Autologous Stem Cell Transplantation in Primary Refractory Hodgkin’s Lymphoma: Supposed Zugzwang or Zwischenzug?

GD Petrova1, KN Melkova1, TZ Chernyavskaya1, NV Gorbunova1, BV Afanasev2, EA Demina1, VN Kostrykina1, VA Doronin1

1 N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

2 R.M. Gorbacheva Scientific Research Institute of Pediatric Hematology and Transplantation; Academician I.P. Pavlov First St. Petersburg State Medical University, 12 Rentgena str., Saint Petersburg, Russian Federation, 197022

For correspondence: Galina Dmitrievna Petrova, graduate student, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-13-59; e-mail: galina_petrova@bk.ru

For citation: Petrova GD, Melkova KN, Chernyavskaya TZ, et al. Autologous Stem Cell Transplantation in Primary Refractory Hodgkin’s Lymphoma: Supposed Zugzwang or Zwischenzug? Clinical oncohematology. 2015;8(3):321–30 (In Russ).


ABSTRACT

Background & Aims. The role of single and double autologous hematopoietic stem cell transplantations (autoSCT) in patients with primary refractory Hodgkin’s lymphoma (HL) has not been determined yet. The aim of the study is to present the results of a one-center prospective study evaluating the role of single and double autoSCT in patients with HL who have not achieved the complete remission (CR) after first line induction polychemotherapy (PCT).

Methods. 62 HL patients were enrolled in the study over the period from 2007 till 2014. High-dose chemotherapy (HDCT) with autoSCT was performed once in 53 patients, and twice in 10 patients.

Results. The study demonstrated an unfavorable prognostic impact of the large volume of previous chemotherapy on the overall survival (OS) rate after the autoSCT (= 0.03). Results of the 1st autoSCT had an independent prognostic value for the OS rate (= 0.004). The study identified the main indication for the 2nd autoSCT, namely, partial remission (PR) or stable disease (SD) achieved after the 1st autoSCT (when the 2nd HDCT with autoSCT should be preferred to the alternative treatment; = 0.004). Progressive disease (PD) after the first autoSCT is a contraindication for the second one. Due to low efficacy and high toxicity, the 2nd autoSCT does not improve outcomes when compared to alternative approaches (= 0.6). The importance of achieving CR at any stage of treatment which is associated with a long life span with no signs of disease and good quality of life has been demonstrated.

Conclusion. AutoSCT is an effective treatment option for patients without complete remission after the first line antitumor treatment. Carrying out 2nd autoSCT is advisable for patients who have reached the PR/SD after the first one. Patients with PD after the 1st autoSCT require an alternative treatment option.


Keywords: Hodgkin’s lymphoma, high-dose chemotherapy, autologous hematopoietic stem cell transplantation, primary resistance, double autoSCT.

Received: May 5, 2015

Accepted: June 2, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Kantarjian H, Pasquini R, Hamerschlak N, et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia after failure of first-line imatinib: a randomized phase 2 trial. Blood. 2007;109(12):5143–50. doi: 10.1182/blood-2006-11-056028.
  2. Kantarjian H, Giles F, Bhalla K, et al. Nilotinib is effective in patients with chronic myeloid leukemia in chronic phase after imatinib resistance or intolerance: 24-month follow-up results. Blood. 2011;117(4):1141–5. doi: 10.1182/blood-2010-03-277152.
  3. Лазорко Н.С., Ломаиа Е.Г., Сбитякова Е.И., Зарицкий А.Ю. Нилотиниб и дазатиниб в первой линии терапии больных хроническим миелолейкозом в хронической фазе. Современная онкология. 2011;13(1):38–40.
    [Lazorko NS, Lomaia EG, Sbityakova EI, Zaritskii AYu. Nilotinib and dazatinib as first line therapy of patients in chronic phase of chronic myeloid leukemia. Sovremennaya onkologiya. 2011;13(1):38–40. (In Russ)]
  4. Ломаиа Е.Г., Романова Е.Г., Сбитякова Е.И., Зарицкий А.Ю. Эффективность и безопасность ингибиторов тирозинкиназ 2-го поколения (дазатиниб, нилотиниб) в терапии хронической фазы хронического миелолейкоза. Онкогематология. 2013;2:22–33.
    [Lomaia EG, Romanova EG, Sbityakova EI, Zaritskii AYu. Efficacy and safety of 2nd generation tyrosine kinase inhibitors (dasatinib, nilotinib) in teatment of chronic phase of chronic myeloid leukemia. Onkogematologiya. 2013;2:22–33. (In Russ)]
  5. Туркина А.Г., Хорошко Н.Д., Гусарова Г.А. и др. Российский опыт применения нилотиниба во второй линии терапии больных хроническим миелолейкозом с резистентностью или непереносимостью иматиниба: оценка безопасности и эффективности в исследовании ENACT (расширенный доступ к нилотинибу в клинических исследованиях). Вестник гематологии. 2010;1(2):92–3.
    [Turkina AG, Khoroshko ND, Gusarova GA, et al. Russian experience in use of nilotinib in second line therapy of patients with chronic myeloid leukemia and imatinib resistance or intolerance: evaluation of safety and efficacy in ENACT trial (Expanding Nilotinib Access in Clinical Trials). Vestnik gematologii. 2010;1(2):92–3. (In Russ)]
  6. http://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm.
  7. Kantarjian H, Giles F, Gattermann N, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood. 2007;110(10):3540–6. doi: 10.1182/blood-2007-03-080689.
  8. Saglio G, Kim D, Issaragrisil S, et al. Nilotinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2010;362(24):2251–9. doi: 10.1517/14656566.2011.534780.
  9. Hochhaus A, Kantarjian H, Baccarani M, et al. Dasatinib induces notable hematologic and cytogenetic responses in chronic phase chronic myeloid leukemia after failure of imatinib therapy. Blood. 2007;109(6):2303–9. doi: 10.1182/blood-2006-09-047266.
  10. Kantarjian H, Shah N, Hochhaus A, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2010;362:2260–70. doi: 10.1056/nejmoa1002315.
  11. Shah R. Drug-induced hepatotoxicity: pharmacokinetic perspectives and strategies for risk reduction. Adv Drug React Toxicol Rev. 1999;18:181–233.
  12. Russmann S, Kullak-Ublick G, Grattagliano I. Current concepts of mechanisms in drug-induced hepatotoxicity. Curr Med Chem. 2009;16(23):3041–53.
  13. Teo YL, Ho HK, Chan A. Risk of tyrosine kinase inhibitors-induced hepatotoxicity in cancer patients: A meta-analysis. Cancer Treat Rev. 2013;39(2):199–206. doi: 10.1016/j.ctrv.2012.09.004.
  14. Saglio G, Pinilla-Ibarz J, Cortes J, et al. Intolerance to tyrosine kinase inhibitors in chronic myeloid leukemia. Blood. 2011;117(4):688−697. doi: 10.1002/cncr.25648.
  15. Rosti G, Castagnetti F, Gugliotta G, et al. Dasatinib and nilotinib in imatinib resistant Philadelphia-positive chronic myelogenous leukemia: a ‘head-to-head’ comparison. Leuk Lymphoma 2010;51(4):583–91. doi: 10.3109/10428191003637282.
  16. Shah R, Morganroth J, Shah D. Hepatotoxicity of Tyrosine Kinase Inhibitors: Clinical and Regulatory Perspectives. Drug Saf. 2013;36(7):491–503. doi: 10.1007/s40264-013-0048-4.
  17. Lammie A, Drobnjak M, Gerald W, et al. Expression of c-kit and kit ligand proteins in normal human tissues. J Histochem Cytochem. 1994;42(11):1417–25. doi: 10.1177/42.11.7523489.
  18. Grichnik J, Burch J, Burchette J, Shea C. The SCF/KIT pathway plays a critical role in the control of normal human melanocyte homeostasis. J Invest Dermatol. 1998;111(2):233–8.
  19. Kantarjian H, Pasquini R, Levy V, et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia resistant to imatinib at a dose of 400 to 600 milligrams daily: two-year follow-up of a randomized phase 2 study (START-R). Cancer. 2009;115(18):4136–47. doi: 10.1002/cncr.24504.
  20. Irvine E, Williams C. Treatment-, Patient-, and Disease-Related Factors and the Emergence of Adverse Events with Tyrosine Kinase Inhibitors for the Treatment of Chronic Myeloid Leukemia. Pharmacotherapy. 2013;33(8):868–81. doi: 10.1002/phar.1266.
  21. Van Etten RA. Cycling, stressed-out and nervous: cellular functions of cAbl. Trends Cell Biol. 1999;9(5):179–86. doi: 10.1016/s0962-8924(99)01549-4.
  22. Wasle B, Edwardson J. The regulation of exocytosis in the pancreatic acinar cell. Cell Signal. 2002;14(3):191–7. doi: 10.1016/s0898-6568(01)00257-1.
  23. Mooren F, Hlouschek V, Finkes T, et al. Early changes in pancreatic acinar cell calcium signalling after pancreatic duct obstruction. J Biol Chem. 2003;278(11):9361–9. doi: 10.1074/jbc.m207454200.
  24. Fitter S, Vandyke K, Gronthos S, Zannettino AC. Suppression of PDGF-induced PI3 kinase activity by imatinib promotes adipogsis and adiponectin secretion. J Mol Endocrinol. 2012;48(3):229–40. doi: 10.1530/jme-12-0003.
  25. Racil Z, Razga F, Drapalova J, et al. Mechanism of impaired glucose metabolism during nilotinib therapy in patients with chronic myelogenous leukemia. Haematologica. 2013;98(10):e124–6. doi: 10.3324/haematol.2013.086355.
  26. le Coutre P, Giles F, Hochhaus A, et al. Analysis of glucose profiles in imatinib resistant or intolerant chronic myelogenous leukemia (CML) patients treated with nilotinib: lack of correlation between glucose levels and nilotinib efficacy. Blood. 2007;110: Abstract 4588.
  27. Breccia M, Alimena G. Pleural/pericardic effusions during dasatinib treatment: incidence, management and risk factors associated to their development. Exp Opin Drug Saf. 2010;9(5):713–21. doi: 10.1517/14740331003742935.
  28. de Lavallade H, Punnialingam S, Milojkovic D, et al. Pleural effusions in patients with chronic myeloid leukaemia treated with dasatinib may have an immune-mediated pathogenesis. Br J Haematol. 2008;141(5):745–7. doi: 10.1111/j.1365-2141.2008.07108.x.
  29. Porkka K, Khoury H, Paquette R, et al. Dasatinib 100 mg once daily minimizes the occurrence of pleural effusion in patients with chronic myeloid leukemia in chronic phase and efficacy is unaffected in patients who develop pleural effusion. Cancer. 2010;116(2):377–86. doi: 10.1002/cncr.24734.
  30. Shah N, Kantarjian H, Kim D, et al. Six-year (yr) follow-up of patients (pts) with imatinib-resistant or -intolerant chronic-phase chronic myeloid leukemia (CML-CP) receiving dasatinib. J Clin Oncol. 2012;30:6506.
  31. Hasinoff BB. The cardiotoxicity and myocyte damage caused by small molecule anticancer tyrosine kinase inhibitors is correlated with lack of target specificity. Toxicol Appl Pharmacol. 2010;244(2):190–5. doi: 10.1016/j.taap.2009.12.032.
  32. Albini A, Pennesi G, Donatelli F, et al. Cardiotoxicity of anticancer drugs: the need for cardio-oncology and cardio-oncological prevention. J Natl Cancer Inst. 2010;102(1):14–25. doi: 10.1093/jnci/djp440.
  33. Strevel E, Ing D, Siu L. Molecularly targeted oncology therapeutics and prolongation of the QT interval. J Clin Oncol. 2007;25(22):3362–71. doi: 10.1200/jco.2006.09.6925.
  34. Haverkamp W, Breithardt G, Camm A, et al. The potential for QT prolongation and proarrhythmia by non-antiarrhythmic drugs: clinical and regulatory implications. Report on a policy conference of the European Society of Cardiology. Eur Heart J. 2000;21(15):1216–31. doi: 10.1053/euhj.2000.2249.
  35. Priori S, Schwartz P, Napolitano C, et al. Risk stratification in the long-QT syndrome. N Engl J Med. 2003;348(19):1866–74. doi: 10.1056/nejmoa022147.
  36. Sauer A, Moss A, McNitt S, et al. Long QT syndrome in adults. J Am Coll Cardiol. 2007;49(3):329–37. doi: 10.1016/j.jacc.2006.08.057.
  37. Center for Drug Evaluation and Research: Nilotinib Pharmacology/Toxicology Review and Evaluation; 2007.
  38. Le Coutre P, Ottmann O, Giles F, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is active in patients with imatinib-resistant or -intolerant accelerated-phase chronic myelogenous leukemia. Blood. 2008;111(4):1834–9. doi: 10.1182/blood-2007-04-083196.
  39. Kantarjian H, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med. 2006;354(24):2542–51. doi: 10.1056/nejmoa055104.
  40. Kim T, Rea D, Schwarz M, et al. Peripheral artery occlusive disease in chronic phase chronic myeloid leukemia patients treated with nilotinib or imatinib. Leukemia. 2013;27(6):1316–21. doi: 10.1038/leu.2013.70.
  41. Larson R, Hochhaus A, Hughes T, et al. Nilotinib vs imatinib in patients with newly diagnosed Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase: ENESTnd 3-year follow-up. Leukemia. 2012;26(10):2197–203. doi: 10.1038/leu.2012.134.
  42. Aichberger K, Herndlhofer S, Schernthaner G, et al. Progressive peripheral arterial occlusive disease and other vascular events during nilotinib therapy in CML. Am J Hematol. 2011;86(7):533–9. doi: 10.1002/ajh.22037.
  43. Verma D, Verstovsek S, Kantarjian H, et al. Malignancies occurring during therapy with tyrosine kinase inhibitors (TKIs) for chronic myeloid leukemia (CML) and other hematologic malignancies. Blood. 2011;118(16):4353–8. doi: 10.1182/blood-2011-06-362889.
  44. Hoffmann V, Baccarani M, Hasford J. The EUTOS population-based registry: incidence and clinical characteristics of 2904 CML patients in 20 European Countries. Leukemia. 2015;29(6):1336–43. doi: 10.1038/leu.2015.73 [Epub 2015 Mar 18]

High-Dose Chemotherapy and Autologous Stem Cells Transplantation for Relapsed/Refractory Hodgkin’s Lymphoma. Is There an Equal Right to Life?

N.V. Zhukov1,2, A.G. Rumyantsev1, A.L. Uss3, N.F. Milanovich3, V.V. Ptushkin1, B.V. Afanasyev4, N.B. Mikhaylova4, V.B. Larionova5, E.A. Demina5, E.E. Karamanesht6, N.G. Tyurina7, M.A. Vernyuk7, A.D. Kaprin7

1 Dmitrii Rogachev Federal Scientific Clinical Centre of Pediatric Hematology, Oncology and Immunology under the RF MH, Moscow, Russian Federation

2 N.I. Pirogov Russian National Research Medical University, Moscow, Russian Federation

3 National Center for Hematology and Bone Marrow Transplantation, Minsk, Belarus

4 R.M. Gorbacheva Memorial Institute of Children Oncology, Hematology and Transplantation under I.P. Pavlov State Medical University, Saint Petersburg, Russian Federation

5 N.N. Blokhin Cancer Research Center of RAMS, Moscow, Russian Federation

6 Kyiv Center for Bone Marrow Transplantation, Kyiv, Ukraine

7 P.A. Hertsen Moscow Oncological Research Institute, Moscow, Russian Federation

For citation: Zhukov N.V., Rumyantsev A.G., Uss A.L., Milanovich N.F., Ptushkin V.V., Afanas’ev B.V., Mikhailova N.B., Larionova V.B., Demina E.A., Karamanesht E.E., Tyurina N.G., Vernyuk M.A., Kaprin A.D. High-Dose Chemotherapy and Autologous Stem Cells Transplantation for Relapsed/Refractory Hodgkin’s Lymphoma. Is There an Equal Right to Life? Klin. onkogematol. 2014; 7(3): 317–26 (In Russ.).


ABSTRACT

Aim. Hodgkin’s lymphoma (HL) patients with primary refractory (PRef) course of disease or relapses refractory to the previous 2nd line therapy (RRel) often are not given high-dose chemotherapy with autologous stem cell support (ASCS), and this refuse is motivated by its poor efficacy and high toxicity in this population. The objective of this study was to evaluate the efficacy and safety of ASCS in this patient population.

Materials and methods. 372 patients with Hodgkin’s lymphoma undergoing ASCS between 01.1990 and 06.2013 were included in the trial. The reason for ASCS was: primary refractory disease in 132 (35.5 %) patients, relapse of the disease resistant to II line chemotherapy (refractory relapse) in 81 (22 %). The remaining 159 patients (42.5 %) either had a relapse for which they received no II line chemotherapy (a relapse with untested sensitivity) or a relapse that proved to be sensitive to previously performed II line therapy (sensitive relapse). These patients were assigned to a chemosensitive HL group.

Results. With a median follow-up of 51 months, the overall survival rate (OS) and the relapse-free survival rate (RFS) did not differ significantly between patients with RRel, PRef and chemosensitive HL group (> 0.05). Only freedom from treatment failure survival (FFTS) was significantly worse in patients with PRef HL (5-yrs EFS 42 % vs 58 % in patients with RRel vs 60 % in patients with chemosensitive HL group; = 0.004). 100-day mortality mostly caused by ASCS toxicity also did not differ significantly between groups (= 0.2). Irrespectively of primary reason for ASCS, long-term ASCS results significantly depended on response to the cytoreductive therapy. The effect of the cytoreductive therapy was assessed in 309 patients. When patients achieved complete, marked partial or partial remission, the 5-year overall survival rate, FFTS, and relapse-free survival rate was 78 %, 64 %, and 68 %, respectively. In patients with stabilization or progression of disease due to the cytoreductive therapy, these parameters were equal to 33 %, 24 % и 52 %, respectively (< 0.001 for OS and FFTS, = 0.005 for RFS).

Conclusion. In patients with primary refractory and refractory relapse of HL, ASCS has acceptable efficacy and early mortality which is comparable to that observed in patients with chemosensitive Hodgkin’s lymphoma, thus permitting to consider ASCS a potential therapeutic approach in patients with primary refractory disease and resistant relapses of Hodgkin’s lymphoma. Irrespectively of the initial disease course, the tumor response to the cytoreductive therapy is the most important predictive factor for the long-term ASCS results.


Keywords: Hodgkin’s lymphoma, high-dose chemotherapy, autologous hematopoietic stem cells transplantation, primary resistance, resistant relapse.

Address correspondence to: zhukov.nikolay@rambler.ru

Accepted: April 13, 2014

Read in PDF (RUS) pdficon


REFERENCES

  1.  Linch D., Winfield D., Goldstone A. et al. Dose intensification with autologous bone-marrow transplantation in relapsed and resistant Hodgkin’s disease: results of a BNLI randomised trial. Lancet 1993; 341: 1051.
  2. Schmitz N., Sextro M., Pfistner B. HDR-1: high-dose therapy (HDT) followed by hematopoietic stem cell transplantation (HSCT) for relapsed chemosensitive Hodgkin’s disease (HD): final results of a randomized GHSG and EBMT trial (HD-R1). Proc. Am. Soc. Clin. Oncol. 1999; 18(Suppl. 5): 18.
  3. Josting A., Franklin J., May M. et al. New prognostic score based on treatment outcome of patients with relapsed Hodgkin’s lymphoma registered in the database of the German Hodgkin’s lymphoma study group. J. Clin. Oncol. 2002; 20: 221–30.
  4. Longo L., Duffey P.L., Young R.C. et al. Conventional-dose salvage combination chemotherapy in patients relapsing with Hodgkin’s disease after combination chemotherapy: the low probability for cure. J. Clin. Oncol. 1992; 10: 210–8.
  5. Brusamolino E., Orlandi E., Canevari A. et al. Results of CAV regimen (CCNU, melphalan, and VP-16) as third-line salvage therapy for Hodgkin’s disease. Ann. Oncol. 1994; 5: 427–32.
  6. Bonfante V., Santoro A., Viviani S. et al. Outcome of patients with Hodgkin’s disease failing after primary MOPP/ABVD. J. Clin. Oncol. 1997; 15: 528–34.
  7. Josting A., Rueffer U., Franklin J. et al. Prognostic factors and treatment outcome in primary progressive Hodgkin lymphoma: a report from the German Hodgkin Lymphoma Study Group. Blood 2000; 96: 1280–6.
  8. Josting A., Rudolph C., Mapara M. et al. Cologne high-dose sequential chemotherapy in relapsed and refractory Hodgkin lymphoma: results of a large multicenter study of the German Hodgkin Lymphoma Study Group (GHSG). Ann. Oncol. 2005; 16(1): 116–23.
  9. Argiris A., Seropian S., Cooper D.L. High-dose BEAM chemotherapy with autologous peripheral blood progenitor-cell transplantation for unselected patients with primary refractory or relapsed Hodgkin’s disease. Ann. Oncol. 2000; 11: 665–72.
  10. Ferme C., Mounier N., Divine M. et al. Intensive salvage therapy with high dose chemotherapy for patients with advanced Hodgkin’s disease in relapse or failure after initial chemotherapy: Results of the Groupe d’Etudes des Lymphomes de l’Adulte H89 Trial. J. Clin. Oncol. 2002; 20: 467–75.
  11. Constans M., Sureda A., Terol M.J. et al. Autologous stem cell transplantation for primary refractory Hodgkin’s disease: Results and clinical variables affecting outcome. Ann. Oncol. 2003; 14: 745–51.
  12. Sweetenham J.W., Carella A.M., Taghipour G. et al. High-dose therapy and autologous stem-cell transplantation for adult patients with Hodgkin’s disease who do not enter remission after induction chemotherapy: Results in 175 patients reported to the European Group for Blood and Marrow Transplantation. Lymphoma Working Party. J. Clin. Oncol. 1999; 17: 3101–9.
  13. Gopal A.K., Metcalfe T.L., Gooley T.A. et al. High-Dose Therapy and Autologous Stem Cell Transplantation for Chemoresistant Hodgkin Lymphoma: The Seattle Experience. Cancer 2008; 113(6): 1344–50.
  14. Sureda A., Arranz R., Iriondo A. et al. Autologous stem-cell transplantation for Hodgkin’s disease: results and prognostic factors in 494 patients from the Grupo Espanol de Linfomas/Transplante Autologo de Medula Osea Spanish Cooperative Group. J. Clin. Oncol. 2001; 19(5): 1395–404.
  15. Czyz J., Dziadziuszko R., Knopinska-Postuszuy W. et al. Outcome and prognostic factors in advanced Hodgkin’s disease treated with high-dose chemotherapy and autologous stem cell transplantation: a study of 341 patients. Ann. Oncol. 2004; 15(8): 1222–30.
  16. Sureda A., Constans M., Iriondo A. et al. Prognostic factors affecting long-term outcome after stem cell transplantation in Hodgkin’s lymphoma autografted after a first relapse. Ann. Oncol. 2005; 16(4): 625–33.