinicheskaya onkogematologiya. 2011;4(2):142–5. (In Russ)]
  9. Twa DD, 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. Bentz M, Barth TF, Bruderlein S, et al. Gain of chromosome arm 9p is characteristic of primary mediastinal B-cell lymphoma (MBL): comprehensive molecular cytogenetic analysis and presentation of a novel MBL cell line. Genes Chromos Cancer. 2001;30(4):393–401. doi: 1002/1098-2264(2001)9999:9999<::aid-gcc1105>3.0.co;2-i.
  11. Huang J, Sanger G, Greiner T, et al. The t(14;18) defines a unique subset of large B-cell lymphoma with a germinal center B-cell gene expression profile. Blood. 2002;99(7):2285–90. doi: 10.1182/blood.v99.7.2285.
  12. Barans S, Connor S, Evans P, et al. Rearrangement of the BCL-6 locus at 3q27 is an independent poor prognostic factor in nodal diffuse large B-cell lymphoma. Br J Hematol. 2002;117(2):322–32. doi: 1046/j.1365-2141.2002.03435.x.
  13. Ковригина А.М., Пробатова Н.А. Лимфома Ходжкина и крупноклеточные лимфомы. М.: МИА, 2007; 108–24.
    [Kovrigina AM, Probatova NA. Limfoma Khodzhkina i krupnokletochnye limfomy. (Hodgkin’s lymphoma and large cell lymphomas.) Moscow: MIA Publ.; 2007. pp. 108–24. (In Russ)]
  14. Yuan J, Wright G, Rosenwald A, et al. Identification of Primary Mediastinal Large B-cell Lymphoma at Nonmediastinal Sites by Gene Expression Profiling. Am J Surg Pathol. 2015;39(10):1322–30. doi: 10.1097/PAS.0000000000000473.
  15. Bishop PC, Wilson WH, Pearson D, et al. CNS involvement in primary mediastinal large B-cell lymphoma. J Clin Oncol. 1999;17(8):2479–85. doi: 10.1200/jco.1999.17.8.2479.
  16. Магомедова А.У., Фастова Е.А., Ковригина А.М. и др. Поражение костного мозга при первичной медиастинальной В-крупноклеточной лимфоме. Терапевтический архив. 2017;89(7):65–8. doi: 10.17116/terarkh201789765-68.
    [Magomedova AU, Fastova EA, Kovrigina AM, et al. Bone marrow involvement in primary mediastinal B-cell lymphoma. Terapevticheskii arkhiv. 2017;89(7):65–8. doi: 17116/terarkh201789765-68. (In Russ)]

Polymerase Chain Reaction for Prognosis Assessment and Monitoring of the Epstein-Barr Virus-Associated Hodgkin’s Lymphoma

MA Katin1, IV Zhil’tsov1, VM Semenov1, DK Novik2

1 Vitebsk State Medical University, 27 Frunze pr-t, Vitebsk, Republic of Belarus, 210023

2 Republican Applied Research Center for Radiation Medicine and Human Ecology, 290 Il’icha str., Gomel, Republic of Belarus, 246040

For correspondence: Prof. Ivan Viktorovich Zhil’tsov, MD, PhD, 27 Frunze pr-t, Vitebsk, Republic of Belarus, 210023; Tel.: +375(29)7104368-93-29; e-mail: zhyltsou@tut.by

For citation: Katin NA, Zhil’tsov IV, Semenov VM, Novik DK. Polymerase Chain Reaction for Prognosis Assessment and Monitoring of the Epstein-Barr Virus-Associated Hodgkin’s Lymphoma. Clinical oncohematology. 2018;11(2):182–6.

DOI: 10.21320/2500-2139-2018-11-2-182-186


ABSTRACT

The review provides the analysis of 34 papers on polymerase chain reaction (PCR) as a method of the Epstein-Barr virus (EBV) DNA detection in biological material of patients with EBV-associated cancer diseases including Hodgkin’s lymphoma (HL). A comparative analysis of different methods of EBV DNA detection in biological material is presented. EBV is associated with HL in 20 to 100 % of cases depending on a geographic region and HIV status. EBV-associated HLs are characterized by latency type II. EBV is found in all the atypical cells and can be detected in blood of EBV-associated HL patients by means of the PCR method. The review includes the results of studies on EBV detection using the PCR method compared to in situ methods of hybridization and immunohistochemistry in various EBV-associated cancer diseases including HL. The obtained data indicate that PCR can be used for quantitative determination of EBV DNA in blood plasma of HL patients for therapeutic efficacy monitoring and prognosis assessment of disease and relapses. Quantitative determination of EBV DNA in blood plasma of HL patients using the real time PCR method is a promising technique. Its further practical application requires standardization of the method, larger trials, and comparison to positron emission tomography.

Keywords: Epstein-Barr virus, Hodgkin’s lymphoma, polymerase chain reaction.

Received: December 20, 2017

Accepted: February 28, 2018

Read in PDF 


REFERENCES

  1. Thorley-Lawson DA. Epstein-Barr virus: exploiting the immune system. Nat Rev Immunol. 2001;1(1):75–82. doi: 10.1038/35095584.
  2. Murata T. Regulation of Epstein-Barr virus reactivation from latency. Microbiol Immunol. 2014;58(6):307–17. doi: 10.1111/1348-0421.12155.
  3. Shannon-Lowe C, Rickinson AB, Bell AI. Epstein–Barr virus-associated lymphomas. Philos Trans R Soc B Biol Sci. 2017;372(1732):20160271.
  4. Young LS, Murray PG. Epstein-Barr virus and oncogenesis: from latent genes to tumours. Oncogene. 2003;22(33):5108–21. doi: 10.1038/sj.onc.1206556.
  5. Rickinson AB, Rowe M, Hart IJ, et al. T-cell-mediated regression of ‘spontaneous’ and of Epstein-Barr virus-induced B-cell transformation in vitro: studies with cyclosporin A. Cell Immunol. 1984;87(2):646–58. doi: 1016/0008-8749(84)90032-7.
  6. Mancao C, Hammerschmidt W. Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood. 2007;110(10):3715–21. doi: 1182/blood-2007-05-090142.
  7. Massini G, Siemer D, Hohaus S. EBV in Hodgkin Lymphoma. Mediterr J Hematol Infect Dis. 2009;1(2):e2009013. doi: 10.4084/MJHID.2009.013.
  8. Flavell KJ, Murray PG. Hodgkin’s disease and the Epstein-Barr virus. Mol Pathol. 2000;53(5):262–9. doi:1136/mp.53.5.262.
  9. Weinreb M, Day PJ, Niggli F, et al. The role of Epstein-Barr virus in Hodgkin’s disease from different geographical areas. Arch Dis Child. 1996;74(1):27–31. doi: 10.1136/adc.74.1.27.
  10. Gandhi MK, Tellam JT, Khanna R. Epstein-Barr virus-associated Hodgkin’s lymphoma. Br J Haematol. 2004;125(3):267–81. doi: 10.1111/j.1365-2141.2004.04902.x.
  11. Glaser SL, Lin RJ, Stewart SL, et al. Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70(4):375–82. doi: 10.1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.0.co;2-t.
  12. Dolcetti R, Boiocchi M, Gloghini A, et al. Pathogenetic and histogenetic features of HIV-associated Hodgkin’s disease. Eur J Cancer. 2001;37(10):1276–87. doi: 10.1016/S0959-8049(01)00105-8.
  13. Gulley ML, Tang W. Laboratory Assays for Epstein-Barr Virus-Related Disease. J Mol Diagn. 2008;10(4):279–92. doi: 2353/jmoldx.2008.080023.
  14. Maurmann S, Fricke L, Wagner H-J, et al. Molecular parameters for precise diagnosis of asymptomatic Epstein-Barr virus reactivation in healthy carriers. J Clin Microbiol. 2003;41(12):5419–28. doi: 10.1128/jcm.41.12.5419-5428.2003.
  15. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17(9):2885–92. doi: 10.1158/1078-0432.CCR-10-3327.
  16. Qi Z-L, Han X-Q, Hu J, et al. Comparison of three methods for the detection of Epstein-Barr virus in Hodgkin’s lymphoma in paraffin-embedded tissues. Mol Med Rep. 2013;7(1):89–92. doi: 10.3892/mmr.2012.1163.
  17. Ryan JL, Fan H, Glaser SL, et al. Epstein-Barr virus quantitation by real-time PCR targeting multiple gene segments: a novel approach to screen for the virus in paraffin-embedded tissue and plasma. J Mol Diagn. 2004;6(4):378–85. doi: 1016/S1525-1578(10)60535-1.
  18. Lo YM, Leung SF, Chan LY, et al. Kinetics of plasma Epstein-Barr virus DNA during radiation therapy for nasopharyngeal carcinoma. Cancer Res. 2000;60(9):2351-5.
  19. To EWH, Chan KCA, Leung S-F, et al. Rapid clearance of plasma Epstein-Barr virus DNA after surgical treatment of nasopharyngeal carcinoma. Clin Cancer Res. 2003;9(9):3254–9.
  20. Zhao F, Liu X, Chen X, et al. Levels of plasma Epstein-Barr virus DNA prior and subsequent to treatment predicts the prognosis of nasopharyngeal carcinoma. Oncol Lett. 2015;10(5):2888–94. doi: 3892/ol.2015.3628.
  21. Au W-Y, Pang A, Choy C, et al. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104(1):243–9. doi: 1182/blood-2003-12-4197.
  22. Lei KIK, Chan LYS, Chan W-Y, et al. Diagnostic and prognostic implications of circulating cell-free Epstein-Barr virus DNA in natural killer/T-cell lymphoma. Clin Cancer Res. 2002;8(1):29–34.
  23. Hohaus S, Giachelia M, Massini G, et al. Cell-free circulating DNA in Hodgkin’s and non-Hodgkin’s lymphomas. Ann Oncol. 2009;20(8):1408–13. doi: 1093/annonc/mdp006.
  24. Gallagher A, Armstrong AA, MacKenzie J, et al. Detection of Epstein-Barr virus (EBV) genomes in the serum of patients with EBV-associated Hodgkin’s disease. Int J Cancer. 1999;84(4):442–8. doi:1002/(sici)1097-0215(19990820)84:4<442::aid-ijc20>3.0.co;2-j.
  25. Chan KCA, Zhang J, Chan ATC, et al. Molecular characterization of circulating EBV DNA in the plasma of nasopharyngeal carcinoma and lymphoma patients. Cancer Res. 2003;63(9):2028–32.
  26. Lei KIK, Chan LYS, Chan WY, et al. Quantitative analysis of circulating cell-free Epstein-Barr virus (EBV) DNA levels in patients with EBV-associated lymphoid malignancies. Br J Haematol. 2000;111(1):239–46. doi:1111/j.1365-2141.2000.02344.x.
  27. Gandhi MK, Lambley E, Burrows J, et al. Plasma Epstein-Barr virus (EBV) DNA is a biomarker for EBV-positive Hodgkin’s lymphoma. Clin Cancer Res. 2006;12(2):460–4. doi: 1158/1078-0432.CCR-05-2008.
  28. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121(18):3547–53. doi: 1182/blood-2012-09-454694.
  29. Sinha M, Rao CR, Shafiulla M, et al. Plasma Epstein Barr viral load in adult-onset Hodgkin lymphoma in South India. Hematol Oncol Stem Cell Ther. 2016;9(1):8–13. doi: 10.1016/j.hemonc.2015.11.004.
  30. Spacek M, Hubacek P, Markova J, et al. Plasma EBV-DNA monitoring in Epstein-Barr virus-positive Hodgkin lymphoma patients. APMIS. 2011;119(1):10–6. doi: 1111/j.1600-0463.2010.02685.x.
  31. Welch JJG, Schwartz CL, Higman M, et al. Epstein-Barr virus DNA in serum as an early prognostic marker in children and adolescents with Hodgkin lymphoma. Blood Adv. 2017;1(11):681–4. doi: 1182/bloodadvances.2016002618.
  32. De Paoli P, Pratesi C, Bortolin MT. The Epstein Barr virus DNA levels as a tumor marker in EBV-associated cancers. J Cancer Res Clin Oncol. 2007;133(11):809–15. doi: 1007/s00432-007-0281-2.
  33. Le Q-T, Jones CD, Yau T-K, et al. A Comparison Study of Different PCR Assays in Measuring Circulating Plasma Epstein-Barr Virus DNA Levels in Patients with Nasopharyngeal Carcinoma. Clin Cancer Res. 2005;11(16):5700–7. doi: 1158/1078-0432.CCR-05-0648.
  34. Bortolin MT, Pratesi C, Dolcetti R, et al. Clinical value of Epstein–Barr virus DNA levels in peripheral blood samples of Italian patients with undifferentiated carcinoma of nasopharyngeal type. Cancer Lett. 2006;233(2):247–54. doi: 10.1016/j.canlet.2005.03.015.

Evolution of Anti-Cancer Treatment and its Impact on Surrogate Prognostic Factors in Multiple Myeloma

AS Luchinin1, SV Semochkin2, NV Minaeva1, NM Pozdeev1, IV Paramonov1

1 Kirov Research Institute of Hematology and Transfusiology, 72 Krasnoarmeiskaya str., Kirov, Russian Federation, 610027

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

For correspondence: Aleksandr Sergeevich Luchinin, 72 Krasnoarmeiskaya str., Kirov, Russian Federation, 610027; Tel.: +7(919)506-87-86; e-mail: glivec@mail.ru

For citation: Luchinin AS, Semochkin SV, Minaeva NV, et al. Evolution of Anti-Cancer Treatment and its Impact on Surrogate Prognostic Factors in Multiple Myeloma. Clinical oncohematology. 2018;11(2):175–81.

DOI: 10.21320/2500-2139-2018-11-2-175-181


ABSTRACT

Aim. To assess prognostic value of surrogate clinical and laboratory markers in current therapy of multiple myeloma (MM).

Materials & Methods. The analysis included 567 patients (215 men and 352 women), the Kirov region inhabitants with newly diagnosed MM over the period from January 1, 1994 to December 31, 2016. The median age was 64 years (range 29–90). Patients were divided into two groups: the first group received treatment from 1994 to 2005 (n = 269), the second group received treatment from 2006 to 2016 (n = 298). Impact of factors on overall survival (OS) was evaluated by multivariate logistic regression analysis using the Cox method.

Results. Over the period from 2006 to 2016 the number of patients treated with traditional chemotherapy decreased from 78.4 to 32.5 %. At the same time the number of patients treated with bortezomib-based regimens increased from 1.9 to 56.3 % and autologous hematopoietic stem cell transplantation (auto-HSCT) protocols — from 1.4 to 14.0 %. Median OS over the period from 1994 to 2005 was 27 months. It increased to 55 months in the period of 2006–2016. In the reference decades 5-year overall survival increased from 21 % (95% confidence interval [95% CI] 17–27 %) to 47 % (95% CI 39–55 %), respectively (hazard ratio [HR] 0.51; 95% CI 0.41–0.64; < 0,0001). In patients treated with bortezomib-based regimens over the period from 2006 to 2016 median OS increased to 73 months compared to 27 months in 1994–2005. In patients aged ≤ 65 years and treated with auto-HSCT median OS was not reached, and median OS in patients without auto-HSCT treatment was 54 months.

Conclusions. Surrogate prognostic markers, such as the age over 65, hemoglobin level < 100 g/L, β2-microglobulin ≥ 6 mg/L, serum creatinine ≥ 177 µmol/L and stage III according to ISS and Durie-Salmon, are unfavourable predictors of survival of MM patients.

Keywords: multiple myeloma, prognosis, bortezomib, auto-HSCT, overall survival.

Received: December 21, 2017

Accepted: February 25, 2018

Read in PDF 


REFERENCES

  1. Менделеева Л.П., Вотякова О.М., Покровская О.С. и др. Национальные клинические рекомендации по диагностике и лечению множественной миеломы. Гематология и трансфузиология. 2016;61(1, прил. 2):1–24. doi: 10.18821/0234-5730-2016-61-1(Прил.2).[Mendeleeva OP, Votyakova OM, Pokrovskaya OS, et al. National clinical recommendations in diagnosis and treatment of multiple myeloma. Gematologiya i transfuziologiya. 2016;61(1, Suppl. 2):1–24. doi: 10.18821/0234-5730-2016-61-1(Прил.2). (In Russ)]
  2. Бессмельцев С.С., Абдулкадыров К.М. Множественная миелома: руководство для врачей. М.: МК, 2016. 504 с.[Bessmel’tsev SS, Abdulkadyrov KM. Mnozhestvennaya mieloma: rukovodstvo dlya vrachei. (Multiple myeloma: manual for physicians.) Moscow: MK Publ.; 2016. 504 p. (In Russ)]
  3. Ghobrial IM, Landgren O. How I treat smoldering multiple myeloma. Blood. 2014;124(23):3380–8. doi: 10.1182/blood-2014-08-551549.
  4. Hsu P, Lin TW, Gau JP, et al. Risk of early mortality in patients with newly diagnosed multiple myeloma. Medicine. 2016;94(50):e2305. doi: 1097/MD.0000000000002305.
  5. Pulte D, Jansen L, Castro FA, et al. Trends in survival of multiple myeloma patients in Germany and the United States in the first decade of the 21st century. Br J Haematol. 2015;171(2):189–96. doi: 10.1111/bjh.13537.
  6. Libby E, Garcia D, Quintana D, et al. Disease-specific survival for patients with multiple myeloma: significant improvements over time in all age groups. Leuk Lymphoma. 2014;55(12):2850–7. doi: 10.3109/10428194.2014.89770
  7. Митина Т.А., Голенков А.К., Трифонова Е.В. и др. Эффективность леналидомида, бортезомиба и преднизолона при лечении пациентов с рецидивирующей и рефрактерной множественной миеломой. Онкогематология. 2015;4(10):8–14. doi: 10.17650/1818-8346-2015-10-4-8-14.[Mitina TA, Golenkov AK, Trifonova EV, et al. Efficacy of lenalidomide, bortezomib, and prednisolone in patients with relapsed or refractory multiple myeloma. Oncohematology. 2015;4(10):8–14. doi: 10.17650/1818-8346-2015-10-4-8-14. (In Russ)]
  8. Hungria VTМ, Maiolino A, Martinez G, et al. Confirmation of the utility of the International Staging System and identification of a unique pattern of disease in Brazilian patients with multiple myeloma. Haematologica. 2008;93(5):791–2. doi: 10.3324/haematol.11637.
  9. Lu J, Lu J, Liu A, et al. The applicability of the International Staging System in Chinese patients with multiple myeloma receiving bortezomib or thalidomide-based regimens as induction therapy: a multicenter analysis. Biomed Res Int. 2015;2015:1–7. doi: 10.1155/2015/856704.
  10. Dosani T, Covut F, Beck R, et al. Significance of the absolute lymphocyte/monocyte ratio as a prognostic immune biomarker in newly diagnosed multiple myeloma. Blood Cancer J. 2017;7(6):e579. doi: 10.1038/bcj.2017.60.
  11. Hanbali A, Hassanein M, Rasheed W, et al. The evolution of prognostic factors in multiple myeloma. Adv Hematol. 2017;2017:1–11. doi: 10.1155/2017/4812637.
  12. Chng WJ, Dispenzieri A, Chim CS, et al. IMWG consensus on risk stratification in multiple myeloma. Leukemia. 2014;28(2):269–77. doi: 10.1038/leu.2013.247.
  13. Rajkumar SV, Kumar S. Multiple Myeloma: diagnosis and treatment. Mayo Clin Proc. 2016;91(1):101–18. doi: 10.1016/j.mayocp.2015.11.007.
  14. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Staging System for multiple myeloma: a report from IMWG. J Clin Oncol. 2015;33(26):2863–6. doi: 10.1200/JCO.2015.61.2267.

Minimal Residual Disease and IGHV-Genes Mutational Status as the Main Predictors of Response to Bendamustine-Rituximab Therapy in Previously Untreated Chronic Lymphocytic Leukemia

YuV Mirolyubova, EA Stadnik, VV Strugov, TO Andreeva, TS Nikulina, YuV Virts, PA Butylin, AG Rumyantsev, AYu Zaritskey

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

For correspondence: Yuliya Vladimirovna Mirolyubova, 2 Akkuratova str., Saint Petersburg, Russian Federation, 197341; e-mail: juli9702@yandex.ru

For citation: Mirolyubova YuV, Stadnik EA, Strugov VV, et al. Minimal Residual Disease and IGHV-Genes Mutational Status as the Main Predictors of Response to Bendamustine-Rituximab Therapy in Previously Untreated Chronic Lymphocytic Leukemia. Clinical oncohematology. 2018;11(2):167–74.

DOI: 10.21320/2500-2139-2018-11-2-167-174


ABSTRACT

Background. In patients with chronic lymphocytic leukemia (CLL) the eradication of minimal residual disease (MRD) is a prognostic factor of overall survival (OS) and progression-free survival (PFS). IGHV mutational status has also independent prognostic value.

Aim. To analyse the impact of mutational status and MRD eradication in CLL patients after first-line standard BR (bendamustine + rituximab) immunochemotherapy.

Materials & Methods. The prospective study included patients with immunophenotypically confirmed CLL who had not previously received anticancer therapy. All patients were treated by BR combination from 2012 to 2015. MRD level was determined in 109 patients after completing the 3rd and the 6th treatment courses. IGHV mutational status data were available for 98 patients. IGHV mutational status was evaluated in accordance with ERIC recommendations. MRD was assessed by standardized method of 4-color flow cytometry.

Results. MRD negativity was achieved in 37 (34 %) out of 109 patients. MRD eradication correlated with the best PFS (= 0.04). IGHV mutational status had a statistically significant impact on PFS (= 0.02). In patients with MRD-negative response and IGHV mutation no unfavorable events occurred during the period of monitoring. Conversely, PFS rates in MRD-negative patients having no IGHV mutation and in MRD-positive patients with mutation were significantly worse. MRD eradication resulted in statistically significant improvement of PFS rates after completing 3 treatment courses, compared with the cases with MRD persistence regardless of residual malignant clone level (= 0.01).

Conclusion. BR therapy as first-line treatment statistically improved PFS in patients who achieved MRD-negative remission after completing the 3rd treatment course. PFS was significantly higher in MRD-negative patients with IGHV mutation after 6 treatment courses. MRD negativity resulting from 6 BR therapies in patients having no IGHV mutation was not accompanied by PFS improvement. It follows that by itself MRD negativity cannot be considered to be a universal prognostic factor.

Keywords: chronic lymphocytic leukemia, minimal residual disease, bendamustine, rituximab, BR, IGHV, mutational status.

Received: December 29, 2017

Accepted: February 27, 2018

Read in PDF 


REFERENCES

  1. Swerdlow SH, Campo E, Harris NL, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  2. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. 2016;127(20):2375–90. doi: 10.1182/blood-2016-01-643569.
  3. Клинические рекомендации по обследованию и лечению больных хроническим лимфолейкозом. Под ред. И.В. Поддубной, В.Г. Савченко [электронный документ]. Доступно по: https://blood.ru/documents/clinical%20guidelines/26.%20klinicheskie-rekomendacii-2014-xll.pdf. Ссылка активна на 12.2017.[Poddubnaya IV, Savchenko VG, eds. Clinical guidelines in examination and treatment of patients with chronic lymphocytic leukemia [Internet]. Available from: https://blood.ru/documents/clinical%20guidelines/26.%20klinicheskie-rekomendacii-2014-xll.pdf. (accessed 27.12.2017) (In Russ)]
  4. Keating MJ, O’Brien S, Albitar M, et al. Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol. 2005;23(18):4079–88. doi: 1200/JCO.2005.12.051.
  5. Eichhorst BF, Busch R, Hopfinger G, et al. Fludarabine plus cyclophosphamide versus fludarabine alone in first-line therapy of younger patients with chronic lymphocytic leukemia. 2006;107(3):885–91. doi: 10.1182/blood-2005-06-2395.
  6. Стадник Е.А., Никитин Е.А., Бидерман Б.В. и др. Ретроспективное сравнение эффективности и токсичности режимов лечения FC и FCR у первичных больных В-клеточным хроническим лимфолейкозом. Онкогематология. 2008;1–2:39–46.[Stadnik EA, Nikitin EA, Biderman BV, et al. Comparison of efficacy and toxicity of FC and FCR regimens in the treatment of primary B-cell chronic lymphocytic leukemia: a retrospective study. Onkogematologiya. 2008;1–2:39–46. (In Russ)]
  7. Vuillier F, Claisse JF, Vandenvelde C, et al. Evaluation of residual disease in B-cell chronic lymphocytic leukemia patients in clinical and bone-marrow remission using CD5-CD19 markers and PCR study of gene rearrangements. Leuk Lymphoma. 1992;7(3):195–204. doi: 10.3109/10428199209053623.
  8. Lenormand B, Bizet M, Fruchart C, et al. Residual disease in B-cell chronic lymphocytic leukemia patients and prognostic value. Leukemia. 1994;8(6):1019–26.
  9. Cabezudo E, Matutes E, Ramrattan M, et al. Analysis of residual disease in chronic lymphocytic leukemia by flow cytometry. Leukemia. 1997;11(11):1909–14. doi: 10.1038/sj.leu.2400835.
  10. Rawstron AC, Villamor N, Ritgen M, et al. International standardized approach for flow cytometric residual disease monitoring in chronic lymphocytic leukaemia. Leukemia. 2007;21(5):956–64. doi: 10.1038/sj.leu.2404584.
  11. Rawstron AC, Bottcher S, Letestu R, et al. Improving efficiency and sensitivity: European Research Initiative in CLL (ERIC) update on the international harmonised approach for flow cytometric residual disease monitoring in CLL. 2013;27(1):142–9. doi: 10.1038/leu.2012.216.
  12. Rawstron AC, Fazi C, Agathangelidis A, et al. A complementary role of multiparameter flow cytometry and high-throughput sequencing for minimal residual disease detection in chronic lymphocytic leukemia: an European Research Initiative on CLL study. Leukemia. 2016;30(4):929–36. doi: 10.1038/leu.2015.313.
  13. Луговская С.А., Почтарь М.Е., Наумова Е.В. Диагностика минимальной остаточной болезни при В-клеточном хроническом лимфолейкозе методом многопараметрической лазерной проточной цитофлюориметрии. Клиническая лабораторная диагностика. 2010;9:20–20а.[Lugovskaya SA, Pochtar’ ME, Naumova EV. Diagnosis of minimal residual diseases in B-cell chronic lympholeukemia by multiparametric laser flow cytofluorometry. Klinicheskaya laboratornaya diagnostika. 2010;9:20–20а. (In Russ)]
  14. Stehlikova O, Chovancova J, Tichy B, et al. Detecting minimal residual disease in patients with chronic lymphocytic leukemia using 8-color flow cytometry protocol in routine hematological practice. Int J Lab Hematol. 2014;36(2):165–71. doi: 10.1111/ijlh.12149.
  15. Bottcher S, Stilgenbauer S, Busch R, et al. Standardized MRD flow and ASO IGH RQ-PCR for MRD quantification in CLL patients after rituximab-containing immunochemotherapy: a comparative analysis. Leukemia. 2009;23(11):2007–17. doi: 10.1038/leu.2009.140.
  16. Thompson PA, Wierda WG. Eliminating minimal residual disease as a therapeutic end point: working toward cure for patients with CLL. Blood. 2016;127(3):279–86. doi: 10.1182/blood-2015-08-634816.
  17. Voena C, Ladetto M, Astolfi M, et al. A novel nested-PCR strategy for the detection of rearranged immunoglobulin heavy-chain genes in B cell tumors. Leukemia. 1997;11(10):1793–8. doi: 10.1038/sj.leu.2400801.
  18. Logan AC, Zhang B, Narasimhan B, et al. Minimal residual disease quantification using consensus primers and high-throughput IGH sequencing predicts post-transplant relapse in chronic lymphocytic leukemia. Leukemia. 2013;27(8):1659–65. doi: 10.1038/leu.2013.52.
  19. Pfitzner T, Engert A, Wittor H, et al. A real-time PCR assay for the quantification of residual malignant cells in B cell chronic lymphatic leukemia. Leukemia. 2000;14(4):754–66. doi: 10.1038/sj.leu.2401706.
  20. Hallek M, Cheson BD, Catovsky D, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: A report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood. 2008;111(12):5446–56. doi: 10.1182/blood-2007-06-093906.
  21. Guideline on the use of minimal residue disease as an endpoint in chronic lymphocytic leukaemia studies [Internet]. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2014/12/WC500179047.pdf. (accessed 27.12.2017).
  22. Bottcher S, Ritgen M, Fischer K, et al. Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: a multivariate analysis from the randomized GCLLSG CLL8 trial. J Clin Oncol. 2012;30(9):980–8. doi: 10.1200/JCO.2011.36.9348.
  23. Kovacs G, Robrecht S, Fink AM, et al. Minimal Residual Disease Assessment Improves Prediction of Outcome in Patients With Chronic Lymphocytic Leukemia (CLL) Who Achieve Partial Response: Comprehensive Analysis of Two Phase III Studies of the German CLL Study Group. J Clin Oncol. 2016;34(31):3758–65. doi: 10.1200/JCO.2016.67.1305.
  24. Damle RN, Wasil T, Fais F, et al. IgV gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood. 1999;94(6):1840–7.
  25. Thompson PA, Tam CS, O’Brien SM, et al. Fludarabine, cyclophosphamide, and rituximab treatment achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood. 2016;127(3):303–9. doi: 10.1182/blood-2015-09-667675.
  26. Kwok M, Rawstron A, Varghese A, et al. Minimal residual disease is an independent predictor for 10-year survival in CLL. Blood. 2016;128(24):2770–3. doi: 10.1182/blood-2016-05-714162.
  27. Strati P, Keating MJ, O’Brien SM, et al. Eradication of bone marrow minimal residual disease may prompt early treatment discontinuation in CLL. Blood. 2014;123(24):3727–32. doi: 10.1182/blood-2013-11-538116.
  28. Стругов В.В., Стадник Е.А., Румянцев А.М. и др. Влияние мутационного статуса IGHV-генов и стереотипности строения BCR на эффективность режима BR в первой линии терапии хронического лимфолейкоза. Клиническая онкогематология. 2017;10(2):141–9. doi: 10.21320/2500-2139-2017-10-2-141-149.[Strugov VV, Stadnik EA, Rumyantsev AM, et al. Effect of IGHV Gene Mutation Status and BCR Structure Stereotypy on Effectiveness of BR Regimen in First-Line Therapy of Chronic Lymphocytic Leukemia. Clinical oncohematology. 2017;10(2):141–9. doi: 10.21320/2500-2139-2017-10-2-141-149. (In Russ)]
  29. Никитин Е.А., Стадник Е.А., Лорие Ю.Ю. и др. Прогностическое значение мутационного статуса генов вариабельного региона иммуноглобулинов у больных хроническим лимфолейкозом, получавших комбинированную терапию флударабином и циклофосфаном. Терапевтический архив. 2007;79(7):66–70.[Nikitin EA, Stadnik EA, Lorie YuYu, et al. Prognostic significance of IGHV mutational status in chronic lymphocytic leukemia patients after combination therapy with fludarabine and cyclophosphan. Terapevticheskii arkhiv. 2007;79(7):66–70. (In Russ)]
  30. Cross NCP, White HE, Colomer D, et al. Laboratory recommendations for scoring deep molecular responses following treatment for chronic myeloid leukemia. Leukemia. 2015;29(5):999–1003. doi: 1038/leu.2015.29.
  31. Chanan-Khan A, Cramer P, Demirkan F, et al. Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (HELIOS): a randomised, double-blind, phase 3 study. Lancet Oncol. 2016;17(2):200–11. doi: 10.1016/S1470-2045(15)00465-9.
  32. Seymour JF, Ma S, Brander DM, et al. Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukaemia: a phase 1b study. Lancet Oncol. 2017;18(2):230–40. doi: 10.1016/s1470-2045(17)30012-8.
  33. Stilgenbauer S, Chyla B, Eichhorst B, et al. Venetoclax in relapsed/refractory chronic lymphocytic leukemia (CLL) with 17p deletion: outcome and minimal residual disease from the full population of the pivotal M13-982 trial. Eur Hematol Assoc. 2017: Abstract S771.
  34. Stilgenbauer S, Morschhauser F, Wendtner C-M, et al. Phase Ib study (GO28440) of venetoclax with bendamustine/rituximab or bendamustine/obinutuzumab in patients with relapsed/refractory or previously untreated chronic lymphocytic leukemia. Blood. 2016;128(22): Abstract 4393.

Epstein-Barr Virus in Patients with Classical Hodgkin’s Lymphoma

VE Gurtsevitch, EA Demina, NB Senyuta, IV Botezatu, KV Smirnova, TE Dushen’kina, DM Maksimovich, UV Paramonova, IS Monin, AV Lichtenshtein

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

For correspondence: Prof. Vladimir Eduardovich Gurtsevitch, MD, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: 8(499)324-25-64; e-mail: gurtsevitch-vlad-88@yandex.ru

For citation: Gurtsevitch VE, Demina EA, Senyuta NB, et al. Epstein-Barr Virus in Patients with Classical Hodgkin’s Lymphoma. Clinical oncohematology. 2018;11(2):160–6.

DOI: 10.21320/2500-2139-2018-11-2-160-166


ABSTRACT

Background. A close relationship between Epstein-Barr virus (EBV) and classical Hodgkin’s lymphoma (cHL) has been established in approximately 1/3 patients. EBV-positive lymphomas are characterized by increased level of EBV specific antibodies emerging long before tumor symptoms, аs well as a high plasma EBV DNA concentration. These viral markers normally correlate with clinical manifestations and the outcome of treatment performed. In patients with EBV-negative lymphomas, however, there has been no attempt to assess the clinical significance of either humoral response to EBV or EBV DNA concentration in plasma.

Aim. To evaluate diagnostic and prognostic significance of EBV markers in patients with EBV-negative lymphomas.

Methods. The clinical trial included 13 cHL-patients admitted at the Department of chemotherapy of hemoblastoses of NN Blokhin National Medical Cancer Research Center. The male to female ratio was 1:1.3, the median age was 26.4 years. Leukocyte and lymphocyte counts were evaluated in all the patients before, during, and after treatment as well as throughout the follow-up period. The same indicators were analysed in the control group which contained 40 healthy persons (with the median age of 41.1 years, male to female ratio 1.5:1). The study was based on serologic test for EBV antibodies and quantitative analysis of the viral DNA copy number in plasma.

Results. The obtained data show a low immunie response to EBV and its diminishment after several polychemotherapy treatment cycles, correlating with decreased leukocyte and lymphocyte levels. As opposed to levels of virus-specific antibodies which do not reflect the efficacy of anticancer therapy, plasma EBV DNA concentration in 2 patients decreased to 0 after remission had been achieved.

Conclusion. Although the number of observations is limited, one could suggest that viral load values in plasma of patients with EBV-negative lymphomas can prove to be a useful marker of anticancer therapeutic effect. Additional studies of these markers are required.

Keywords: Epstein-Barr virus (EBV), classical Hodgkin’s lymphoma, EBV DNA, EBV-negative classical Hodgkin’s lymphoma, level of virus-specific antibodies.

Received: November 13, 2017

Accepted: February 8, 2018

Read in PDF 


REFERENCES

  1. Alexander FE, Jarrett RF, Lawrence D, et al. Risk factors for Hodgkin’s disease by Epstein-Barr virus (EBV) status: prior infection by EBV and other agents. Br J Cancer. 2000;82(5):1117–21.
  2. Mueller N, Evans A, Harris NL, et al. Hodgkin’s disease and Epstein-Barr virus. Altered antibody pattern before diagnosis. N Engl J Med. 1989;320(11):689–95. doi: 10.1056/nejm198903163201103.
  3. Anagnostopoulos I, Herbst H, Niedobitek G, et al. Demonstration of monoclonal EBV genomes in Hodgkin’s disease and Ki-1-positive anaplastic large cell lymphoma by combined Southern blot and in situ hybridization. Blood. 1989;74(2):810–6.
  4. Tanyildiz HG, Yildiz I, Bassullu N, et al. The Role of Epstein-Barr Virus LMP-1 Immunohistochemical Staining in Childhood Hodgkin Lymphoma. Iran J Pediatr. 2015;25(6):e2359. doi: 10.5812/ijp.2359.
  5. Iwakiri D, Takada K. Role of EBERs in the pathogenesis of EBV infection. Adv Cancer Res. 2010;107:119–36. doi: 10.1016/s0065-230x(10)07004-1.
  6. Glaser SL, Lin RJ, Stewart SL, et al. Epstein-Barr virus-associated Hodgkin’s disease: epidemiologic characteristics in international data. Int J Cancer. 1997;70(4):375–82. doi: 10.1002/(sici)1097-0215(19970207)70:4<375::aid-ijc1>3.3.co;2-l.
  7. Jarrett AF, Armstrong AA, Alexander E. Epidemiology of EBV and Hodgkin’s lymphoma. Ann Oncol. 1996;7(Suppl 4):s5–s10. doi: 10.1093/annonc/7.suppl_4.s5.
  8. Ambinder RF. Gammaherpesviruses and “Hit-and-Run” oncogenesis. Am J Pathol. 2000;156(1):1–3. doi: 10.1016/s0002-9440(10)64697-4.
  9. Meij P, Vervoort MB, Bloemena E, et al. Antibody responses to Epstein-Barr virus-encoded latent membrane protein-1 (LMP1) and expression of LMP1 in juvenile Hodgkin’s disease. J Med Virol. 2002;68(3):370–7. doi: 10.1002/jmv.10213.
  10. Chang ET, Zheng T, Lennette ET, et al. Heterogeneity of risk factors and antibody profiles in Epstein-Barr virus genome-positive and -negative Hodgkin lymphoma. J Infect Dis. 2004;189(12):2271–81. doi: 10.1086/420886.
  11. Gallagher A, Perry J, Freeland J, et al. Hodgkin lymphoma and Epstein-Barr virus (EBV): no evidence to support hit-and-run mechanism in cases classified as non-EBV-associated. Int J Cancer. 2003;104(5):624–30. doi: 10.1002/ijc.10979.
  12. Staratschek-Jox A, Kotkowski S, Belge G, et al. Detection of Epstein-Barr virus in Hodgkin-Reed-Sternberg cells: no evidence for the persistence of integrated viral fragments in Latent membrane protein-1 (LMP-1)-negative classical Hodgkin’s disease. Am J Pathol. 2000;156(1):209–16. doi: 10.1016/s0002-9440(10)64721-9.
  13. zur Hausen H, de Villiers EM. Virus target cell conditioning model to explain some epidemiologic characteristics of childhood leukemias and lymphomas. Int J Cancer. 2005;115(1):1–5. doi: 10.1002/ijc.20905.
  14. Jelcic I, Hotz-Wagenblatt A, Hunziker A, et al. Isolation of multiple TT virus genotypes from spleen biopsy tissue from a Hodgkin’s disease patient: genome reorganization and diversity in the hypervariable region. J Virol. 2004;78(14):7498–507. doi: 10.1128/jvi.78.14.7498-7507.2004.
  15. Feng H, Shuda M, Chang Y, et al. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science. 2008;319(5866):1096–100. doi: 10.1126/science.1152586.
  16. Volter C, Hausen H, Alber D, et al. Screening human tumor samples with a broad-spectrum polymerase chain reaction method for the detection of polyomaviruses. Virology. 1997;237(2):389–96. doi: 10.1006/viro.1997.8772.
  17. Lo YM, Leung SF, Chan LY, et al. Kinetics of plasma Epstein-Barr virus DNA during radiation therapy for nasopharyngeal carcinoma. Cancer Res. 2000;60(9):2351–5.
  18. Wang WY, Twu CW, Chen HH, et al. Plasma EBV DNA clearance rate as a novel prognostic marker for metastatic/recurrent nasopharyngeal carcinoma. Clin Cancer Res. 2010;16(3):1016–24. doi: 10.1158/1078-0432.ccr-09-2796.
  19. Au WY. Quantification of circulating Epstein-Barr virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and EBV-positive lymphomas in immunocompetent patients. Blood. 2004;104(1):243–9. doi: 10.1182/blood-2003-12-4197.
  20. Hohaus S, Santangelo R, Giachelia M, et al. The viral load of Epstein-Barr virus (EBV) DNA in peripheral blood predicts for biological and clinical characteristics in Hodgkin lymphoma. Clin Cancer Res. 2011;17(9):2885–92. doi: 10.1158/1078-0432.ccr-10-3327.
  21. Kasamon YL, Jacene HA, Gocke CD, et al. Phase 2 study of rituximab-ABVD in classical Hodgkin lymphoma. Blood. 2012;119(18):4129–32. doi: 10.1182/blood-2012-01-402792.
  22. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013;121(18):3547–53. doi: 10.1182/blood-2012-09-454694.
  23. Dinand V, Sachdeva A, Datta S, et al. Plasma Epstein Barr Virus (EBV) DNA as a Biomarker for EBV associated Hodgkin lymphoma. Indian Pediatr. 2015;52(8):681–5. doi: 10.1007/s13312-015-0696-9.
  24. Stein H, Delsol G, Pileri SA, et al. Classical Hodgkin lymphoma, introduction. In: Swerdlow SH, Campo E, Harris NL, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  25. Lo YM, Chan LY, Chan AT, et al. Quantitative and temporal correlation between circulating cell-free Epstein-Barr virus DNA and tumor recurrence in nasopharyngeal carcinoma. Cancer Res. 1999;59(21):5452–5.
  26. Botezatu IV, Kondratova VN, Shelepov VP, et al. DNA melting analysis: application of the “open tube” format for detection of mutant KRAS. Anal Biochem. 2011;419(2):302–8. doi: 10.1016/j.ab.2011.08.015.
  27. Srinivas SK, Sample JT, Sixbey JW. Spontaneous loss of viral episomes accompanying Epstein-Barr virus reactivation in a Burkitt’s lymphoma cell line. J Infect Dis. 1998;177(6):1705–9. doi: 10.1086/517427.
  28. Razzouk BI, Srinivas S, Sample CE, et al. Epstein-Barr Virus DNA recombination and loss in sporadic Burkitt’s lymphoma. J Infect Dis. 1996;173(3):529–35. doi: 10.1093/infdis/173.3.529.

First-Line Treatment of Mantle-Cell Lymphoma: Analysis of Effectiveness and Cost-Effectiveness

KD Kaplanov1, NP Volkov1, TYu Klitochenko1, AL Shipaeva1, IV Matveeva1, MN Shirokova1, AC Proskurina1, NA Red’kina1, EG Gemdzhyan2

1 Volgograd Regional Clinical Oncologic Centre, 78 Zemlyachki str., Volgograd, Russian Federation, 400138

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

For correspondence: Kamil’ Daniyalovich Kaplanov, MD, PhD, 78 Zemlyachki str., Volgograd, Russian Federation, 400138; e-mail: kamilos@mail.ru

For citation: Kaplanov KD, Volkov NP, Klitochenko TYu, et al. First-Line Treatment of Mantle-Cell Lymphoma: Analysis of Effectiveness and Cost-Effectiveness. Clinical oncohematology. 2018;11(2):150–9.

DOI: 10.21320/2500-2139-2018-11-2-150-159


ABSTRACT

Aim. To study the correlation between efficacy of mantle-cell lymphoma treatment in clinical practice and failure of first-line therapy and direct expenses depending on the first-line therapy selection.

Methods. During the period from 2008 to 2016 a comparative single-center controlled trial was performed to evaluate the effectiveness and toxicity of R-hyper-CVAD-R-HD-AraC (n = 16) regimen. The control group included patients treated with 6–8 cycles of R-CHOP (n = 39). Cytarabine dose was lower than the original regimen and contained not more than 1 g/m2 twice a day for 2 days. R-hyper-CVAD regimen included the standard drug doses. R-HD-AraC treatment started on day 28 from the beginning of the R-hyper-CVAD therapy. The R-hyper-CVAD-R-HD-AraC group consisted of patients with the following characteristics: the median age was 56 years (range 40–66), older than 60 — 6 (38 %), male patients — 12 (75 %), stage IV — 12 (75 %), bulky — 7 (44 %), with bone marrow involved — 11 (69 %), MIPIb high-risk — 8 (50 %), blastoid variant — 7 (44 %). Only 2 patients of the R-hyper-CVAD-R-HD-AraC group received high-dose consolidation treatment with autologous HSC transplantation. HSCT was not performed in the control group. The results of comparative analysis were adjusted to age. In terms of the other significant factors the groups under comparison were similar.

Results. All the patients of the study group were treated with 3 R-hyper-CVAD and 3 R-HD-AraC regimens. The rate of complete remission was significantly higher than in the control group —12 (75 %) vs. 14 (36 %). No differences were observed in the 5-year overall survival: 55 % in the R-hyper-CVAD-R-HD-AraC group and 58 % in the R-CHOP group (= 0.75). Second-line therapy was received by 8 out of 15 (47 %) patients treated with R-hyper-CVAD-R-HD-AraC, and by 18 out of 23 (78 %) patients treated with R-CHOP. Median time before second-line therapy was significantly higher in the R-hyper-CVAD-R-HD-AraC group — 26 vs. 6 months (= 0.018). The costs of the first and subsequent therapy lines were analysed using a Markov model. Cost analysis of first-line therapy variants to be compared was based on cost-effectiveness ratio (CER) and incremental cost-effectiveness ratio (ICER). The analysis proved the cost-effectiveness of R-hyper-CVAD-R-HD-AraC program.

Conclusion. R-hyper-CVAD-R-HD-AraC program meets eligibility criteria for effectiveness, toxicity and cost-effectiveness and can, therefore, be recommended as first-line therapy of mantle-cell lymphoma and be used for the further comparative clinical trials.

Keywords: mantle-cell lymphoma, immunochemotherapy, pharmacoeconomics, Markov model, cost-effectiveness analysis.

Received: January 7, 2018

Accepted: March 3, 2018

Read in PDF 


REFERENCES

  1. A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin’s lymphoma. The Non-Hodgkin’s Lymphoma Classification Project. Blood. 1997;89(11):3909–18.
  2. Zelenetz AD, Gordon LI, Wierda WG, et al. Non-Hodgkin’s lymphomas, version 4.2014. J Natl Compr Canc Netw. 2014;12(9):1282–303.
  3. Aschebrook-Kilfoy B, Caces DB, Ollberding NJ, et al. An upward trend in the age-specific incidence patterns for mantle cell lymphoma in the USA. Leuk Lymphoma. 2013;54(8):1677–83. doi: 10.3109/10428194.2012.760041.
  4. Zhou Y, Wang H, Fang W, et al. Incidence Trends of Mantle Cell Lymphoma in the United States Between 1992 and 2004. Cancer. 2008;113(4):791–8. doi: 10.1002/cncr.23608.
  5. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375–90. doi: 10.1182/blood-2016-01-643569.
  6. Swerdlow SH, Campo E, Harris NL, et al. (eds) WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  7. Kelemen K, Peterson LC, Helenowski I, et al. CD23+ Mantle Cell Lymphoma. Am J Clin Pathol. 2008;130(2):166–77. doi: 10.1309/R94MAFJY5EA4A8C3.
  8. Vose JM. Mantle cell lymphoma: 2015 update on diagnosis, risk-stratification, and clinical management. Am J Hematol. 2015;90(8):739–45. doi: 10.1002/ajh.24094.
  9. Liu Z, Dong HY, Gorczyca W, et al. CD5– mantle cell lymphoma. Am J Clin Pathol. 2002;118(2):216–24. doi: 10.1309/TE56-A43X-29TT-5H8G.
  10. Zanetto U, Dong H, Huang Y, et al. Mantle cell lymphoma with aberrant expression of CD10. Histopathology. 2008;53(1):20–9. doi: 10.1111/j.1365-2559.2008.03060.x.
  11. Torlakovic E, Nielsen S, Vyberg M, et al. Antibody selection in immunohistochemical detection of cyclin D1 in mantle cell lymphoma. Am J Clin Pathol. 2005;124(5):782–9. doi: 10.1309/TYE7-K2CQ-MQ70-7FRT.
  12. Belaud-Rotureau MA, Parrens M, Dubus P, et al. A comparative analysis of FISH, RT-PCR, PCR, and immunohistochemistry for the diagnosis of mantle cell lymphomas. Mod Pathol. 2002;15(5):517–25. doi: 10.1038/modpathol.3880556.
  13. Vegliante MC, Palomero J, Perez-Galan P, et al. SOX11 regulates PAX5 expression and blocks terminal B-cell differentiation in aggressive mantle cell lymphoma. Blood. 2013;121(12):2175–85. doi: 10.1182/blood-2012-06-438937.
  14. Rosenwald A, Wright G, Wiestner A, et al. The proliferation gene expression signature is a quantitative integrator of oncogenic events that predicts survival in mantle cell lymphoma. Cancer Cell. 2003;3(2):185–97. doi: 10.1016/S1535-6108(03)00028-X.
  15. Chandran R, Gardiner SK, Simon M, Spurgeon SE. Survival trends in mantle cell lymphoma in the United States over 16 years 1992–2007. Leuk Lymphoma. 2012;53(8):1488–93. doi: 10.3109/10428194.2012.656628.
  16. Spurgeon SE, Till BG, Martin P, et al. Recommendations for clinical trial development in mantle cell lymphoma. J Natl Cancer Inst. 2017;109(1):1–10. doi: 10.1093/jnci/djw263.
  17. Sandoval-Sus JD, Sotomayor EM, Shah BD. Mantle cell lymphoma: Contemporary diagnostic and treatment perspectives in the age of personalized medicine. Hematol Oncol Stem Cell Ther. 2017;10(3):99–115. doi: 10.1016/j.hemonc.2017.02.003.
  18. LaCasce AS, Vandergrift JL, Rodriguez MA, et al. Comparative outcome of initial therapy for younger patients with mantle cell lymphoma: an analysis from the NCCN NHL Database. Blood. 2013;119(9):2093–9. doi: 10.1182/blood-2011-07-369629.
  19. Chen R, Li H, Bernstein SH, et al. Pre-Transplant R-Bendamustine Induces High Rates of Minimal Residual Disease in MCL Patients: Updated Results of S1106: US Intergroup Study of a Randomized Phase II Trial of R-HCVAD Vs. R-Bendamustine Followed By Autologous Stem Cell Transplants for Patients. Blood. 2015;126: Abstract 518.
  20. Khouri IF, Romaguera J, Kantarjian H, et al. Hyper-CVAD and high-dose methotrexate/cytarabine followed by stem-cell transplantation: An active regimen for aggressive mantle-cell lymphoma. J Clin Oncol. 1998;16(12):3803–9. doi: 10.1200/JCO.1998.16.12.3803.
  21. Romaguera JE, Fayad L, Rodriguez MA, et al. High rate of durable remissions after treatment of newly diagnosed aggressive mantle-cell lymphoma with rituximab plus hyper-CVAD alternating with rituximab plus high-dose methotrexate and cytarabine. J Clin Oncol. 2005;23(28):7013–23. doi: 10.1200/JCO.2005.01.1825.
  22. Damon LE, Johnson JL, Niedzwiecki D. Immunochemotherapy and autologous stem-cell transplantation for untreated patients with mantle-cell lymphoma: CALGB 59909. J Clin Oncol. 2009;27(36):6101–8. doi: 10.1200/JCO.2009.22.2554.
  23. Dreyling M, Lenz G, Hoster E, et al. Early consolidation by myeloablative radiochemotherapy followed by autologous stem cell transplantation in first remission significantly prolongs progression-free survival in mantle-cell lymphoma: results of a prospective randomized trial of the European MCL Network. Blood. 2005;105(7):2677–84. doi: 10.1182/blood-2004-10-3883.
  24. Hermine O, Hoster E, Walewski J, et al. Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): a randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. Lancet. 2016;388(10044):565–75. doi: 10.1016/S0140-6736(16)00739-X.
  25. Merli F, Luminari S, Ilariucci F, et al. Rituximab plus HyperCVAD alternating with high dose cytarabine and methotrexate for the initial treatment of patients with mantle cell lymphoma, a multicentre trial from Gruppo Italiano Studio Linfomi. Br J Haematol. 2012;156(3):346–53. doi: 10.1111/j.1365-2141.2011.08958.x.
  26. Geisler CH, Kolstad A, Laurell A, et al. Nordic MCL2 trial update: Six-year follow-up after intensive immunochemotherapy for untreated mantle cell lymphoma followed by BEAM or BEAC + autologous stem-cell support: Still very long survival but late relapses do occur. Br J Haematol. 2012;158(3):355–62. doi: 10.1111/j.1365-2141.2012.09174.x.
  27. Delarue R, Haioun C, Ribrag V, et al. CHOP and DHAP plus rituximab followed by autologous stem cell transplantation in mantle cell lymphoma: a phase 2 study from the Groupe d’Etude des Lymphomes de l’Adulte. Blood. 2012;121(1):48–53. doi: 10.1182/blood-2011-09-370320.
  28. Sachs JD. Macroeconomics and health: Investing in health for economic development. Revista Panamericana de Salud Publica. 2002;12(2):143–4. doi: 10.1590/s1020-49892002000800017.
  29. Laurell A, Kolstad A, Jerkeman M, et al. High dose cytarabine with rituximab is not enough in first-line treatment of mantle cell lymphoma with high proliferation: early closure of the Nordic Lymphoma Group Mantle Cell Lymphoma 5 trial. Leuk Lymphoma. 2014;55(5):1206–8. doi: 10.3109/10428194.2013.825906.
  30. Le Gouill S, Thieblemont C, Oberic L, et al. Rituximab maintenance after autologous stem-cell transplantation in patients with mantle cell lymphoma, final result of the LyMA trial conducted on behalf the LYSA group. Hematol Oncol. 2017;35:209. doi: 10.1002/hon.2438_74.
  31. Klener P, Fronkova E, Belada D, et al. Alternating R-CHOP and R-cytarabine is a safe and effective regimen for transplant-ineligible patients with a newly diagnosed mantle cell lymphoma. Hematol Oncol. 2017;1–6. doi: 10.1002/hon.2483
  32. Chen R, Li H, Bernstein SH, et al. Results of a randomized phase II trial of R-Hyper-CVAD versus bendamustine and rituximab followed by consolidation with ASCT in previously untreated patients with MCL. ICML 2015:062.
  33. van Keep M, Gairy K, Seshagiri D, et al. Cost-effectiveness analysis of bortezomib in combination with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone (VR-CAP) in patients with previously untreated mantle cell lymphoma. BMC Cancer. 2016;16(1):598. doi: 10.1186/s12885-016-2633-2.
  34. Widmer F, Balabanov S, Soldini D, et al. R-hyper-CVAD versus R-CHOP/cytarabine with high-dose therapy and autologous haematopoietic stem cell support in fit patients with mantle cell lymphoma: 20 years of single-center experience. Ann Hematol. 2017;97(2):277–87. doi: 10.1007/s00277-017-3180-x.
  35. Hoster E, Rosenwald A, Berger F, et al. Prognostic value of Ki-67 index, cytology, and growth pattern in mantle-cell lymphoma: Results from randomized trials of the European mantle cell lymphoma network. J Clin Oncol. 2016;34(12):1386–94. doi: 10.1200/JCO.2015.63.8387.

Conventional and Conformal Radiotherapy with and without Beam Intensity Modulation in Patients with Stage II Hodgkin’s Lymphoma with Supradiaphragmal Lesions

YuN Vinogradova1, EI Ivanova1, AI Chumachenko1, EV Smirnova2, GI Andreev2, AM Kalesnik2, NA Vorob’ev2, NV Il’in1

1 AM Granov Russian Research Centre of Radiology and Surgery Technologies, 70 Leningradskaya str., Pesochnyi, Saint Petersburg, Russian Federation, 197758

2 Nuclear Medicine Centre of SM Berezin International Institute of Biological Systems, 43 Karl Marx str., Pesochnyi, Saint Petersburg, Russian Federation, 197758

For correspondence: Nikolai Vasil’evich Il’in, PhD, Professor, 70 Leningradskaya str., Pesochnyi, Saint Petersburg, Russian Federation, 197758; Tel.: +7(812)596-90-35; e-mail: ilyin_prof@mail.ru.

For citation: Vinogradova YuN, Ivanova EI, Chumachenko AI, et al. Conventional and Conformal Radiotherapy with and without Beam Intensity Modulation in Patients with Stage II Hodgkin’s Lymphoma with Supradiaphragmal Lesions. Clinical oncohematology. 2018;11(1):70-7.

DOI: 10.21320/2500-2139-2018-11-1-70-77


ABSTRACT

Aim. To increase the efficacy of chemo-radiotherapy in patients with stage II Hodgkin’s lymphoma (HL) with supradiaphragmal lesions by different fractionation (conventional fractionation [CF] and multi-fractionation [MF]) and various radiation volume (mantle radiotherapy, involved field radiotherapy [IFRT] or involved site radiotherapy [ISRT]).

Materials & Methods. The clinical trial included 317 patients with stage II classical HL with supradiaphragmal lesions who have received chemo-radiotherapy in AM Granov Russian Scientific Centre of Radiology and Surgery Technologies from 1986 to 2015 (n = 301) and in SM Berezin International Institute of Biological Systems from 2014 to 2016 (n = 16). The mean age was 30.9 years (range 18–65); the study group included 107 men and 210 women. The diagnosis in all the cases was confirmed by immunomorphologic analysis. The treatment program included 2 to 6 cycles of ABVD regimen followed by CF (n = 153) or MF (n = 148) radiation therapy. The patients received mantle radiotherapy or IFRT with the cumulative dose of 30 to 40 Gy (2D planning, 237 patients) or ISRT with the cumulative dose of 30 to 40 Gy (3D planning, n = 80, CF only). The MF-regimen was administered only with 2D planning, mantle radiotherapy and IFRT. The CF-regimen was administered with 2D-planning (n = 89) with the same volume of radiation and 3D planning using conformal RT (3D-CRT) as ISRT with the cumulative dose of 30 to 36 Gy (n = 80). From the total of 80 patients 16 patients were treated with beam intensity modulation (IMRT) and 64 patients were treated without IMRT.

Results. The treatment response (both complete and partial remission) was reported in 235 (99.2 %) of 237 patients of 2D planning group. In 2 cases (0.8 %) a progression of disease was diagnosed. Out of 235 patients with remission 222 (94.5 %) had a complete remission and 13 (5.5 %) had a partial remission. Within the group treated with the conventional RT 19 (8.1 %) patients had recurrent disease. Overall 5- and 10-year survival (OS) was equal and accounted for 98.0 ± 1.4 %, the disease-free survival (DFS) was also equal between groups and accounted for 85.9 ± 1.3 %. The rate of 5- and 10-year OS in patients who received CF was 97.8 ± 1.7 %; the DFS rate for the same period was 85.0 ± 1.5 % with standard fractionation and 86.2 ± 1.6 % with MF (> 0.1). The total number of local radiation reactions and the number of radiation pulmonitis were significantly smaller with the exposure twice daily compared to CF at 2D planning and mantle irradiation. Decreasing the radiation volume from the mantle type to IFRT was shown to reduce the incidence of pulmonitis. The incidence of esophagitis remained the same with different fractionation and the irradiation volumes specified above. The results of the analysis proved the total absence of radiation pericarditis with both types of conformal RT with or without IMRT. No pulmonitis cases were observed with IMRT; the use of 3D-CRT and 2D-RT significantly increased the incidence of pulmonitis. The incidence of esophagitis within the CRT-group (n = 80) was 2 times lower compared to the conventional RT (22.5 % and 43.9 %; < 0.01).

Conclusion. The innovative technologies of radiation therapy allowed to reduce the incidence of early local radiation reactions. These technologies will be the basis for preventing severe late radiation complications that reduce the life expectancy and quality of life of patients with HL.

Keywords: Hodgkin’s lymphoma, radiotherapy, fractionation, 2D and 3D planning.

Received: September 16, 2017

Accepted: December 5, 2017

Read in PDF 


REFERENCES

  1. Yahalom J, Hoppe RT. Principles of Radiation Techniques in Hodgkin Lymphoma. In: Engert A, Horning SJ, eds. Hodgkin Lymphoma. A Comprehensive Update on Diagnostics and Clinics. Berlin, Heidelberg: Springer; 2011. pp. 117–39. doi: 10.1007/978-3-642-12780-9.
  2. Даниленко А.А., Шахтарина С.В. Лучевая терапия лимфомы Ходжкина: от «радикальной программы» лечения до современных технологий. Медицинская радиология и радиационная безопасность. 2015;60(4):71–9. [Danilenko AA, Shakhtarina Radiation therapy of Hodgkin’s lymphoma: from the “radical radiation therapy” to the actual technology. Meditsinskaya radiologiya i radiatsionnaya bezopasnost’. 2015;60(4):71–9. (In Russ)]
  3. Lugtenburg E, Hagenbeek A. Treatment of Early Favorable Hodgkin Lymphoma. In: Engert A, Horning SJ, eds. Hodgkin Lymphoma. A Comprehensive Update on Diagnostics and Clinics. Berlin, Heidelberg: Springer; 2011. pp. 163–81. doi: 10.1007/978-3-642-12780-9.
  4. Hoppe R. Radiotherapy Planning for the Lymphomas: Expanding Roles for Biologic Imaging. In: Meyer JL, ed. IMRT, IGRT, SBRT: Advances in the Treatment Planning and Delivery of Radiotherapy, 2nd, revised and extended edition. Basel: Karger; 2011. 487 p.
  5. Pavlovsky S, Maschio M, Santarelli MT, et al. Randomized trial of chemotherapy versus chemotherapy plus radiotherapy for stage I–II Hodgkin’s disease. J Natl Cancer Inst. 1988;80(18):1466–73. doi: 10.1093/jnci/80.18.1466.
  6. Laskar S, Gupta T, Vimal S, et al. Consolidation radiation after complete remission in Hodgkin’s disease following six cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine chemotherapy: is there a need? J Clin Oncol. 2004;22(1):62–8. doi: 10.1200/jco.2004.01.021.
  7. Straus DJ, Portlock CS, Qin J, et al. Results of a prospective randomized clinical trial of doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) followed by radiation therapy (RT) versus ABVD alone for stages III, and IIIA non bulky Hodgkin disease. Blood. 2004;104(12):3483–9. doi: 10.1182/blood-2004-04-1311.
  8. Meyer RM, Gospodarowicz MK, Connors JM, et al. Randomized comparison of ABVD chemotherapy with a strategy that includes radiation therapy in patients with limited-stage Hodgkin’s lymphoma: National Cancer Institute of Canada Clinical Trial Group and the Eastern Cooperative Oncology Group. J Clin Oncol. 2005;23(21):4634–42. doi: 10.1200/JCO.2005.09.085.
  9. Engert A, Shiller P, Josting A, et al. Involved-field radiotherapy is equally effective and less toxic compared with extended-field radiotherapy after four cycles of chemotherapy in patients with early-stage unfavorable Hodgkin’s lymphoma: results of the HD8 trial of the German Hodgkin’s Study Group. J Clin Oncol. 2003;21(19):3601–8. doi: 10.1200/jco.2003.03.023.
  10. Bonadonna G, Bonfante V, Viviani S, et al. ABVD plus subtotal nodal versus involved-field radiotherapy in early-stage Hodgkin’s disease: long-term results. J Clin Oncol. 2004;22(14):2835–41. doi: 10.1200/jco.2004.12.170.
  11. Ferme C, Eghbali H, Meerwaldt JH, et al. Chemotherapy plus involved-field radiation in early-stage Hodgkin’s disease. N Engl J Med. 2007;357(19):1916–27. doi: 10.1056/NEJMoa064601.
  12. Von Treskow B, Gorgen H, Plutschow A, et al. Infradiaphragmatic Hodgkin lymphoma in patients treated with state-of-the-art therapies: a risk factors analysis from the German Hodgkin Study Group (GHSG) HD13 and HD14 trials. Haematologica. 2013;98(2):12–3. (abstr. P036).
  13. Акимов А.А., Ильин Н.В. Роль фракционирования дозы в повышении эффективности лучевой терапии опухолей. СПб.: Берсерк, 2005. 29 с.[Akimov AA, Il’in NV. Rol’ fraktsionirovaniya dozy v povyshenii effektivnosti luchevoi terapii opukholei. (Significance of dose fractionation in increasing of efficacy of tumor radiation therapy.) Saint Petersburg: Berserk Publ.; 2005. 29 p. (In Russ)]
  14. Yahalom J. Principles, Indications, and Techniques of Radiation Therapy of Lymphomas. In: Canellos G, Lister TA, Young B, eds. The Lymphomas, 2nd edition. Elsevier; 2006. pp. 203–24. doi: 10.1016/b978-0-7216-0081-9.50016-9.
  15. Крутилина Н.И. Эффективность нетрадиционной лучевой терапии больных лимфогранулематозом IIA-IIIA стадий. Медицинская радиология. 1990;9:44. [Krutilina NI. Efficacy of non-traditional radiotherapy of stages IIA/IIIA Hodgkin’s lymphoma. Meditsinskaya radiologiya. 1990;9:44. (In Russ)]
  16. Ильин Н.В. Оптимизация методов лучевой терапии лимфогранулематоза: Автореф. дис. … д-ра мед. наук. СПб., 1993. 42 с.[Il’in NV. Optimizatsiya metodov luchevoi terapii limfogranulematoza. (Optimization of methods of radiation treatment of Hodgkin’s disease.) [dissertation] Saint Petersburg; 1993. 42 p. (In Russ)]
  17. Акимов А.А. Анализ дозно-временных зависимостей при лучевой терапии злокачественных опухолей: Автореф. дис. … д-ра мед. наук. СПб., 2000. 48 с. [Akimov AA. Analiz dozno-vremennykh zavisimostei pri luchevoi terapii zlokachestvennykh opukholei. (Analysis of dose and time correlations in radiation treatment of malignant tumors.) [dissertation] Saint Petersburg; 2000. 48 p. (In Russ)]
  18. Ильин Н.В., Виноградова Ю.Н., Николаева Е.Н., Смирнова Е.В. Значение мультифракционирования дозы радиации при первичном лучевом лечении больных лимфомой Ходжкина. Онкогематология. 2007;4:47–53. [Il’in NV, Vinogradova YuN, Nikolaeva EN, Smirnova EV. Value of accelerated hyperfraction doses of radiation in primary radiation treatment for Hodgkin’s lymphoma. Onkogematologiya. 2007;4:47–53. (In Russ)]
  19. Изотов Б.М. Исходные прогностические факторы и оптимизация терапии больных лимфогранулематозом IIIA стадии: Автореф. дис. … канд. мед. наук. СПб., 2000. 24 с. [Izotov BM. Iskhodnye prognosticheskie faktory i optimizatsiya terapii bol’nykh limfogranulematozom IIIA stadii. (Background prognostic factors and optimization of treatment of stage IIIA Hodgkin’s lymphoma.) [dissertation] Saint Petersburg; 2000. 24 p. (In Russ)]
  20. Николаева Е.Н. Оценка эффективности режима ускоренного мультифракционирования дозы в лучевом и комбинированном лечении больных лимфомой Ходжкина II–III стадий: Автореф. дис. … канд. мед. наук. СПб., 2005. 23 с. [Nikolaeva EN. Otsenka effektivnosti rezhima uskorennogo mul’tifraktsionirovaniya dozy v luchevom i kombinirovannom lechenii bol’nykh limfomoi Khodzhkina II–III stadii. (Efficacy evaluation of the accelerated dose multifranctionation in radiation and combined treatment of stages II/III Hodgkin’s lymphoma.) [dissertation] Saint Petersburg; 2005. 23 p. (In Russ)]
  21. Specht L, Yahalom J, Illidge T, et al. Modern Radiation Therapy for Hodgkin’s lymphoma: Field and Dose Guidelines from the International Lymphoma Radiation Oncology Group (ILROG). Int J Rad Oncol Biol Phys. 2014;89(4):854–62. doi: 10.1016/j.ijrobp.2013.05.005.
  22. Carbone PP, Kaplan HS, Musshoff K, et al. Report of the Committee on Hodgkin’s disease staging classification. Cancer Res. 1971;31:1860–1.
  1. Lister TA, Crowther D, Sutcliffe SB, et al. Report of a committee convened to discuss the evaluation and staging of patients with Hodgkin’s disease: Cotswolds meeting. J Clin Oncol. 1989;7(11):1630–6. doi: 10.1200/jco.1989.7.11.1630.
  2. Симбирцева Л.П., Синицын Р.В., Виноградов В.М., Ватницкий С.М. Планирование и проведение лучевой терапии больных лимфогранулематозом по радикальной программе с помощью крупных полей сложных конфигураций: Методические рекомендации. Ленинград, 1979. 27 с. [Simbirtseva LP, Sinitsyn RV, Vinogradov VM, Vatnitskii SM. Planirovanie i provedenie luchevoi terapii bol’nykh limfogranulematozom po radikal’noi programme s pomoshch’yu krupnykh polei slozhnykh konfiguratsii. Metodicheskie rekomendatsii. (Radiotherapy planning and realization in patients with Hodgkin’s disease according to the radical program using large fields with complicated configuration. Methodological guidelines.) Leningrad; 1979. 27 p. (In Russ)]
  3. Kaplan H. The radical radiotherapy of regionally localized Hodgkin’s disease. Radiology. 1962;78(4):553–61. doi: 10.1148/78.4.553.
  4. Yahalom J, Mauch P. The involved field is back: issues in delineating the radiation field in Hodgkin’s disease. Ann Oncol. 2002;13(Suppl 1):79–83. doi: 10.1093/annonc/13.s1.79.
  5. Girinsky T, van der Maazen, Specht L, et al. Involved-node radiotherapy (INRT) in patients with early Hodgkin lymphoma: concepts and guidelines. Radiother Oncol. 2006;79(3):270–7. doi: 10.1016/j.radonc.2006.05.015.
  6. World health statistics annual. Volume II. Infectious diseases: Cases and deaths 228 World Health Organization Geneva 1978 Sw. fr. 32 EW. Public Health. 1979;93(3):199–200. doi: 10.1016/s0033-3506(79)80127-4.
  7. Cox J, Stetz J, Pajak T. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31(5):1341–6. doi: 10.1016/0360-3016(95)00060-c.
  8. Cheson BD, Horning SJ, Coiffer B, et al. Report of an international workshop to standardize response criteria for non-Hodgkin’s lymphomas. J Clin Oncol. 1999:17(4):1244–53. doi: 10.1200/jco.1999.17.4.1244.
  9. Cheson BD, Pfisther B, Juweid ME, et al. Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579–86. doi: 10.1200/jco.2006.09.2403.
  10. Волчков В.А. Краткое практическое руководство по биометрии для врачей, 2-е издание. СПб., 2004. 60 с. [Volchkov VA. Kratkoe prakticheskoe rukovodstvo po biometrii dlya vrachei. (Quick practical guide on biometry for medical doctors.) 2nd edition. Saint Petersburg; 2004. 60 p. (In Russ)]
  11. Macdonald DA, DingK, Gospodarowicz MK, et al. Patterns of disease progression and outcomes in a randomized trial testing ABVD alone for patients with limited-stage Hodgkin lymphoma. Ann Oncol. 2007;18(10):1680–4. doi: 10.1093/annonc/mdm
  12. Количественный анализ повреждений здоровых органов и тканей при проведении лучевой терапии злокачественных новообразований (Проект QUANTEC). Обзор толерантности нормальных тканей: Пер. с англ. под ред. С.И. Ткачева. М., 2015. 250 c. [Quantitative analysis of normal organ and tissue injuries in radiotherapy of malignant neoplasms (QUANTEC project). Quantitative Analyses of Normal Tissue Effects in the Clinic. (Russ. ed.: Tkachev SI. Kolichestvennyi analiz povrezhdenii zdorovykh organov i tkanei pri provedenii luchevoi terapii zlokachestvennykh novoobrazovanii (Proekt QUANTEC). Obzor tolerantnosti normal’nykh tkanei. Moscow; 2015. 250 p.)]
  13. Terezakis SA, Hunt M, Specht L, Yahalom J. Traditional and Modern Techniques for Radiation Treatment Planning. In: Specht L, Yahalom J, eds. Radiotherapy for Hodgkin Lymphoma. Berlin, Heidelberg: Springer; 2011. pp. 123–51. doi: 10.1007/978-3-540-78944-4_10.