Mantle Cell Lymphoma: History, Current Principles of Diagnosis, and Treatment (Literature Review)

GS Tumyan

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

For correspondence: Gayane Sepugovna Tumyan, MD, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: gaytum@mail.ru

For citation: Tumyan GS. Mantle Cell Lymphoma: History, Current Principles of Diagnosis, and Treatment (Literature Review). Clinical oncohematology. 2020;13(4):366–81. (In Russ).

DOI: 10.21320/2500-2139-2020-13-4-366-381


ABSTRACT

Mantle cell lymphoma (MCL) is a heterogeneous disease with a broad spectrum of clinical manifestations from rare indolent cases requiring no immediate treatment to aggressive fast-proliferating tumors. Differences in clinical behavior are rooted in molecular grounds which in the latest edition of WHO hematopoietic and lymphoid tissue tumor classification formed the basis for dividing MCL into two variants: classical (in most cases) and indolent. In last decades, our insight into biology and disease development mechanisms has been considerably enhanced. Further, it will help to risk stratify patients not only according to clinical factors (MIPI) but also taking into account molecular and biological properties of tumor (Ki-67 proliferation index, ТР53, NOTCH1, and NOTCH2 mutations, complex karyotype, and unmutated IGHV status). Treatment algorithms based on intensive chemotherapy with high-dose cytarabine and autologous hematopoietic stem cell transplantation with further rituximab maintenance therapy ensure long-term monitoring of the disease in many MCL patients. The use of new “chemo-free” regimens and rational combinations (bortezomib, BTK inhibitors, lenalidomide, and venetoclax) offers the hope of a departure from conventional chemotherapy for a certain part of patients. Novel drugs with unique modes of action enabled, to some extent, to deconstruct the stigma of MCL fatality.

Keywords: mantle cell lymphoma, treatment.

Received: June 17, 2020

Accepted: September 2, 2020

Read in PDF


REFERENCES

  1. Rappaport H, Winter WJ, Hicks EB. Follicular lymphoma. A re-evaluation of its position in the scheme of malignant lymphoma, based on a survey of 253 cases. 1956;9(4):792–821. doi: 10.1002/1097-0142(195607/08)9:4<792::aid-cncr2820090429>3.0.co;2-b.

  2. Gerard-Marchant R, Hamlin I, Lennert K, et al. Classification of non-Hodgkin’s lymphomas. Lancet. 1974;2:406–8.

  3. Tubbs RR, Fishleder A, Weiss RA, et al. Immunohistologic cellular phenotypes of lymphoproliferative disorders. Comprehensive evaluation of 564 cases including 257 non-Hodgkin’s lymphomas classified by the International Working Formulation. Am J Pathol. 1983;113(2):207–21.

  4. Banks PM, Chan J, Cleary ML, et al. Mantle cell lymphoma: a proposal for unification of morphologic, immunologic, and molecular data. Am J Surg Pathol. 1992;16(7):637–40. doi: 10.1097/00000478-199207000-00001.

  5. Harris NL, Jaffe ES, Stein H, Banks PM. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood. 1994;84(5):1361–92. doi: 10.1182/blood.v84.5.1361.bloodjournal8451361.

  6. Dreyling M, Klapper W, Rule S. Blastoid and pleomorphic mantle cell lymphoma: still a diagnostic and therapeutic challenge! Blood. 2018;132(26):2722–9. doi: 10.1182/blood-2017-08-737502.

  7. Hernandez L, Fest T, Cazorla M, et al. p53 gene mutations and protein overexpression are associated with aggressive variants of mantle cell lymphomas. Blood. 1996;87(8):3351–9. doi: 10.1182/blood.v87.8.3351.bloodjournal8783351.

  8. Stefancikova L, Moulis M, Fabian P, et al. Loss of the p53 tumor suppressor activity is associated with negative prognosis of mantle cell lymphoma. Int J Oncol. 2010;36(3):699–706. doi: 10.3892/ijo_00000545.

  9. Aukema SM, Hoster E, Rosenwald A, et al. Expression of TP53 is associated with the outcome of MCL independent of MIPI and Ki-67 in trials of the European MCL Network. Blood. 2018;131(4):417–20. doi: 10.1182/blood-2017-07-797019.

  10. 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.

  11. Jiang W, Kahn SM, Zhou P, et al. Overexpression of Cyclin D1 in Rat Fibroblasts Causes Abnormalities in Growth Control, Cell Cycle Progression and Gene Expression. Oncogene. 1993;8(12):3447–57.

  12. Jaffe ES, Harris NL, Stein H, Isaacson PG. Classification of lymphoid neoplasms: the microscope as a tool for disease discovery. Blood. 2008;112(12):4384–99. doi: 10.1182/blood-2008-07-077982.

  13. Klener P. Advances in Molecular Biology and Targeted Therapy of Mantle Cell Lymphoma. Int J Mol Sci. 2019;20(18):4417. doi: 10.3390/ijms20184417.

  14. Jares P, Colomer D, Campo E. Genetic and molecular pathogenesis of mantle cell lymphoma: Perspectives for new targeted therapeutics. Nat Rev Cancer. 2007;7(10):750–62. doi: 10.1038/nrc2230.

  15. Vincent-Fabert C, Fiancette R, Rouaud P, et al. A defect of the INK4-Cdk4 checkpoint and Myc collaborate in blastoid mantle cell lymphoma-like lymphoma formation in mice. Am J Pathol. 2012;180(4):1688–701. doi: 10.1016/j.ajpath.2012.01.004.

  16. Choe JY, Yun JY, Na HY, et al. MYC overexpression correlates with MYC amplification or translocation, and is associated with poor prognosis in mantle cell lymphoma. Histopathology. 2016;68(3):442–9. doi: 10.1111/his.12760.

  17. Salaverria I, Royo C, Carvajal-Cuenca A, et al. CCND2 rearrangements are the most frequent genetic events in cyclin D1– mantle cell lymphoma. Blood. 2013;121(8):1394–402. doi: 10.1182/blood-2012-08-452284.

  18. Bea S, Valdes-Mas R, Navarro A, et al. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci USA. 2013;110(45):18250–5. doi: 10.1073/pnas.1314608110.

  19. 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.

  20. Kuo PY, Jatiani SS, Rahman AH, et al. SOX11 augments BCR signaling to drive MCL-like tumor development. Blood. 2018;131(20):2247–55. doi: 10.1182/blood-2018-02-832535.

  21. Balsas P, Palomero J, Eguileor A, et al. SOX11 promotes tumor protective microenvironment interactions through CXCR4 and FAK regulation in mantle cell lymphoma. Blood. 2017;130(4):501–13. doi: 10.1182/blood-2017-04-776740.

  22. Narurkar R, Alkayem M, Liu D. SOX11 is a biomarker for cyclin D1-negative mantle cell lymphoma. Biomark Res. 2016;4(1):6. doi: 10.1186/s40364-016-0060-9.

  23. Zhang J, Jima D, Moffitt AB, et al. The genomic landscape of mantle cell lymphoma is related to the epigenetically determined chromatin state of normal B cells. Blood. 2014;123(19):2988–96. doi: 10.1182/blood-2013-07-517177.

  24. Yang P, Zhang W, Wang J, et al. Genomic landscape and prognostic analysis of mantle cell lymphoma. Cancer Gene Ther. 2018;25(5–6):129–40. doi: 10.1038/s41417-018-0022-5.

  25. Eskelund CW, Dahl C, Hansen JW, et al. TP53 Mutations Identify Younger Mantle Cell Lymphoma Patients Who Do Not Benefit From Intensive Chemoimmunotherapy. Blood. 2017;130(17):1903–10. doi: 10.1182/blood-2017-04-779736.

  26. Morello L, Rattotti S, Giordano L, et al. Mantle Cell Lymphoma of Mucosa-Associated Lymphoid Tissue. A European Mantle Cell Lymphoma Network Study. Hemasphere. 2019;4(1):e302. doi: 10.1097/hs9.0000000000000302.

  27. Hoster E, Dreyling M, Klapper W, et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood. 2008;111(2):558–65. doi: 10.1182/blood-2007-06-095331.

  28. Scott DW, Abrisqueta P, Wright GW, et al. New Molecular Assay for the Proliferation Signature in Mantle Cell Lymphoma Applicable to Formalin-Fixed Paraffin-Embedded Biopsies. J Clin Oncol. 2017;35(15):1668–77. doi: 10.1200/jco.2016.70.7901.

  29. Kridel R, Meissner B, Rogic S, et al. Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma. Blood. 2012;119(9):1963–71. doi: 10.1182/blood-2011-11-391474.

  30. Halldorsdottir AM, Lundin A, Murray F, et al. Impact of TP53 mutation and 17p deletion in mantle cell lymphoma. Leukemia. 2011;25(12):1904–8. doi: 10.1038/leu.2011.162.

  31. Delfau-Larue MH, Klapper W, Berger F, et al. European Mantle Cell Lymphoma Network. High-dose cytarabine does not overcome the adverse prognostic value of CDKN2A and TP53 deletions in mantle cell lymphoma. Blood. 2015;126(5):604–11. doi: 10.1182/blood-2015-02-628792.

  32. Cohen JB. TP53 mutations in MCL: more therapy is not better. Blood. 2017;130(17):1876–7. doi: 10.1182/blood-2017-08-803551.

  33. Jain P, Wang M. Mantle cell lymphoma: 2019 update on the diagnosis, pathogenesis, prognostication, and management. Am J Hematol. 2019;94(6):710–25. doi: 10.1002/ajh.25487.

  34. Chihara D, Cheah CY, Westin JR, et al. Rituximab plus hyper-CVAD alternating with MTX/Ara-C in patients with newly diagnosed mantle cell lymphoma: 15-year follow-up of a phase II study from the MD Anderson Cancer Center. Br J Haematol. 2016;172(1):80–8. doi: 10.1111/bjh.13796.

  35. Romaguera JE, Wang M, Feng L, et al. Phase 2 trial of bortezomib in combination with rituximab plus hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with bortezomib, rituximab, methotrexate, and cytarabine for untreated mantle cell lymphoma. Cancer. 2018;124(12):2561–9. doi: 10.1002/cncr.31361.

  36. 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.

  37. Bernstein SH, Epner E, Unger JM, et al. A phase II multicenter trial of hyperCVAD MTX/Ara-C and rituximab in patients with previously untreated mantle cell lymphoma; SWOG 0213. Ann Oncol. 2013;24(6):1587–93. doi: 10.1093/annonc/mdt070.

  38. Eskelund CW, Kolstad A, Jerkeman M, et al. 15-year follow-up of the second Nordic mantle cell lymphoma trial (MCL2): prolonged remissions without survival plateau. Br J Haematol. 2016;175(3):410–8. doi: 10.1111/bjh.14241.

  39. 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.

  40. Armand P, Redd R, Bsat J, et al. A phase 2 study of Rituximab-Bendamustine and Rituximab-Cytarabine for transplant-eligible patients with mantle cell lymphoma. Br J Haematol. 2016;173(1):89–95. doi: 10.1111/bjh.13929.

  41. Le Gouill S, Thieblemont C, Oberic L, et al. Rituximab after autologous stem-cell transplantation in mantle-cell lymphoma. N Engl J Med. 2017;377(13):1250–60. doi: 10.1056/NEJMoa1701769.

  42. Kluin-Nelemans HC, Hoster E, Hermine O, et al. Treatment of Older Patients With Mantle Cell Lymphoma (MCL): Long-Term Follow-Up of the Randomized European MCL Elderly Trial. J Clin Oncol. 2020;38(3):248–56. doi: 10.1200/JCO.19.01294.

  43. 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.

  44. Flinn IW, van der Jagt R, Kahl BS, et al. Randomized trial of bendamustine-rituximab or RCHOP/RVP in first line treatment of indolent NHL or MCL: the BRIGHT study. Blood. 2014;123(19):2944–52. doi: 10.1182/blood-2013-11-531327.

  45. Kluin-Nelemans JC, Hoster E, Hermine O, et al. Treatment of older patients with mantle cell lymphoma. N Engl J Med. 2012;367(6):520–31. doi: 10.1056/NEJMoa1200920.

  46. Robak T, Huang H, Jin J, et al. Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N Engl J Med. 2015;372(10):944–53. doi: 10.1056/NEJMoa1412096.

  47. Robak T, Jin J, Pylypenko H, et al. Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: final overall survival results of a randomised, open-label, phase 3 study. Lancet Oncol. 2018;19(11):1449–58. doi: 10.1016/S1470-2045(18)30685-5.

  48. Ratnasingam S, Casan J, Shortt J, et al. Cytarabine-based induction immunochemotherapy in the front-line treatment of older patients with mantle cell lymphoma. Sci Rep. 2019;9(1):13544. doi: 10.1038/s41598-019-49776-9.

  49. Visco C, Finotto S, Zambello R, et al. Combination of Rituximab, Bendamustine, and Cytarabine for Patients With Mantle-Cell Non-Hodgkin Lymphoma Ineligible for Intensive Regimens or Autologous Transplantation. J Clin Oncol. 2013;31(11):1442–9. doi: 10.1200/JCO.2012.45.9842.

  50. 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.

  51. Ruan J, Martin P, Christos P, et al. Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma. Blood. 2018;132(19):2016–25. doi: 10.1182/blood-2018-07-859769.

  52. Hoster E, Pott C. Minimal residual disease in mantle cell lymphoma: insights into biology and impact on treatment. Hematology. 2016;2016(1):437–45. doi: 10.1182/asheducation-2016.1.437.

  53. Pott C, Bruggemann M, Ritgen M, et al. MRD detection in B-cell non-Hodgkin lymphomas using Ig gene rearrangements and chromosomal translocations as targets for real-time quantitative PCR. In: R Kuppers (ed). Lymphoma. Methods in Molecular Biology (Methods and Protocols). Vol. 971. Totowa: Humana Press; 2013. рр. 175–200. doi: 10.1007/978-1-62703-269-8_10.

  54. Kumar A, Sha F, Toure A, et al. Patterns of survival in patients with recurrent mantle cell lymphoma in the modern era: Progressive shortening in response duration and survival after each relapse. Blood Cancer J. 2019;9(6):50. doi: 10.1038/s41408-019-0209-5.

  55. Herrmann A, Hoster E, Zwingers T, et al. Improvement of overall survival in advanced stage mantle cell lymphoma. J Clin Oncol. 2009;27(4):511–8. doi: 10.1200/JCO.2008.16.8435.

  56. Fisher RI, Bernstein SH, Kahl BS, et al. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2006;24(30):4867–74. doi: 10.1200/JCO.2006.07.9665.

  57. Chen RW, Palmer JM, Tomassetti S, et al. Multi-center phase II trial of bortezomib and rituximab maintenance combination therapy in patients with mantle cell lymphoma after consolidative autologous stem cell transplantation. J Hematol Oncol. 2018;11(1):87. doi: 10.1186/s13045-018-0631-3.

  58. Albertsson-Lindblad A, Kolstad A, Laurell A, et al. Lenalidomide-bendamustine-rituximab in patients older than 65 years with untreated mantle cell lymphoma. Blood. 2016;128(14):1814–20. doi: 10.1182/blood-2016-03-704023.

  59. Wang ML, Blum KA, Martin P, et al. Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood. 2015;126(6):739–45. doi: 10.1182/blood-2015-03-635326.

  60. Rule S, Jurczak W, Jerkeman M, et al. Ibrutinib vs temsirolimus: 3-year follow-up of patients with previously treated mantle cell lymphoma from the phase 3, international, randomized, open-label RAY study. Leukemia. 2018;32(8):1799–803. doi: 10.1038/s41375-018-0023-2.

  61. Rule S, Dreyling M, Goy A, et al. Ibrutinib for the treatment of relapsed/refractory mantle cell lymphoma: extended 3.5-year follow-up from a pooled analysis. Haematologica. 2019;194(5):е211–е214. doi: 10.3324/haematol.2018.205229.

  62. Wang M, Rule S, Zinzani PL, et al. Long-Term Follow-Up of Acalabrutinib Monotherapy in Patients with Relapsed/Refractory Mantle Cell Lymphoma. Clin Lymphoma Myel Leuk. 2019;19:S316. doi: 10.1016/j.clml.2019.07.291.

  63. Telford C, Kabadi SM, Abhyankar S, et al. Matching-adjusted Indirect Comparisons of the Efficacy and Safety of Acalabrutinib Versus Other Targeted Therapies in Relapsed/Refractory Mantle Cell Lymphoma. Clin Ther. 2019;41(11):2357–79.e1. doi: 10.1016/j.clinthera.2019.09.012.

  64. Davids MS, Roberts AW, Seymour JF, et al. Phase I first-in-human study of Venetoclax in patients with relapsed or refractory non-Hodgkin lymphoma. J Clin Oncol. 2017;35(8):826–33. doi: 10.1200/JCO.2016.70.4320.

  65. Tam CS, Anderson MA, Pott C, et al. Ibrutinib plus Venetoclax for the treatment of mantle-cell lymphoma. N Engl J Med. 2018;378(13):1211–23. doi: 10.1056/NEJMoa1715519.

  66. Hess G, Herbrecht R, Romaguera J, et al. Phase III study to evaluate temsirolimus compared with investigator’s choice therapy for the treatment of relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2009;27(23):3822–9. doi: 10.1200/JCO.2008.20.7977.

  67. Kahl BS, Spurgeon SE, Furman RR, et al. A phase 1 study of the PI3Kδ inhibitor idelalisib in patients with relapsed/refractory mantle cell lymphoma (MCL). Blood. 2014;123(22):3398–405. doi: 10.1182/blood-2013-11-537555.

Intermediate Results of Prospective Observational Study: The 2-year Experience of Ibrutinib Therapy in Relapsed/Refractory Mantle Cell Lymphoma in Clinical Practice

VI Vorob’ev, VA Zherebtsova, EI Dubrovin, LA Bychenkova, YuB Kochkareva, LA Mukha, VL Ivanova, NK Khuazheva, VV Ptushkin

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

For correspondence: Vladimir Ivanovich Vorob’ev, MD, PhD, 5 2-i Botkinskii pr-d, Moscow, Russian Federation, 125284; e-mail: morela@mail.ru

For citation: Vorob’ev VI, Zherebtsova VA, Dubrovin EI, et al. Intermediate Results of Prospective Observational Study: The 2-year Experience of Ibrutinib Therapy in Relapsed/Refractory Mantle Cell Lymphoma in Clinical Practice. Clinical oncohematology. 2019;12(2):165-72.

DOI: 10.21320/2500-2139-2019-12-2-165-172


ABSTRACT

Aim. To assess efficacy and toxicity of ibrutinib monotherapy in patients with relapsed/refractory mantle cell lymphoma (MCL).

Materials & Methods. In this group of patients ibrutinib has been used since April 2016. Ibrutinib administration criteria were the age > 18 years and the confirmed MCL diagnosis with nuclear hyperexpression of cyclin D1 and t(11;14)(q13;q32) translocation. Poor physical status, pancytopenia, infectious complications (except for life-threatening conditions), blastoid variant, and the number of previous treatment lines were not regarded as contraindications to ibrutinib therapy. Oral ibrutinib was administered once a day at a dose of 560 mg before progression and until intolerable toxicity was observed.

Results. From April 20, 2016 to April 6, 2018 ibrutinib therapy was provided to 42 patients with relapsed/refractory MCL. The median age was 69 years (range 40–81); 64 % of patients were men; ECOG > 2 was registered in 14 % of patients; 38 % of patients had blastoid variant; the median number of previous treatment lines was 2 (range 1–11). The overall response rate was 85 % (35 % were in complete remission); 57 % (24/42) of patients remain on ibrutinib treatment for the period of 4–667 days. The median event-free survival (EFS) was 365 days (95% confidence interval was 31–698 days). The median overall survival was not achieved. In blastoid variant the median EFS was 92 days, in the alternative group the median was not achieved and EFS was 76 % for 12 months (< 0.001). In the majority of cases ibrutinib was well tolerated by patients. The most common complications were myalgia and muscle cramps (57 % cases), diarrhea (46 %, and grade 3 in 5 % cases), hemorrhagic complications (63 %, all of them of grade 1–2), and arrhythmia (7 %). Infectious complications were reported in 31 % of patients. In one case the start of ibrutinib treatment appeared to be problematic due to neutropenia of grade 4. Relative dose intensity was > 98 % (range 91.6–100 %). In 10 (24 %) patients ibrutinib treatment had to be adjusted (dose reduction or treatment interruption) due to toxicity and planned surgeries. None of ibrutinib recipients had to completely discontinue ibrutinib therapy due to complications.

Conclusion. These data on the use of ibrutinib in actual clinical practice are comparable with the results of international multicenter studies (PCYC-1104, SPARK, and RAY). Reduced toxicity profile and rather high speed of antitumor response allow for ibrutinib administration in cases of poor physical status, low blood count, and even infectious complications. However, some adverse effects are manifested not earlier than after 6-month treatment, which calls for continuous monitoring, especially when preparing for surgeries.

Keywords: mantle cell lymphoma, ibrutinib, relapse, refractory course, targeted therapy.

Received: November 4, 2018

Accepted: February 11, 2019

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. 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.

  3. Smith A, Roman E, Appleton S, et al. Impact of novel therapies for mantle cell lymphoma in the real world setting: a report from the UK’s Haematological Malignancy Research Network (HMRN). Br J Haemotol. 2018;181(2):215–28. doi: 10.1111/bjh.15170.

  4. Leux C, Maynadie M, Troussard X, et al. Mantle cell lymphoma epidemiology: a population-based study in France. Ann Hematol. 2014;93(8):1327–33. doi: 10.1007/s00277-014-2049-5.

  5. 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.

  6. Romaguera JE, Fayad LE, Feng L, et al. Ten-year follow-up after intense chemoimmunotherapy with Rituximab-HyperCVAD alternating with Rituximab-high dose methotrexate/cytarabine (R-MA) and without stem cell transplantation in patients with untreated aggressive mantle cell lymphoma. Br J Haematol. 2010;150(2):200–8. doi: 10.1111/j.1365-2141.2010.08228.x.

  7. 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.

  8. Le Gouill S, Thieblemont C, Oberic L, et al. Rituximab after Autologous Stem-Cell Transplantation in Mantle-Cell Lymphoma. N Engl J Med. 2017;377(13):1250–60. doi: 10.1056/nejmoa1701769.

  9. Воробьев В.И., Кравченко С.К., Гемджян Э.Г. и др. Мантийноклеточная лимфома: программное лечение первичных больных в возрасте до 65 лет. Клиническая онкогематология. 2013;6(3):274–81.

    [Vorob’ev VI, Kravchenko SK, Gemdzhian EG, et al. Mantle cell lymphoma: program therapy for untreated patients under 65 years. Klinicheskaya onkogematologiya. 2013;6(3):274–81. (In Russ)]

  10. 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. 2013;381(9873):1203–10. doi: 10.1016/s0140-6736(12)61763-2.

  11. Flinn IW, van der Jagt R, Kahl BS, et al. Randomized trial of bendamustine-rituximab or R-CHOP/R-CVP in first-line treatment of indolent NHL or MCL: the BRIGHT study. Blood. 2014;123(19):2944–52. doi: 10.1182/blood-2013-11-531327.

  12. Kluin-Nelemans HC, Hoster E, Hermine O, et al. Treatment of older patients with mantle-cell lymphoma. N Engl J Med. 2012;367(6):520–31. doi: 10.1056/nejmoa1200920.

  13. Robak T, Huang H, Jin J, et al. Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N Engl J Med. 2015;372(10):944–53. doi: 10.1056/nejmoa1412096.

  14. Fisher RI, Bernstein SH, Kahl BS, et al. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol, 2006;24(30):4867–74. doi: 10.1200/jco.2006.07.9665.

  15. Goy A, Sinha R, Williams ME, et al. Single-agent lenalidomide in patients with mantle-cell lymphoma who relapsed or progressed after or were refractory to bortezomib: phase II MCL-001 (EMERGE) study. J Clin Oncol. 2013;31(29):3688–95. doi: 10.1200/jco.2013.49.2835.

  16. Dreyling M, Jurczak W, Jerkeman M, et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet. 2016;387(10020):770–8. doi: 10.1016/s0140-6736(15)00667-4.

  17. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013,369(6):507–16. doi: 10.1056/nejmoa1306220.

  18. Davids SM, Roberts AW, Seymour JF, et al. Phase I First-in-Human Study of Venetoclax in Patients with Relapsed or Refractory Non-Hodgkin Lymphoma. J Clin Oncol. 2017;35(8):826–33. doi: 10.1200/jco.2016.70.4320.

  19. Khan WN. Colonel Bruton’s kinase defined the molecular basis of X-linked agammaglobulinemia, the first primary immunodeficiency. J Immunol. 2012;188(7):2933–5. doi: 10.4049/jimmunol.1200490.

  20. Herrera AF, Jacobsen ED. Ibrutinib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2014;20(21):5365–71. doi: 10.1158/1078-0432.ccr-14-0010.

  21. Honigberg LA, Smith AM, Sirisawad M, et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci USA. 2010;107(29):13075–80. doi: 10.1073/pnas.1004594107.

  22. Cinar M, Hamedani F, Mo Z, et al. Bruton tyrosine kinase is commonly over expressed in mantle cell lymphoma and its attenuation by Ibrutinib induces apoptosis. Leuk Res. 2013;37(10):1271–7. doi: 10.1016/j.leukres.2013.07.028.

  23. de Rooij MFM, Kuil A, Geest CR, et al. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012;119(11):2590–4. doi: 10.1182/blood-2011-11-390989.

  24. Ponader S, Chen S-S, Buggy JJ, et al. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood. 2012;119(5):1182–9. doi: 10.1182/blood-2011-10-386417.

  25. Buggy JJ, Elias L. Bruton tyrosine kinase (BTK) and its role in B-cell malignancy. Int Rev Immunol. 2012;31(2):119–32. doi: 10.3109/08830185.2012.664797.

  26. Herman SE, Gordon AL, Hertlein E, et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood. 2011;117(23):6287–96. doi: 10.1182/blood-2011-01-328484.

  27. Cheng S, Ma J, Guo A, et al. BTK inhibition targets in vivo CLL proliferation through its effects on B-cell receptor signaling activity. Leukemia. 2014;28(3):649–57. doi: 10.1038/leu.2013.358.

  28. Advani RH, Buggy JJ, Sharman JP, et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J Clin Oncol. 2013;31(1):88–94. doi: 10.1200/jco.2012.42.7906.

  29. Wang M, Rule S, Martin P, et al. Single-agent ibrutinib demonstrates safety and durability of response at 2 years follow-up in patients with relapsed or refractory mantle cell lymphoma: updated results of an international, multicenter, open-label phase 2 study. Blood. 2014;124(21):4453, abstract.

  30. Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059–68. doi: 10.1200/jco.2013.54.8800.

  31. Rule S, Dreyling M, Goy A, et al. Outcomes in 370 patients with mantle cell lymphoma treated with ibrutinib: a pooled analysis from three open-label studies. Br J Haematol. 2017;179(3):430–8. doi: 10.1111/bjh.14870.

  32. Wang ML, Rule S, Martin P, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369(6):507–16. doi: 10.1056/nejmoa1306220.

  33. Cheah CY, Chihara D, Romaguera JE, et al. Patients with mantle cell lymphoma failing ibrutinib are unlikely to respond to salvage chemotherapy and have poor outcomes. Ann Oncol. 2015;26(6):1175–9. doi: 10.1093/annonc/mdv111.

  34. Martin P, Maddocks K, Noto K, et al. Poor overall survival of patients with ibrutinib-resistant mantle cell lymphoma. Blood. 2014;124(21):3047, abstract.

  35. Balasubramanian S, Schaffer M, Deraedt W, et al. Mutational analysis of patients with primary resistance to single-agent ibrutinib in relapsed or refractory mantle cell lymphoma (MCL). Blood. 2014;124(21):78, abstract.

  36. Woyach JA, Furman RR, Liu T-M, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370(24):2286–94. doi: 10.1056/nejmoa1400029.

  37. Sarkozy C, Traverse-Glehen A, Bachy E, et al. Comparative Effectiveness of Single-Agent Ibrutinib in the Ray Trial Versus Real-World Treatment in the Lyon-Sud Database in Patients with Relapsed or Refractory Mantle Cell Lymphoma. Blood. 2017;130: 2770, abstract.

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.

Role of Superficial CD200 Marker in Differential Diagnosis of Malignant B-Cell Lymphoproliferative Diseases

YuV Mirolyubova, EA Stadnik, TS Nikulina, VV Strugov, TO Andreeva, YuV Virts, RV Grozov, AYu Zaritskey

Federal Almazov North-West Medical Research Centre, 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, Nikulina TS, et al. Role of Superficial CD200 Marker in Differential Diagnosis of Malignant B-Cell Lymphoproliferative Diseases. Clinical oncohematology. 2017;10(2):169–75 (In Russ).

DOI: 10.21320/2500-2139-2017-10-2-169-175


ABSTRACT

Background & Aims. Flow cytometry is successfully used for diagnosis of malignant lymphoproliferative disorders. However, there are atypical cases that are difficult to interpret; thus, new markers relevant for the differential diagnosis are to be searched for. The aim is to analyze CD200 expression in patients with B-cell lymphoproliferative disorders.

Materials & Methods. 187 patients with chronic lymphocytic leukemia (CLL), 14 patients with mantle cell lymphoma (MCL), 9 patients with marginal zone lymphoma (MZL), and 5 patients with hairy cell leukemia (HCL) were enrolled in the study. Neoplasm was not confirmed in 12 subjects. The patients underwent the following tests: CBC, immunophenotyping of peripheral blood or bone marrow lymphocytes, and a cytogenetic test. In some cases, an additional immunohistochemical test of bone marrow trepanobiopsy or lymph node biopsy samples was required.

Results. In all cases of CLL and HCL, the CD200 expression was positive; mean fluorescence intensity was higher in these cases as compared to other groups. Negative expression of CD200 prevailed in MCL patients; however, at the same time 2 cases of intermediate and positive expression were reported, both showing moderate fluorescence intensity values. CD200 expression was heterogeneous in MZL patients.

Conclusion. The CD200 negative expression excludes typical HCL and CLL. Additional cytogenetic and immunnohistoсhemical tests should be performed in such cases to verify the diagnosis, first of all, MCL or MZL.

Keywords: CD200, flow cytometry, diagnosis, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone lymphoma, hairy cell leukemia.

Received: September 7, 2016

Accepted: January 3, 2017

Read in PDF (RUS)pdficon


REFERENCES

  1. Купрышина Н.А., Тупицын Н.Н. Проточная цитометрия в онкогематологии. Часть II. Основы и нововведения в диагностике хронического лимфолейкоза. Клиническая онкогематология. 2012;5(4):349–54.
    [Kupryshina NA, Tupitsyn NN. Flow cytometry in hematology malignancies. Part II: ABC and news in diagnostics of chronic lymphocytic leukaemia. Klinicheskaya onkogematologiya. 2012;5(4):349–54. (In Russ)]
  2. Стадник Е.А., Стругов В.В., Вирц Ю.В., Зарицкий А.Ю. Хронический лимфолейкоз. Рекомендации по диагностике и лечению. Трансляционная медицина. 2012;17:104–15.
    [Stadnik EA, Strugov VV, Virts YuV, Zaritskey AYu. Chronic lymphocytic leukemia. Guidelines for diagnosis and treatment. Translyatsionnaya meditsina. 2012;17:104–15. (In Russ)]
  3. Swerdlow SH, Campo E, Harris NL, et al, eds. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon: IARC Press; 2008.
  4. Kohnke T, Wittmann VK, Sauter D, et al. Proposal For a Novel Scoring System For The Diagnosis оf CLL. Blood. 2013;122(21):47–5599 (Plenary Abstracts).
  5. Morice WG, Kurtin PJ, Hodnefield JM, et al. Predictive Value of Blood and Bone Marrow Flow Cytometry in B-Cell Lymphoma Classification: Comparative Analysis of Flow Cytometry and Tissue Biopsy in 252 Patients. Mayo Clin Proc. 2008;83(7):776–85. doi: 10.4065/83.7.776.
  6. Луговская С.А., Кисиличина Д.Г., Почтарь М.Е. и др. Новые маркеры (CD160, CD200, LAIR-1) в диагностике В-клеточных лимфопролиферативных заболеваний. Клиническая онкогематология. 2013;6(1):45–52.
    [Lugovskaya SA, Kisilichina DG, Pochtar’ ME, et al. New markers (CD160, CD200, and LAIR-1) in diagnosis of B-cell lymphoproliferative disorders. Klinicheskaya onkogematologiya. 2013;6(1):45–52. (In Russ)]
  7. Brunetti L, Di Noto R, Abate G, et al. CD200/OX2, a cell surface molecule with immuno-regulatory function is consistently expressed on hairy cell leukaemia neoplastic cells. Br J Haematol. 2009;145(5):665–78. doi: 10.1111/j.1365-2141.2009.07644.x.
  8. Palumbo GA, Parrinello N, Fargione G, et al. CD200 expression may help in differential diagnosis between mantle cell lymphoma and B-cell chronic lymphocytic leukemia. Leuk Res. 2009;33(9):1212–6. doi: 10.1016/j.leukres.2009.01.017.
  9. Dorfman DM, Shahsafaei A. CD200 (OX-2 Membrane Glycoprotein) Expression in B Cell–Derived Neoplasms. Am J Clin Pathol. 2010;134(5):726–33. doi: 10.1309/ajcp38xrrugsqovc.
  10. Sander B. Mantle cell lymphoma: recent insights into pathogenesis, clinical variability, and new diagnostic markers. Semin Diagn Pathol. 2011;28(3):245–55. doi: 10.1053/j.semdp.2011.02.010.
  11. Alapat D, Coviello-Malle J, Owens R, et al. Diagnostic Usefulness and Prognostic Impact of CD200 Expression in Lymphoid Malignancies and Plasma Cell Myeloma. Am J Clin Pathol. 2012;137(1):93–100. doi: 10.1309/ajcp59uorcyzevqo.
  12. El Desoukey NA, Afify RA, Amin DG, et al. CD200 expression in B-cell chronic lymphoproliferative disorders. J Investig Med. 2012;60(1):56–61. doi: 10.2310/jim.0b013e31823908f9.
  13. Pillai V, Pozdnyakova O, Charest K, et al. CD200 flow cytometric assessment and semiquantitative immunohistochemical staining distinguishes hairy cell leukemia from hairy cell leukemia-variant and other B-cell lymphoproliferative disorders. Am J Clin Pathol. 2013;140(4):536–43. doi: 10.1309/ajcpebk31vqqnddr.
  14. Challagundla P, Medeiros LJ, Kanagal-Shamanna R, et al. Differential Expression of CD200 in B-Cell Neoplasms by Flow Cytometry Can Assist in Diagnosis, Subclassification, and Bone Marrow Staging. Am J Clin Pathol. 2014;142(6):837–44. doi: 10.1309/ajcpbv9elxc0ecvl.
  15. Sandes AF, de Lourdes Chauffaille M, Regina C, et al. CD200 Has an Important Role in the Differential Diagnosis of Mature B-Cell Neoplasms by Multiparameter Flow. Cytometry. 2013;86(2):98–105. doi: 10.1002/cyto.b.21128.
  16. McCaughan GW, Clark MJ, Barclay AN. Characterization of the human homolog of the rat MRC OX-2 membrane glycoprotein. Immunogenetics. 1987;25(5):329–35. doi: 10.1007/bf00404426.
  17. Wright GJ, Jones M, Puklavec MJ, et al. The unusual distribution of the neuronal/lymphoid cell surface CD200 (OX2) glycoprotein is conserved in humans. Immunology. 2001;102(2):173–9. doi: 10.1046/j.1365-2567.2001.01163.x.
  18. Kretz-Rommel A, Qin F, Dakappagari N, et al. CD200 expression on tumor cells suppresses antitumor immunity: new approaches to cancer immunotherapy. J Immunol. 2007;178(9):5595–605. doi: 10.4049/jimmunol.178.9.5595.
  19. Moreaux J, Hose D, Reme T, et al. CD200 is a new prognostic factor in multiple myeloma. Blood. 2006;108(13):4194–7. doi: 10.1182/blood-2006-06-029355.
  20. Tonks A, Hills R, White P, et al. CD200 as a prognostic factor in acute myeloid leukemia. Leukemia. 2007;21(3):566–8. doi: 10.1038/sj.leu.2404559.
  21. Moreaux J, Veyrune JL, Reme T, et al. CD200: a putative therapeutic target in cancer. Biochem Biophys Res Commun. 2008;366(1):117–22. doi: 10.1016/j.bbrc.2007.11.103.
  22. Kretz-Rommel A, Bowdish KS. Rationale for anti-CD200 immunotherapy in B-CLL and other hematologic malignancies: new concepts in blocking immune suppression. Expert Opin Biol Ther. 2008;8(1):5–15. doi: 10.1517/14712598.8.1.5.

Mantle cell lymphoma: program therapy for untreated patients under 65 years

V.I. Vorobyev1, S.K. Kravchenko1, E.G. Gemdjian1, Yu. Yu. Lorie 2, A.U. Magomedova1, A.L. Melikyan1, J.K. Mangasarova1, D.S. Mar’yin1, E.I. Dubrovin1, T.N. Obukhova1, S.A. Makhinya, V.A. Zherebtsova3, M.A. Vernyuk4, N.G. Tyurina4, and V.G. Savchenko1

1 Hematology Research Center, RF Ministry of Health, Moscow, Russian Federation

2 Moscow Oncology Clinic #3, Russian Federation

3 Central Clinical Hospital with Polyclinic, RF Presidential Executive Office, Moscow, Russian Federation

4 P.A. Hertzen Moscow Oncology Research Institute, Moscow, Russian Federation


ABSTRACT

Background: Mantle cell lymphoma (MCL) is aggressive B-cell neoplasm which is diagnosed predominantly among older men. The use of high-dose Ara-C (12 g/m2 per course), autoSCT, and rituximab at all stages of therapy is the most effective approach but it is feasible only in patients under 60–65 years. High efficacy of gemcitabine and oxaliplatin-based regimens and irinotecan in relapsed or refractory MCL justifies their use in first-line therapy.

Objective: Assessment of toxicity and efficacy of R-DA-EPOCH/R-GIDIOX- and R-DA-EPOCH/R-HD-Met-Ara-C-regimens in primary MCL patients selected for autoSCT.

Patients and Methods: Since May 2008, 41 untreated MCL pts (median age: 54 years [29–64], M/F: 73%/27%, MIPIb: 29.3% low, 36.6% intermediate, 34.1% high risk) have been enrolled. After first R-EPOCH course (W. Wilson, 2003) completed, the patients were stratified according to toxicity emerged into 2 therapeutic groups: R-DA-EPOCH/R-HD-Met-AraC or R-DA-EPOCH/R-GIDIOX. In absence of grade 4 hematological toxicity for more than 3 days, serious infectious complications, or signs of renal failure, the pts received the R-HD-Met-Ara-C (R, 375 mg/m2 on Day 0; methotrexate, 1000 mg/m2 for 24 hours, Day 1; cytarabine, 3000 mg/m2 q 12 hrs on Days 2–3) regimen. When any of the above complications was present, the pts received the R-GIDIOX (R, 375 mg/m2 on Day 0; gemcitabine, 800 mg/m2 on Days 1 and 4; oxaliplatin, 120 mg/m2 on Day 2; irinotecan, 100 mg/m2 on Day 3; dexamethasone, 10 mg/m2 IV on Days 1–5; ifosfamide, 1000 mg/m2 on Days 1–5) regimen. Then, these regimens were reversed into either R-DA-EPOCH/R-HD-Met-Ara-C or R-DA-EPOCH/R-GIDIOX. Depending on the time until the complete response was achieved, pts received 6 to 8 therapeutic courses and autoSCT (BEAM-R) with in vivo purging using rituximab. Pts with residual tumor after autoSCT underwent local irradiation. R-maintenance was performed every 3 months for 3 years. Since Nov. 2011, all pts had received intrathecal CNS prophylaxis (including the patients who had undergone autoSCT during the year preceding Nov. 2011). The protocol was approved by the local ethics committee. Pts were analyzed using the intention-to-treat model. Toxicity assessment was performed for 124 R-DA-EPOCH, 87 R-HD-Met-Ara-C, and 51 R-GIDIOX courses.

Results: The median follow-up was 22 months (range 4–60). By April 2013, 35 patients had undergone autoSCT: 21 and 14 from R-HD-Met-Ara-C- and R-GIDIOX arm, respectively. One patient died from acute renal failure and septic shock at the induction stage after first HD-Met-AraC course. R-maintenance therapy was completed in 5 patients. In all patients who had received R-HD-Met-Ara-C, CR was achieved. In the R-GIDIOX arm, OR rate was 93%: 12 CR, 2 PR, and 1 case of disease progression after 5 courses. The most common non-hematological R-GIDIOX toxicity was related to the liver with elevated aminotransferases up to Grades 1–2 and 3–4 in 64.7% and 7.8% of cases, respectively, with no clinical manifestations. The sources of stem cells was PB in 27 out of 31 patients, and in 4 cases of harvest failure after 3 R-GIDIOX and 1 HD-Met-AraC BM was used. Hematological toxicity of R-GIDIOX course included grade 4 leukopenia in 74.5% (medium duration: 5 days, range: 1–13) and grade 4 thrombocytopenia in 39.2%. The estimated 5-years OS for the R-GIDIOX and R-HD-Met-AraC groups was 93 ± 7% and 79 ± 12%, respectively. The estimated 5-years EFS for the R-GIDIOX and R-HD-Met-AraC groups was 59 ± 19% and 74 ± 12%, respectively.

Conclusions: The HD-Met-Ara-C regimen is highly toxic, and it can be used only in 2/3 of patients under 65 years. The R-GIDIOX regimen is less toxic than HD-Met-Ara-C and equally effective with regard to the response induction and mobilizing necessary amount of autologous stem cells, so it can be recommended for the patients in whom Ara-C and methotrexate in high doses carry the high risk of life-threatening consequences.


Keywords: Mantle cell lymphoma, treatment, autoSCT, maintenance therapy.

Read in PDF (RUS)pdficon


REFERNCES

  1. Лорие Ю.Ю. Лимфома из клеток мантийной зоны: клинические формы, морфологические варианты, диагностика и лечение: Автореф. дис. … канд. мед. наук. М., 2005. [Loriye Yu.Yu. Limfoma iz kletok mantiynoy zony: klinicheskiye formy,  kand. med.¼morfologicheskiye varianty, diagnostika i lecheniye: Avtoref. dis.  nauk (Mantle cell lymphoma: clinical forms, morphological variants, diagnosis, and management. Author’s summary of dissertation for the degree of PHD). M., 2005.]
  2. Argatoff L.H., Connors J.M., Klasa R.J. et al. Mantle cell lymphoma: a clinicopathologic study of 80 cases. Blood 1997; 89: 2067–78.
  3. Yatabe Y., Suzuki R., Tobinai K. et al. Significance of cyclin D1 overexpression for the diagnosis of mantle cell lymphoma: a clinicopathologic comparison of cyclin D1-positive MCL and cyclin D1-negative MCL-like B-cell lymphoma. Blood 2000; 95: 2253–61.
  4. Wlodarska I., Meeus P., Stul M. et al. Variant t(2;11)(p11;q13) associated with the IgK-CCND1 rearrangement is a recurrent translocation in leukemic small-cell B-non-Hodgkin lymphoma. Leukemia 2004; 18: 1705–10.
  5. Komatsu H., Iida S., Yamamoto K. et al. A variant chromosome translocation at 11q13 identifying PRAD1/Cyclin Dl as the BCL-1 Gene. Blood 1994; 84: 1226–31.
  6. Vose J.M. Mantle cell lymphoma: 2012 update on diagnosis, risk-stratification, and clinical management. J. Hematol. 2012; 87(6): 604–9.
  7. Vegliante M.C., Palomero J., Perez-Galen P. et al. SOX11 regulates PAX5 expression and blocks terminal B-cell differentiation in aggressive mantle cell lymphoma. Blood 2013; 121(12): 2175–85.
  8. Mozos A., Royo A., Hartmann E. et al. SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype. Haematologica 2009; 94(11): 1555–62.
  9. Fernandez V., Salamero O., Espinet B. et al. Genomic and gene expression profiling defines indolent forms of mantle cell lymphoma. Cancer Res. 2010; 70(4): 1408–18.
  10. 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: 791–8.
  11. Howard O.M., Gribben J.G., Neuberg D.S. et al. Rituximab and CHOP induction therapy for newly diagnosed mantle cell lymphoma: molecular complete responses are not predictive of progression-free survival. Clin. Oncol. 2002; 20: 1288–94.
  12. Lenz G., Dreyling M., Hoster E. et al. Immunochemotherapy With Rituximab and Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone Significantly Improves Response and Time to Treatment Failure, But Not Long-Term Outcome in Patients With Previously Untreated Mantle Cell Lymphoma: Results of a Prospective Randomized Trial of the German Low Grade Lymphoma Study Group (GLSG). Clin. Oncol. 2005; 23: 1984–92.
  13. Romaguera J., Fayad L., Feng L. et al. Ten-year follow-up after intense chemoimmunotherapy with Rituximab-HyperCVAD alternating with Rituximabhigh dose methotrexate/cytarabine (R-MA) and without stem cell transplantation in patients with untreated aggressive mantle cell lymphoma. J. Haematol. 2010; 150(2): 200–8.
  14. Geisler C.H., Kolstad A., Laurell A. et al. Nordic MCL2 trial update: sixyear follow-up after intensive immunochemotherapy for untreated mantle celllymphoma followed by BEAM or BEAC + autologous stem-cell support: still very long survival but late relapses do occur. J. Haematol. 2012; 158(3): 355–62.
  15. Hermine O., Hoster E., Walewski J. et al. Alternating Courses of 3x CHOP and 3x DHAP Plus Rituximab Followed by a High Dose ARA-C Containing Myeloablative Regimen and Autologous Stem Cell Transplantation (ASCT) Is Superior to 6 Courses CHOP Plus Rituximab Followed by Myeloablative Radiochemotherapy and ASCT In Mantle Cell Lymphoma: Results of the MCL Younger Trial of the European Mantle Cell Lymphoma Network (MCL net). Blood 2010; 116: Abstract 110.
  16. Pott C., Hoster E., Beldjord K. et al. R-CHOP/R-DHAP Compared to RCHOP Induction Followed by High Dose Therapy with Autologous Stem Cell Transplantation Induces Higher Rates of Molecular Remission In MCL: Results of the MCL Younger Intergroup Trial of the European MCL Network. Blood 2010; 116: Abstract 965.
  17. Tam C., Bassett R., Ledesma C. et al. Mature results of the M. D. Anderson Cancer Center risk-adapted transplantation strategy in mantle cell Lymphoma. Blood 2009; 113(1): 8.
  18. Bernstein S.H., Epner E., Unger J.M. et al. A phase II multicenter trial of hyperCVAD MTX/Ara-C and rituximab in patients with previously untreated mantle cell lymphoma; SWOG 0213. Oncol. 2013 Mar 15.
  19. 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 2013; 121(1): 48–53.
  20. Hoster E., Dreyling M., Klapper W. et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood 2008; 111: 558–65.
  21. Tiemann M., Schrader C., Klapper W. et al. Histopathology, cell proliferation indices and clinical outcome in 304 patients with mantle cell lymphoma (MCL): a clinicopathological study from the European MCL Network. J. Haematol. 2005; 131: 29–38.
  22. Geisler C., Kolstad A., Laurell A. et al. The Mantle Cell Lymphoma International Prognostic Index (MIPI) is superior to the International Prognostic Index (IPI) in predicting survival following intensive first-line immunochemotherapy and autologous stem cell transplantation (ASCT). Blood 2010; 115: 1530–3.
  23. Geisler C. Front-line therapy of MCL. Hematologica 2010; 95: 1241–3.
  24. Damon L.E., Johnson J.L., Niedzwiecki D. et al. Immunochemotherapy and autologous stem-cell transplantation for untreated patients with mantle-cell lymphoma: CALGB 59909. Clin. Oncol. 2009; 27(36): 6101–8.
  25. Fayad L., Thomas D., Romaguera J. Update of the M. D. Anderson Cancer Center experience with hyper-CVAD and rituximab for the treatment of mantle cell and Burkitt-type lymphomas. Lymph. Myeloma 2007; 8(Suppl. 2): S57–62.
  26. Vigouroux S., Gaillard F., Moreau P. et al. High-dose therapy with autologous stem cell transplantation in first response in mantle cell lymphoma. Haematologica 2005; 90(11): 1580–2.
  27. Rodriguez J., Gutierrez A., Palacios A. et al. Rituximab, gemcitabine and oxaliplatin: An effective regimen in patients with refractory and relapsing mantle cell lymphoma. Lymphoma 2007; 48: 2172–8.
  28. Corazzelli G., Capobianco G., Arcamone M. et al. Long-term results of gemcitabine plus oxaliplatin with and without rituximab as salvage treatment for transplant-ineligible patients with refractory/relapsing B-cell lymphoma. Cancer Chemother. Pharmacol. 2009; 64: 907–16.
  29. Corazzelli G., Russo F., Capobianco G. et al. Gemcitabine, ifosfamide, oxaliplatin and rituximab (R-GIFOX), a new effective cytoreductive/mobilizing salvage regimen for relapsed and refractory aggressive non-Hodgkin’s lymphoma: results of a pilot study. Oncol. 2006; 17(Suppl. 4): iv18–24.
  30. Park B.B., Kim W.S., Eom H.S. et al. Salvage therapy with gemcitabine, ifosfamide, dexamethasone, and oxaliplatin (GIDOX) for B-cell non-Hodgkin’s lymphoma: a consortium for improving survival of lymphoma (CISL) trial. New Drugs 2011; 29: 154–60.
  31. Suzumiya J., Suzushima H., Maeda K. et al. Phase I study of the combination of irinotecan hydrochloride, carboplatin, and dexamethasone for the treatment of relapsed or refractory malignant lymphoma. J. Hematol. 2004; 79(3): 266–70.
  32. Hara S., Yokote T., Akioka T. et al. Successful treatment of refractory mantle cell lymphoma with irinotecan. Rinsho Ketsueki. 2005; 46(5): 358–62.
  33. Wilson W., Dunleavy K., Pittaluga S. et al. Phase II study of dose-adjusted EPOCH and rituximab in untreated diffuse large B-cell lymphoma with analysis of germinal center and post-germinal center biomarkers. Clin. Oncol. 2008; 26: 2717–24.
  34. Cheson B., Pfistner B., Juweid M. et al. Revised response criteria for malignant lymphoma. Clin. Oncol. 2007; 25: 579–86.