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.

Haploidentical hematopoietic stem cell transplantation in children with acute myeloid leukemia: evolution of method and our data

N.N. Subbotina, I.S. Dolgopolov, A.V. Popa, V.K. Boyarshinov, R.I. Pimenov, and G.L. Mentkevich

Pediatric Oncology and Hematology Research Institute, N.N. Blokhin Russian Cancer Research Center, RAMS, Moscow, Russian Federation


ABSTRACT

This article presents the results of haploidentical stem cell transplantation in children with prognostically unfavorable AML. The study group included 18 pts at the age of 1–18. The disease status at the transplantation time was as follows: high risk AML in first remission (n=4, 22 %), more than two remissions (n=7, 39 %), no remission (n=4, 22 %), or secondary AML in remission (n=3, 17 %). All patients received reduced-intensity conditioning regimen followed by HSCT from haploidentical donors. Hematologic recovery occurred in 17 out of 18 pts in a mean time of 11 days and 12 days for WBC and platelets, respectively. One patient with no remission at the time of transplantation died from leukemia progression and infection with no signs of hematologic recovery. The regimen toxicity was mild and manageable. Acute GVHD of I/II and III degree occurred in 88 % and 6 % of pts, respectively. Chronic GVHD occurred in 85 % of pts, having been quite severe in one pt. The causes of death were infection (n=2, 11 %) or disease relapse/progression (n = 5, 28 %). Eleven pts (61 %) are still alive and disease-free. EFS is 57.5 % with a mean follow-up of 84 (1–144) months. TRM is 13.3 % with a mean follow-up of 124 months.


Keywords: pediatric acute myeloid leukemia, unfavorable prognosis, haploidentical hematopoietic stem cell transplantation.

Read in PDF (RUS) pdficon


REFERENCES

  1. Rubnitz J.E. Childhood acute myeloid leukemia. Curr. Treat. Options Oncol. 2008; 9: 95–105.
  2. Creutzig U., Zimmermann M., Henze G. et al. Treatment strategies and long-term results in paediatric patients treated in four consecutive AML BFM trials. Leukemia 2005; 19(12): 2030–42.
  3. Perel Y., Auvrignon A., Vannier J.P. et al. Treatment of childhood acute myeloblastic leukemia: dose intensification improves outcome and maintenance therapy is of no benefit—multicenter studies of the French LAME (Leucemie Aigue Myeloblastique Enfant) Cooperative Group. Leukemia 2005; 19(12): 2082–9.
  4. August K.J., Narendran A., Neville K.A. Pediatric Relapsed or Refractory Leukemia: New Pharmacotherapeutic Developments and Future Directions. Drugs 2013; 73: 439–61.
  5. Grimwade D., Walker H., Oliver F. et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children’s Leukaemia Working Parties. Blood 1998; 92: 2322–33.
  6. Gorman M.F., Ji L., Hutchinson R. et al. Outcome for children treated for relapsed or refractory acute myelogenous leukemia (rAML): a Therapeutic Advances in Childhood Leukemia (TACL) Consortium study. Pediatr. Blood Cancer 2010; 55(3): 421–9.
  7. Liu D.-H., Xu L.-P., Liu K.-Y. et al. Long-term outcomes of unmanipulated haploidentical HSCT for paediatric patients with acute leukaemia. Bone Marrow Transplant. 2013; 48: 1519–24.
  8. Shaw P.J., Kan F., Pulsipher M.A. et al. Outcomes of pediatric bone marrow transplantation for leukemia and myelodysplasia using matched sibling, mismatched related, or matched unrelated donors. Blood 2010; 116: 4007–15.
  9. Gluckman E., Vanderson R., William A. et al. Outcome of cord-blood transplantation from related and unrelated donors. N. Engl. J. Med. 1997; 337(6): 373–81.
  10. Kurtzberg J., Laughlin M., Graham M.L. et al. Placental blood as a source of hematopoietic stem cells for transplantation into unrelated recipients. N. Engl. J. Med. 1996; 335: 157–66.
  11. Wagner J.E., Rosenthal J., Sweetman R. et al. Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: analysis of engraftment and acute graft-versus-host disease. Blood 1996; 88: 795–802.
  12. Silberstein L.E., Jefferies L.C. Placental-blood banking — a new frontier in transfusion medicine. N. Engl. J. Med. 1996; 335: 199–201.
  13. Rubinstein P., Rosenfield R.E., Stevens C.E. Stored placental blood for unrelated bone marrow reconstitution. Blood 1993; 81: 1679–90.
  14. Rubinstein P., Dobrila L., Rosenfield R.E. et al. Processing and cryopreservation of placental/umbilical cord blood for unrelated bone marrow reconstitution. Proc. Natl. Acad. Sci. U S A 1995; 92: 10119–22.
  15. Rocha V., Gluckman E., Frassoni F. et al. Unrelated cord blood transplantation: outcomes after single-unit intrabone injection compared with double-unit intravenous injection in patients with hematological malignancies. Transplantation 2013; 95(10): 1284–91.
  16. Page K.M., Zhang L., Kurtzberg J. et al. Total colony-forming units are a strong, independent predictor of neutrophil and platelet engraftment after unrelated umbilical cord blood transplantation: a single-center analysis of 435 cord blood transplants. Biol. Blood Marrow Transplant. 2011; 17(9): 1362–74.
  17. Barker J.N., Scaradavou A., Stevens C.E. Combined effect of total nucleated cell dose and HLA match on transplantation outcome in 1061 cord blood recipients with hematologic malignancies. Blood 2010; 115: 1843–9.
  18. Sideri A., Neokleous N., Gluckman E. An overview of the progress on double umbilical cord blood transplantation. Haematologica 2011; 96(8): 1213–20.
  19. Rocha V., Crotta A., Gluckman E. et al. Double cord blood transplantation: extending the use of unrelated umbilical cord blood cells for patients with hematological diseases. Best Pract. Res. Clin. Haematol. 2010; 23(2): 223–9.
  20. Powles R.L., Morgenstern G.R., Robinson B. et al. Mismatched family donors for bone marrow transplantation as treatment for acute leukaemia. Lancet 1983; 1: 612.
  21. Beatty P.G., Clift R.A., Storb R. et al. Marrow transplantation from related donors other than HLA identical siblings. N. Engl. J. Med. 1985; 313: 765.
  22. Hows J.M., Yin J.L., Goldman J.M. et al. Histocompatible unrelated volunteer donors compared with HLA nonidentical family donors in marrow transplantation for aplastic anemia and leukemia. Blood 1986; 68(6): 1322–8.
  23. Reisner Y., Kapoor N., Good R.A. et al. Transplantation for acute leukeamia with HLA-A and B non identical parental marrow cells fractionated with soybean agglutinin and sheep red blood cells. Lancet 1981; 2(8242): 327–31.
  24. Mehta J., Singhal S., Gee A.P. et al. Bone marrow transplantation from partially HLA-mismatched family donors for acute leukemia: single-center experience of 201 patients. Bone Marrow Transplant. 2004; 33: 389–96.
  25. O’Reilly R.J., Kernan N.A., Cunningham I. Allogeneic transplants depleted of T cells by soybean lectin agglutination and E-rosette depletion. Bone Marrow Transplant. 1988; 3: 3–6.
  26. Schwartz E., Lapidot T., Reisner Y. et al. Abrogation of bone marrow allograft resistance in mice by increased total body irradiation correlates with eradication of host clonable T cells and alloreactive cytotoxic precursors. J. Immunol. 1987; 138(2): 460–5.
  27. Terenzi A., Lubin I., Rabi I. et al. Enhancement of T-cell depleted bone marrow allografts inmice by thiotepa. Transplantation 1990; 50(4): 717–20.
  28. Cobbold S.P., Martin G., Waldmann H. et al. Monoclonal antibodies to promote marrow engraftment and tissue graft tolerance. Nature 1986; 323(6084): 164–6.
  29. Reisner Y., Itzicovitch L., Sharon N. et al. Hematopoietic stem cell transplantation using mouse bone-marrow and spleen cells fractionated by lectins. Proc. Natl. Acad. Sci. U S A 1978; 75(5): 2933–6.
  30. Aversa F., Tabilio A., Giannoni C. et al. Successful engraftment of T-celldepleted haploidentical “threeloci” incompatible transplants in leukemia patients by addition of recombinant human granulocyte colony-stimulating factor mobilized peripheral blood progenitor cells to bone marrow inoculum. Blood 1994; 84(11): 3948–55.
  31. Aversa F., Terenzi A., Ballanti S. et al. Full haplotype-mismatched hematopoietic stem-cell transplantation: A phase II study in patients with acute leukemia at high risk of relapse. J. Clin. Oncol. 2005; 23(15): 3447–54.
  32. Schumm M., Lang P., Taylor G. et al. Isolation of highly purified autologous and allogeneic peripheral CD34+ cells using the CliniMACS device. J. Hematother. 1999; 8: 209–18.
  33. Klingebiel T., Cornish J., Labopin M. et al. Pediatric Diseases and Acute Leukemia Working Parties of the European Group for Blood and Marrow Transplantation (EBMT). Results and factors influencing outcome after fully haploidentical hematopoietic stem cell transplantation in children with very high-risk acute lymphoblastic leukemia: impact of center size: an analysis on behalf of the Acute Leukemia and Pediatric Disease Working Parties of the European Blood and Marrow Transplant group. Blood 2010; 115: 3437–46.
  34. Ciceri F., Labopin M., Rocha V. et al. Acute Leukemia Working Party (ALWP) of European Blood and Marrow Transplant (EBMT) Group. A survey of fully haploidentical hematopoietic stem cell transplantation in adults with highrisk acute leukemia: a risk factor analysis of outcomes for patients in remission at transplantation. Blood 2008; 112(9): 3574–81.
  35. Barfield R.C., Otto M., Houston J. et al. A one-step large-scale method for T- and B-cell depletion of mobilized PBSC for allogeneic transplantation. Cytotherapy 2004; 6: 1–6.
  36. Oevermann L., Handgretinger R. New strategies for haploidentical transplantation. Pediatr. Res. 2012; 71(4 Pt. 2): 418–26.
  37. Hale G.A., Kasow K., Gan K. et al. Haploidentical Stem Cell Transplantation with CD3 Depleted Mobilized Peripheral Blood Stem Cell Grafts for Children with Hematologic Malignancies. 47th Annual Meeting of the American Society of Hematology, 10–13 December 2005, Atlanta, GA, USA.
  38. Hale G.A., Kasow K., Madden R. et al. Mismatched family member donor transplantation for patients with refractory hematologic malignancies: Long-term follow-up of a prospective clinical trial. 48th Annual Meeting of the American Society of Hematology, 9–12 December 2006, Orlando, FL, USA.
  39. Handretinger R., Chen X., Lang P. et al. Feasibility and Outcome of Reduced-Intensity Conditioning in Haploidentical Transplantation. Ann. N.Y. Acad. Sci. 2007; 1106: 279–89.
  40. Federmann B., Handgretinger R., Bethge W.A. et al. Haploidentical allogeneic hematopoietic cell transplantation in adults using CD3/CD19 depletion and reduced intensity conditioning: a phase II study. Haematologica 2012; 97(10): 1523–31.
  41. Bader P., Koehl U., Klingebiel T. et al. Rapid immune recovery and low TRM in haploidentical stem cell transplantation in children and adolescence using CD3/CD19-depleted stem cells. Best Pract. Res. Clin. Haematol. 2011; 24: 331–7.
  42. Dufort G., Pisano S., Castillo L. et al. Feasibility and outcome of haploidentical SCT in pediatric high risk hematologic malignancies and Fanconianemia in Uruguay. Bone Marrow Transplant. 2012; 47(5): 663–8.
  43. Palma J., Handgretinger R., Rivera G.K. et al. Haploidentical stem cell transplantation for children with high-risk leukemia. Pediatr. Blood Cancer 2012; 59(5): 895–901.
  44. Gonzalez-Vicent M., Ramirez M., Diaz M.A. et al. Graft manipulation and reduced-intensity conditioning for allogeneic hematopoietic stem cell transplantation from mismatched unrelated and mismatched/haploidentical related donors in pediatric leukemia patients. J. Pediatr. Hematol. Oncol. 2010; 32(3): e85–90.
  45. Oevermann L., Lang P., Handgretinger R. et al. Immune reconstitution and strategies for rebuilding the immune system after haploidentical stem cell transplantation. Ann. N. Y. Acad. Sci. 2012; 1266: 161–70.
  46. Locatelli F., Vinti L., Moretta L. et al. Strategies to optimize the outcome of children given T-cell depleted HLA-haploidentical hematopoietic stem cell transplantation. Best Pract. Res. Clin. Haematol. 2011; 24(3): 339–49.
  47. Azevedo R.I., Soares M.V., Sousa A.E. et al. Long-term immune reconstitution of naive and memory T cell pools after haploidentical hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2013; 19(5): 703–12.
  48. Handgretinger R. Negative depletion of CD3(+) and TcRab(+) T cells. Curr. Opin. Hematol. 2012; 19(6): 434–9. 49. Bonneville M., O’Brien R.L., Born W.K. Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity. Nat. Rev. Immunol. 2010; 10: 467–78.
  49. Chiplunkar S., Dhar S., Wesch D., Kabelitz D. Gammadelta T cells in cancer immunotherapy: current status and future prospects. Immunotherapy 2009; 1: 663–78.
  50. Godder K.T., Henslee-Downey P.J., Mehta J. et al. Long term diseasefree survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation. Bone Marrow Transplant. 2007; 39: 751–7.
  51. Locatelli F., Bauquet A., Bertaina A. et al. Negative depletion of a/b+ T cells and of CD19+ B lymphocytes: A novel frontier to optimize the effect of innate immunity in HLA-mismatched hematopoietic stem cell transplantation. Immunol. Lett. 2013 Sep 30.
  52. Dodero A., Carniti C., Raganato A. et al. Haploidentical stem cell transplantation after a reduced-intensity conditioning regimen for the treatment of advanced hematologic malignancies: posttransplantation CD8-depleted donor lymphocyte infusions contribute to improve T-cell recovery. Blood 2009; 113: 4771–9.
  53. Amrolia P.J., Muccioli-Casadei G., Huls H. et al. Adoptive immunotherapy with allodepleted donor T-cells improves immune reconstitution after haploidentical stem cell transplantation. Blood 2006; 108: 1797–808.
  54. Mielke S., Nunes R., Rezvani K. et al. A clinical-scale selective allodepletion approach for the treatment of HLA-mismatched and matched donor- recipient pairs using expanded T lymphocytes as antigen-presenting cells and a TH9402-based photodepletion technique. Blood 2008; 111: 4392–402.
  55. Feuchtinger T., Matthes-Martin S., Richard C. et al. Safe adoptive transfer of virus-specific T-cell immunity for the treatment of systemic adenovirus infection after allogeneic stem cell transplantation. Br. J. Haematol. 2006; 134: 64–76.
  56. Feuchtinger T., Opherk K., Bethge W.A. et al. Adoptive transfer of pp65- specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood 2010; 116: 4360–7.
  57. Perruccio K., Tosti A., Burchielli E. et al. Transferring functional immune responses to pathogens after haploidentical hematopoietic transplantation. Blood 2005; 106: 4397–406.
  58. Lugthart G., Albon S.J., Ricciardelli I. et al. Simultaneous generation of multivirus-specific and regulatory T cells for adoptive immunotherapy. J. Immunother. 2012; 35: 42–53.
  59. Di I.M., Falzetti F., Carotti A. et al. Tregs prevent GVHD and promote immune reconstitution in HLA haploidentical transplantation. Blood 2011; 117(14): 3921–8.
  60. Brehm C., Huenecke S., Quaiser A. et al. IL-2 stimulated but not unstimulated NK cells induce selective disappearance of peripheral blood cells: concomitant results to a phase I/II study. PLoS One 2011; 6(11): e27351.
  61. Rizzieri D.A., Storms R., Chen D.F. et al. Natural killer cell-enriched donor lymphocyte infusions from A 3-6/6 HLA matched family member following nonmyeloablative allogeneic stem cell transplantation. Biol. Blood Marrow Transplant. 2010; 16: 1107–14.
  62. Passweg J.R., Tichelli A., Meyer-Monard S. et al. Purified donor NKlymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation. Leukemia 2004; 18(11): 1835–8.
  63. Rubnitz J.E., Inaba H., Ribeiro R.C. et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J. Clin. Oncol. 2010; 28(6): 955–9.
  64. Ji S.Q., Chen H.R., Xun C.Q. et al. G-CSF-primed haploidentical marrow transplantation without ex vivo T cell depletion: an excellent alternative for highrisk leukemia. Bone Marrow Transplant. 2002; 30(12): 861–6.
  65. Lu D.P., Dong L., Liu K.Y. et al. Conditioning including antithymocyte globulin followed by unmanipulated HLA-mismatched/haploidentical blood and marrow transplantation can achieve comparable outcomes with HLA-identical sibling transplantation. Blood 2006; 107(8): 3065–73. Epub 2005 Dec 27.
  66. Yabe H., Inoue H., Yabe M. et al. Unmanipulated HLA-haploidentical bone marrow transplantation for the treatment of fatal, nonmalignant diseases in children and adolescents. Int. J. Hematol. 2004; 80(1): 78–82.
  67. Ikegame K., Tanji Y., Ogawa H. et al. Successful treatment of refractory T-cell acute lymphoblastic leukemia by unmanipulated stem cell transplantation from an HLA 3-loci mismatched (haploidentical) sibling. Bone Marrow Transplant. 2003; 31(6): 507–10.
  68. Shimazaki C., Ochiai N., Nakagawa M. et al. Non-T-cell-depleted HLA haploidentical stem cell transplantation in advanced hematologic malignancies based on the feto-maternal michrochimerism. Blood 2003; 101(8): 3334–6.
  69. Huang X., Liu D., Zhang X. et al. Haploidentical hematopoietic stem cell transplantation without in vitro T cell depletion for treatment of hematologic malignancies in children. Biol. Blood Marrow Transplant. 2009; 15(1): 91–4.
  70. Mochizuki K., Kikuta A., Hosoya M. et al. Feasibility of tacrolimus, methotrexate, and prednisolone as a graft-versus-host disease prophylaxis in non-T-cell depleted haploidentical hematopoietic stem cell transplantation for children. Clin. Transplant. 2011; 25(6): 892–7.
  71. Субботина Н. Режимы кондиционирования со сниженной интенсивностью. Современный взгляд и собственный опыт применения в детской онкологии (обзор литературы). Дет. онкол. 2009; 3–4: 3–14. [Subbotina N. Reduced intensity conditioning regimens. Current view and own experience with usage in pediatric oncology (literature review). Det. onkol. 2009; 3–4: 3–14. (In Russ.)].
  72. Leung W., Handgretinger R., Pui C.H. et al. High success rate of hematopoietic cell transplantation regardless of donor source in children with very high-risk leukemia. Blood 2011; 118(2): 223–30.
  73. Grupp S.A., Kalos M., Barrett D. et al. Chimeric Antigen Receptor– Modified T Cells for Acute Lymphoid Leukemia. N. Engl. J. Med. 2013; 368(16): 1509–18.
  74. Tettamanti S., Marin V., Pizzitola I. et al. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor. Br. J. Haematol. 2013; 161(3): 389–401.

Successful allogeneic bone marrow transplantation in patients with severe gram-negative sepsis and septic shock

G.M. Galstyan, P.M. Makarova, L.A. Kuzmina, Ye.N. Parovichnikova, G.A. Klyasova, O.S. Pokrovskaya, M.Yu. Drokov, V.A. Novikov, V.V. Troitskaya, I.E. Kostina, and V.G. Savchenko

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


ABSTRACT

We present two cases of successful allogeneic bone marrow transplantation in the patients with severe gram-negative sepsis and septic shock. The features of the post-transplantation period and management of patients are described.


Keywords: allogeneic hematopoietic stem cell transplantation, conditioning, sepsis, septic shock, post-transplantation period, neutropenic enterocolitis, prolonged neutropenia, Pseudomonas aeruginosa, acute respiratory failure, invasive pulmonary aspergillosis, invasive lung ventilation, noninvasive ventilation, mesenchymal stromal cells.

Read in PDF (RUS) pdficon


REFERENCES

  1. Morena M.T., Gatti R.A. A History of Bone Marrow Transplantation. Hematol. Oncol. Clin. 2011; 25: 1–15.
  2. Wingard J.R., Majhail N.S., Brazauskas R. et al. Long-Term Survival and Late Deaths After Allogeneic Hematopoietic Cell Transplantation. J. Clin. Oncol. 2011; 29: 2230–9.
  3. Gyurkocza B., Rezvanil A., Storb R.F. Allogeneic hematopoietic cell transplantation: the state of the art. Exp. Rev. Hematol. 2010; 3(3): 285–99.
  4. Савченко В.Г., Любимова Л.С., Паровичникова Е.Н. и др. Трансплантации аллогенных и аутологичных гемопоэтических стволовых клеток при острых лейкозах (итоги 20-летнего опыта). Тер. арх. 2007; 79: 30–5. [Savchenko V.G., Lyubimova L.S., Parovichnikova Ye.N., et al. Transplantation of allogeneic and autologous hematopoietic stem cells in acute leukemias (summary of 20-year experience). Ter. arkh., 2007; 79: 30–5. (In Russ.)].
  5. Blume K.G., Forman S.J., Appelbaum F.R. The evaluation and counseling of candidates for hematopoietic cell transplantation. Thomas’ Hematopoietic Cell Transplantation, 3rd ed. Malden: Blackwell, 2004: 449–67.
  6. Ball E.D., Lister J., Law P. Evaluation of patients before hematopoietic stem cell transplantation. Hematopoietic Stem Cell Therapy. New York: Churchill Livingstone, 2001: 225–32.
  7. Majhail N.S., Rizzo J.D. Surviving the cure: long term follow up of hematopoietic cell transplant recipients. Bone Marrow Transplant. 2013: 1–7.
  8. Soubani A.O., Kseibi E., Bander J.J. et al. Outcome and Prognostic Factors of Hematopoietic Stem Cell Transplantation Recipients Admitted to a Medical ICU. Chest 2004; 126(5): 1604–11.
  9. Marena C., Zecca M., Carenini M.L. et al. Incidence of, and risk factors for, nosocomial infections among hematopoietic stem cell transplantation recipients, with impact on procedure-related mortality. Infect. Control Hosp. Epidemiol. 2001; 22: 510–7.
  10. Huynh T.N., Weigt S.S., Belperio J.A., Territo M., Keane M.P. Outcome and Prognostic Indicators of Patients with Hematopoietic Stem Cell Transplants Admitted to the Intensive Care Unit. J. Transplant. 2009; 2009: 917294.
  11. Afessa B., Azoulay E. Critical Care of the Hematopoietic Stem Cell Transplant Recipient. Crit. Care Clin. 2010; 26: 133–50.
  12. Degoricija V., Sharma M., Legac A. et al. Survival analysis of 314 episodes of sepsis in medical intensive care unit in university hospital: impact of intensive care unit performance and antimicrobial therapy. Croat. Med. J. 2006; 47(3): 385–97.
  13. Shirazi M.H., Ranjbar R., Ghasemi A. et al. A Survey of Bacterial Infections in Bone Marrow Transplant Recipients. Iran. J. Publ. Health 2007; 36: 77–81.
  14. Ali N., Adil S.M., Shaikh M.U., Moosajee M., Masood N. Outcome of match related allogeneic stem cell transplantation procedures performed from 2004 till 2011. Exper. Hematol. Oncol. 2012; 1: 13.
  15. George B., Mathews V., Srivastava A., Chandy M. Infections among allogeneic bone marrow transplant recipients in India. Bone Marrow Transplant. 2004; 33: 311–5.
  16. Donnelly P. Bacterial complications of transplantation: diagnosis and treatment. J. Antimicrob. Chemother. 1995; 36: 59–72.
  17. van Kraaij M.G., Dekker A.W., Verdonck L.F. et al. Infectious gastroenteritis: an uncommon cause of diarrhoea in adult allogeneic and autologous stem cell transplant recipients. Bone Marrow Transplant. 2000; 26(3): 299–303.
  18. Schulenburg A., Turetschek K., Wrba F. et al. Early and late gastrointestinal complications after myeloablative and nonmyeloablative allogeneic stem cell transplantation. Ann. Hematol. 2004; 83(2): 101–6.
  19. Holler E., Kolb H.J., Greinix H. et al. Bleeding events and mortality in SCT patients: a retrospective study of hematopoietic SCT patients with organ dysfunctions due to severe sepsis or GVHD. Bone Marrow Transplant. 2009; 43(6): 491–7.
  20. Mikulska M., Del Bono V., Bruzzi P. et al. Mortality after bloodstream infections in allogeneic haematopoietic stem cell transplant (HSCT) recipients. Infection 2012; 40: 271–8.
  21. Legrand M., Max A., Peigne V. et al. Survival in neutropenic patients with severe sepsis or septic shock. Crit. Care Med. 2012; 40(1): 43–9.
  22. Reikvam H., Hatfiel K.J., Kittang A.O., Hovland R., Bruserud O. Acute myeloid leukemia with the t(8;21) translocation: clinical consequences and biological implications. doi: 10.1155/2011/104631. Epub, May 3, 2011.
  23. Cho E.K., Bang S.M., Ahn J.Y. et al. Prognostic value of AML 1/ETO fusion transcripts in patients with acute myelogenous leukemia. Korean J. Intern. Med. 2003; 18(1): 13–20.
  24. Программное лечение заболеваний системы крови. Под ред. В.Г. Савченко. М.: Практика, 2012: 720–34. [Programmnoye lecheniye zabolevaniy sistemy krovi. Pod red. V.G. Sav chenko (Program therapy for hematological malignancies. Ed. by: V.G. Savchenko). M.: Praktika, 2012: 720–34.]
  25. De Pauw B., Walsh T.J., Donnelly J.P. et al. European Organization for Research and Treatment of Cancer/Invasive Fungal Infections Cooperative Group; National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Revised Definitions of Invasive Fungal Disease from the European Organization for Research and Treatment of Cancer/ Invasive Fungal Infections Cooperative Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus Group. Clin. Infect. Dis. 2008; 46(12): 1813–21.
  26. Afessa B., Tefferi A., Dunn W.F. et al. Intensive care unit support and Acute Physiology and Chronic Health Evaluation III performance in hematopoietic stem cell transplant recipients. Crit. Care Med. 2003; 31(6): 1715–21.
  27. Leung A.N., Gosselin M.V., Napper C.H. et al. Pulmonary Infections after Bone Marrow Transplantation: Clinical and Radiographic Findings. Radiology 1999; 210: 699–710.
  28. Champlin R.E., Perez W.S., Passweg J.R. et al. Bone marrow transplantation for severe aplastic anemia: a randomized controlled study of conditioning regimens. Blood 2007; 109(10): 4582–5.
  29. Georges G.E., Storb R. Stem cell transplantation for aplastic anemia. Int. J. Hematol. 2002; 75(2): 141–6.
  30. Quenot J.P., Binquet C., Kara F., Martinet O. The epidemiology of septic shock in French intensive care units: the prospective multicenter cohort EPISS study. Crit. Care. 2013; 17: R65.
  31. Jawad I., Luksic I., Rafnsson S.B. Assessing available information on the burden of sepsis: global estimates of incidence, prevalence and mortality. Glob. Health 2012; 2(1): 10404.
  32. Ghosh I., Raina V., Kumar L. et al. Profile of infections and outcome in high-risk febrile neutropenia: experience from a tertiary care cancer center in India. Med. Oncol. 2012; 29: 1354–60.
  33. Klastersky J., Ameye L., Maertens J. et al. Bacteraemia in febrile neutropenic cancer patients. Int. J. Antimicrob. Agents 2007; 30: 51–9.
  34. Sakamoto M., Saruta K., Nakazawa Y. et al. Sepsis Associated with Hematological Malignancies: Prophylaxis of Pseudomonas aeruginosa Sepsis. 69th General Meeting of the Japanese Association for Infectious Diseases (Fukuoka). Kansenshogaku Zasshi 1996; 70(2): 116–22.
  35. Клясова Г.А. Инфекции при гемобластозах и депрессиях кроветворения: клиника, диагностика и лечение: Автореф. дис. ¼ д-ра мед. наук. М., 2009. [Klyasova G.A. Infektsii pri gemoblastozakh i depressiyakh krovetvoreniya: klinika, diagnostika i lechenie. Dokt. diss. (Infections in hematological malignancies and depressed hematopoiesis. Dr. med. sci. diss.). M., 2009]
  36. Mokart D., Craenenbroeck T. Prognosis of acute respiratory distress syndrome (ARDS) in neutropenic cancer patients. Eur. Respir. J. 2012; 40(1): 169–76.
  37. Regazzoni C.J., Irrazabal C., Luna C.M., Poderoso J.J. Cancer patients with septic shock: mortality predictors and neutropenia. Supp. Care Cancer 2004; 12: 833–9.
  38. Groeger J.S., Lemeyow S., Price K., Nierman J. Multicenter outcome study of cancer patients admitted to the intensive care unit: a probability of mortality model. Clin. Oncol. 1998; 16: 761–70.
  39. Gronlykke L., Brandstrup S.L., Perner A. Data from clinical database on septic shock are valid. Dan. Med. J. 2012; 59(10): A4522.
  40. Горелов В.Г. Эффективность искусственной вентиляции легких при острой дыхательной недостаточности у больных гемобластозами: Автореф. дис. ¼ канд. мед. наук. М., 1994. [Gorelov V.G. Effektivnost iskusstvennoy ventilyatsii legkikh pri ostroy dykhatelnoy nedostatochnosti u bolnykh gemoblastozami. Kand. diss. (Efficacy of mechanical lung ventilation in acute respiratory failure in patients with hematological malignancies. Cand med. sci.diss.). M., 1994]
  41. Бычинин М.В., Галстян Г.М., Шулутко Е.М., Клясова Г.А., Городецкий В.М. Катетеризация артерий у гематологических больных. Гематол. и трансфузиол. 2013; 58: 14–22. [Bychinin M.V., Galstyan G.M., Shulutko Ye.M., Klyasova G.A., Gorodetzky V.M. Artery catheterization in hematological patients. Gematol. i transfuziol. 2013; 58: 14–22. (In Russ.)].
  42. Shirley H., Mei J. Mesenchymal Stem Cells Reduce Inflammation while Enhancing Bacterial Clearance and Improving Survival in Sepsis. Am. J. Respir. Crit. Care Med. 2010; 182(8): 1047–57.
  43. Gilbert C., Vasu T.S., Baram M. Use of mechanical ventilation and renal replacement therapy in critically ill hematopoietic stem cell transplant recipients. Biol. Blood Marrow Transplant. 2013; 19(2): 321–4.
  44. Azoulay E., Alberti C., Bornstain C. et al. Improved survival in cancer patients requiring mechanical ventilatory support: impact of noninvasive mechanical ventilatory support. Crit. Care Med. 2001; 29(3): 519–25.
  45. Avivi I., Oren I., Haddad N., Rowe J.M. Stem Cell Transplantation Post Invasive Fungal Infection Is a Feasible Task. Am. J. Hematol. 2004; 75: 6–11.
  46. Bjerke J.W., Meyers J.D., Bowden R.A. Hepatosplenic candidiasis — a contraindication to marrow transplantation? Blood 1994; 84: 2811–4.
  47. Wang J.T., Yao M., Tang J.L., Chang S.C., Hung C.C. Prior invasive fungal infection is not a contraindication for subsequent allogeneic bone marrow transplantation in adult patients with hematologic malignancies. J. Clin. Oncol. 2001; 19(1): 4000–1.
  48. Aki Z.S., Sucak G.T., Yegin Z.A. et al. Hematopoietic Stem Cell Transplantation in Patients With Active Fungal Infection: Not a Contraindication for Transplantation. Transplant. Proceed. 2008; 40: 1579–85.
  49. El-Cheikh J., Castagna L., Wang L. et al. Impact of prior invasive aspergillosis on outcome in patients receiving reduced-intensity conditioning allogeneic hematopoietic stem cell transplant. Leuk. Lymphoma 2010; 51(9): 1705–10.
  50. Lee J.Y., Jung C.W., Kim K., Jang J.H. Impact of previous invasive pulmonary aspergillosis on the outcome of allogeneic hematopoietic stem cell transplantation. Korean J. Hematol. 2012; 47(4): 255–9.
  51. Dellinger R.P., Levy M.M., Rhodes A. et al. Surviving Sepsis Campaign Guidelines Committee including The Pediatric Subgroup. Surviving Sepsis Campaign: International Guidelines for Management of Severe Sepsis and Septic Shock, 2012. Intens. Care Med. 2013; 39(2): 165–228.
  52. Pittenger M.F., Mackay A.M., Beck S.C. et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999; 284: 143–7.
  53. Deans R.J., Moseley A.B. Mesenchymal stem cells: biology and potential clinical uses. Exp. Hematol. 2000; 28: 875–84.
  54. Jones B.J., McTaggart S.J. Immunosuppression by mesenchymal stromal cells: from culture to clinic. Exp. Hematol. 2008; 36: 733–41.
  55. Петинати Н.А. Профилактика реакции трансплантат против хозяина у больных гемобластозами после трансплантации аллогенных гемопоэтических стволовых клеток с помощью мультипотентных мезенхимальных стромальных клеток донора: Автореф. дис. ¼ канд. мед. наук. М., 2013. [Petinati N.A. Profilaktika reaktsii transplantat protiv khozyaina u bolnykh gemoblastozami posle transplantatsii allogennykh gemopoeticheskikh stvolovykh kletok s pomoshchyu multipotentnykh mezenkhimalnykh stromalnykh kletok donora. Kand. diss. (Prevetion of graft-versus-host disease in patients with hematological malignancies after transplantation of allogeneic hematopoietic stem cells using multipotent mesenchymal stromal donor cells. Cand med. sci. diss.). M., 2013].
  56. Kuzmina L.A., Petinati N.A., Parovichnikova E.N. et al. Multipotent Mesenchymal Stromal Cells for the Prophylaxis of Acute Graft-versus-Host Disease — A Phase II Study. Stem Cells Int. 2012; 2012: 968213.
  57. Kebriaei P., Robinson S. Treatment of graft-versus-host-disease with mesenchymal stromal cells. Cytotherapy 2011; 13(3): 262–8.
  58. Lucchini G., Dander E., Rovelli A. et al. Platelet-lysate-expanded mesenchymal stromal cells as a salvage therapy for severe resistant graftversus-host disease in a pediatric population. Biol. Blood Marrow Transplant. 2010; 16: 1293–301.
  59. Toubai Т., Paczesny S., Shono Y. et al. Mesenchymal stem cells for treatment and prevention of graft-versus-host disease after allogeneic hematopoietic cell transplantation. Curr. Stem Cell Res. Ther. 2009; 4: 252–9.
  60. Osuchowski M.F., Welch K., Siddiqui J., Remick D.G. Circulating cytokine/inhibitor profiles reshape the understanding of the sirs/cars continuum in sepsis and predict mortality. J. Immunol. 2006; 177: 1967–74.
  61. Chien M.H., Bien M.Y., Ku C.C. et al. Systemic human orbital fat-derived stem/stromal cell transplantation ameliorates acute inflammation in lipopolysaccharide-induced acute lung injury. Crit. Care Med. 2012; 40(4): 1245–53.
  62. Kim E.S., Sil Y. Intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells attenuates Escherichia coli induced acute lung injury in mice. Respir. Res. 2011; 12: 108.
  63. Adult stem cell transplantation in severe blood poisoning. 2011-MSC-1 Erasmus MC Rotterdam. http://www.trialregister.nl/ 64. OHRI, Ottawa to lead first stem cell therapy trial for septic shock/2012. http://nationalhealthwatch.ca/

Stable donor hematopoiesis reconstitution after post-transplantation relapse of acute myeloid leukemia in patient with inv(3)(q21q26), –7 and EVI1 oncogene overexpression treated by donor lymphocyte infusions and hypomethylating agents

N.N. Mamaev, A.V. Gorbunova, T.L. Gindina, O.A. Slesarchuk, V.N. Ovechkina, S.N. Bondarenko, O.V. Goloshchapov, V.M. Kravtsova, and B.V. Afanasev

I.P. Pavlov Saint Petersburg State Medical University, R.M. Gorbacheva Institute of Pediatric Oncology, Hematology and Transplantology, Saint Petersburg, Russian Federation


ABSTRACT

We present the case of successful treatment of post-transplantation relapse of prognostically unfavorable AML with inv(3)(q21q26), –7 and EVI1 oncogene overexpression, when stable donor hematopoiesis reconstitution was achieved due to one high-dose cytarabine course, DLI, and hypomethylating agents (decitabine, 5-azacitidine). Possible molecular mechanisms of this effect are discussed with respect to the new approaches to management of such patients.

Keywords: acute myeloid leukemia, inv(3)(q21q26), EVI1 high expression, hematopoietic stem cell transplantation, relapse, treatment, donor lymphocyte infusions, hypomethylating agents.

Read in PDF (RUS)pdficon


REFERENCES

  1. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миелодис- пластические синдромы с высокой экспрессией гена EVI1: теоретические и клинические аспекты. Клин. онкогематол. 2012; 5(4): 361–4.[Mamayev N.N., Gorbunova A.V., Gindina T.L. et al. Leukemias and myelodisplastic syndromes with high EVI1 gene expression: theoretical and clinical aspects. Klin. onkogematol. 2012; 5(4): 361–4. (In Russ.)].
  2. Barrett A.J., Battiwalla M. Relapse after allogeneic stem cell transplantation. Expert. Rev. Hematol. 2010; 3(4): 429–41.
  3. Arellano M.L., Langston A., Winton E. et al. Treatment of relapsed acute leukemia after allogeneic transplantation: a single center experience. Biol. Blood Marrow Transplant. 2007; 13(1): 116–23.
  4. Porter D.L., Alyea E.P., Antin J.H. et al. NCI First International Workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: Report from the Committee on Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2010; 16: 1467–503.
  5. Pavletic S.Z., Kumar S., Mohty M. et al. NCI First International Workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: Report from the Committee on the Epidemiology and Natural History of Relapse following Allogeneic Cell Transplantation. Biol. Blood Marrow Transplant. 2010; 16: 871–90.
  6. Wang Y., Liu D.-H., Fan Z.-P. et al. Prevention of relapse using DLI can increase survival following HLA-identical transplantation in patients with advanced-stage acute leukemia: a multi-center study. Clin. Transplant. 2012. doi: 10.111/j.1399-0012.2012.01626.x.
  7. Lubbert M., Bertz H., Wasch R. et al. Efficacy of a 3-day, low-dose treatment with 5-azacytidine followed by donor lymphocyte infusions in older patients with acute myeloid leukemia or chronic myelomonocytic leukemia relapsed after allografting. Bone Marrow Transplant. 2009; 45(4): 627–32.
  8. Craddock C., Quek L., Goardon N. et al. Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia. Leukemia 2012; doi: 10.1038/leu.2012.312.
  9. Candoni A., Tiribelli M., Toffoletti E. et al. Quantitative assessment of WT1 gene expression after allogeneic stem cell transplantation is a useful tool for monitoring minimal residual disease in acute myeloid leukemia. Eur. J. Haematol. 2009; 82(1): 61–8.
  10. Zhao X.-S., Jin S., Zhu H.-H. et al. Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2011. doi:10.1038/bmt.2011.121.

Impact of molecular genetic and cytogenetic characteristics on outcomes of allogeneic hematopoietic stem cell transplantation in chronic myeloid leukemia

A.V. Gorbunova, T.L. Gindina, E V. Morozova, I.M. Barkhatov, N.N. Mamayev, and B.V. Afanasyev

R.M. Gorbacheva Institute of Pediatric Oncology, Hematology and Transplantology, I.P. Pavlov State Medical University, Saint Petersburg, Russian Federation


ABSTRACT

Point mutations in the BCR-ABL kinase domain, BCR-ABL and EVI1 gene expression alterations, and additional chromosomal aberrations in Philadelphia chromosome-positive chronic myeloid leukemia are strongly associated with resistance to tyrosine kinase inhibitors (TKIs) and disease progression, but their effect on the outcome of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is uncertain. This retrospective study included 35 CML patients with resistance to TKI therapy who received a related or unrelated HSCT. Additional chromosomal aberrations were associated with the decreased rate of the complete molecular response (CMR) after allo-HSCT. EVI1 expression level was associated with a decreased disease-free survival (DFS). BCR-ABL kinase domain mutations showed no influence on CMR, OS, and DFS in this patient cohort. 9 out of 10 patients with T315I mutation achieved CMR. EVI1-directed stratification of patients during the post-transplantation period may improve outcome of HSCT.


Keywords: chronic myeloid leukemia, CML, allogeneic hematopoietic stem cells transplantation, allo-HSCT, BCR-ABL, EVI1.

Read in PDF (RUS)pdficon


Refernces

  1. Oyekunle A., Klyuchnikov E., Ocheni S. et al. Challenges for allogeneic hematopoietic stem cell transplantation in chronic myeloid leukemia in the era of tyrosine kinase inhibitors. Acta Haematol. 2011; 126(1): 30–9.
  2. Baccarani M., Cortes J., Pane F. et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. Clin. Oncol. 2009; 27(35): 6041–51.
  3. Soverini S., Hochhaus A., Nicolini F.E. et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011; 118(5): 1208–15.
  4. Cortes J.E., Kantarjian H., Shah N.P. et al. Ponatinib in refractory Philadelphia chromosome-positive leukemias. Engl. J. Med. 2012; 367(22): 2075–88.
  5. Hochhaus A., Kreil S., Corbin A.S. et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia 2002; 16: 2190–6.
  6. Wang Y., Cai D., Brendel C. et al. Adaptive secretion of granulocytemacrophage colony-stimulating factor (GM-CSF) mediates imatinib and nilotinib resistance in BCR/ABL+ progenitors via JAK-2/STAT-5 pathway activation. Blood 2007; 109: 2147–55.
  7. Chu S., Holtz M., Gupta M. et al. BCR/ABL kinase inhibition by imatinib mesylate enhances MAP kinase activity in chronic myelogenous leukemia CD34+ cells. Blood 2004; 103: 3167–74.
  8. Burchert A., Wang Y., Cai D. et al. Compensatory PI3-kinase/Akt/mTOR activation regulates imatinib resistance development. Leukemia 2005; 19: 1774–82.
  9. Daghistani M., Marin D., Khorashad J.S. et al. EVI-1 oncogene expression predicts survival in chronic-phase CML patients resistant to imatinib treated with second-generation tyrosine kinase inhibitors. Blood 2010; 116(26): 6014–7.
  10. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миело- диспластические синдромы с экспрессией гена EVI1: теоретические и клинические аспекты. Клин. онкогематол. 2012; 5(4): 361–4. [Mamayev N.N., Gorbunova A.V., Gindina T.L. i dr. Leykozy i miyelodis_ plasticheskiye sindromy s vysokoy ekspressiyey gena EVI1: teoreticheskiye i klinicheskiye aspekty (Leukemias and myelodisplastic syndromes with high EVI1 gene expression: theoretical and clinical aspects. In: Clin. oncohematol.). Klin. onkogematol. 2012; 5(4): 361–4.]
  11. Groschel S., Lugthart S., Schlenk R.F. et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. Clin. Oncol. 2010; 28(12): 2101–7.

 

Hematopoietic stem cell transplantation in AML patients with t(8;21)(q22;q22) translocation

N.N. Mamayev, A.V. Gorbunova, T.L. Gindina, I.M. Barkhatov, S.N. Bondarenko, M.Yu. Averyanova, О.V. Pirogova, O.V. Goloshchapov, Ye.V. Kondakova, and B.V. Afanasyev

R.M. Gorbacheva Institute of Pediatric Oncology, Hematology and Transplantology, I.P. Pavlov State Medical University, Saint Petersburg, Russian Federation


ABSTRACT

The outcomes of bone marrow transplantation for treatment of relapses in 7 AML patients with t(8;21)(q22;q22) translocation are presented and analyzed. Two of them were transplanted in the 1st remission, and 5 patients received HSCT during resistance to the therapy. Three patients underwent unrelated allo-HSCT with various sources of HSC. Three others were treated with related allo-, auto-, or haplo-HSCT, respectively. In the last patient, auto-HSCT followed by related haplo-HSCT was performed. The course of disease was monitored using the serial levels of both AML1-ETO and WT1 gene expression. The high AML1-ETO levels and t(8;21) translocation were detected in all studied patients, whereas no FLT3 gene mutations were found in any patients, and a classic V617F JAK2 mutation was present in 1 patient. The levels of AML1-ETO and WT1 gene expression decreased in parallel with the relapse reduction in 2 patients, but remained elevated in 3 other patients despite the normalization of bone marrow morphologic picture, including 2 cases of development of extramedullary AML relapses. Relapses were accompanied by the high levels of the above gene expression. The study led to the conclusion that bone marrow transplantation is indicated for some AML patients with t(8;21) translocation. The treatment efficacy can be monitored using serial measurements of WT1 gene expression levels.


Keywords: AML with t(8;21) translocation, bone marrow transplantation, AML1-ETO and WT1 gene expression monitoring, molecular monitoring during treatment of leukemia.

Read in PDF (RUS) pdficon


Refernces

  1. Zander A.R., Bacher U., Finke J. Allogeneic stem cell transplantation in acute myeloid leukemia establishment of indications on the basis of individual risk stratification. Arztebl. Int. 2008; 105(39): 663–9.
  2. Numata A., Fujimaki K., Aoshima T. et al. Retrospective analysis of treatment outcomes in 70 patients with t(8;21) acute myeloid leukemia. Rinsho Ketsueki 2012; 53(7): 698–704.
  3. Kawamura M., Kaku H., Ito T. et al. FLT3-internal tandem duplication in a pediatric patient with t(8;21) acute myeloid leukemia. Cancer Genet. Cytogenet. 2010; 203(2): 292–6.
  4. Chen Y.M., Liu T.F., Ruan M. et al. Prognosis and chromosomal abnormalities in 79 children with t(8;21) acute myeloid leukemia. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2009; 31(5): 542–6.
  5. Mamaev N., Mamaeva S. Two cases of acute myeloblastic leukemia (M2 type) with karyotypes 45X,-X,t(6;8)(q27;q22),inv(9) and 46,XY,t(8;21) (q22;q22),del(9)(q22). Cancer Genet. Cytogenet. 1985; 18(2): 105–11.
  6. Мамаев Н.Н., Горбунова А.В., Гиндина Т.Л. и др. Лейкозы и миело- диспластические синдромы с высокой экспрессией гена EVI1: теоретиче- ские и клинические аспекты. Клин. онкогематол. 2012; 5(4): 361–4. [Mamayev N.N., Gorbunova A.V., Gindina T.L. i dr. Leykozy i miyelodisplasticheskiye sindromy s vysokoy ekspressiyey gena EVI1: teoreticheskiye i klinicheskiye aspekty (Leukemias and myelodisplastic syndromes with high EVI1 gene expression: theoretical and clinical aspects. In: Clin. oncohematol.). Klin. onkogematol. 2012; 5(4): 361–4.]
  7. Forman S.J., Rowe J.M. The myth of the second remission of acute leukemia in the adult. Blood 2013; 121(7): 1077–82.
  8. Kuwatsuka Y., Miyamura K., Suzuki R. et al. Hematopoietic stem cell transplantation for core binding factor acute myeloid leukemia: t(8;21) and inv(16) represent different clinical outcomes. Blood 2009; 113: 2096–103.
  9. Zhao X.S., Jin S., Zhu H.H. et al. Wilms’ tumor gene 1 expression: an independent acute leukemia prognostic indicator following allogeneic hematopoietic SCT. Bone Marrow Transplant. 2012; 47(4): 499–507.
  10. Candoni A., Toffoletti E., Gallina R. et al. Monitoring of minimal residual disease by quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem cell transplantation in acute myeloid leukemia. Clin. Transplant. 2011; 25: 308–16.
  11. Lange T., Hubmann M., Burkhardt R. et al. Monitoring of WT1 expression in PB and CD34+ donor chimerism of BM predicts early relapse in AML and MDS patients after hematopoietic cell transplantation with reduced-intensity conditioning. Leukemia 2011; 25: 498–505.
  12. Kwon M., Martinez-Laperche C., Infante M. et al. Evaluation of minimal residual disease by real-time quantitative PCR of Wilms’ Tumor 1 expression in patients with acute myelogenous leukemia after allogeneic stem cell transplantation: Correlation with flow cytometry and chimerism. Biol. Blood Marrow Transplant. 2012; 18: 1235–42.

Mixed chimerism following allogeneic bone marrow transplantation: cases report

K.N. Melkova, N.V. Gorbunova, T.Z. Cherniavskaya

FSBI «N.N. Blokhin Russian Cancer Research Center» RAMS, Moscow, Russian Federation


ABSTRACT

Cimerism monitoring after the bone marrow transplantation (BMT) by a method based on the quantitative polymerase chain reaction (PCR), is important for the assessment of efficiency of transplantation, early identification of recurrence and timely correction of therapy. Clinical examples of the mixed chimerism after allogeneic BMT are presented.


Keywords: allogeneic transplantation, chimerism, mixed chimerism, bone marrow, hematopoietic stem cells.

Read in PDF (RUS)pdficon


REFERENCES

  1. Appelbaum F.R., Forman S. J., Negrin R.S., Blume K.G. Thomas’ Hematopoietic Cell Transplantation, 4th ed. Malden: Blackwell Publishing Ltd., 2009: 316–27.
  2. Blum W., Brown R., Lin H.-S. et al. Low-dose (550 cGy), single-exposure total body irradiation and cyclophosphamide: Consistent, durable engraftment of related-donor peripheral blood stem cells with low treatment-related mortality and fatal organ toxicity. Biol. Blood Marrow Transplant. 2002; 8(11): 608–18.
  3. Tutschka P.J., Copelan E.A., Klein J.P. Bone Marrow transplantation for leukemia following a new busulfan and cyclophosphamide regimen. Blood 1987; 70(5): 1382–8.
  4. Munker R., Lasarus H.M., Atkinson K. The BMT Data Book, 2nd ed. Cambridge University Press, 2009: 331–56.
  5. Matutes E., Pickl W.F., van’t Veer M. et al. Mixed-phenotype acute leukemia: clinical and laboratory features and outcome in 100 patients defined according to the WHO 2008 classification. Blood 2011; 117: 3163–71.
  6. Румянцев А.Г., Масчан А.А. Трансплантация гемопоэтических стволовых клеток у детей. М.: МИА, 2003: 405. [Rumyantsev A.G., Maschan A.A. Transplantatsiya gemopoeticheskikh stvolovykh kletok u detei (Hematopoietic stem cell transplantation in children). M.: MIA, 2003: 405.]

Chimerism following allogeneic hematopoietic stem cell transplantation

O.V. Blau

Charité University School of Medicine, Department of Hematology and Oncology, Berlin, Germany


ABSTRACT

Allogenic hematopoietic stem cell transplantation (HSCT) is a potentially curative treatment for various hematological disorders. Different molecular genetics methods are useful to monitor engraftment, relapse of the underlying disease, rejection of the graft, and minimal residual disease. Cytogenetic investigations, fluorescence in situ hybridization, PCR on chromosome abnormalities, lymphocyte-receptor gene rearrangement, and chimerism analysis are widely employed, although they are characterized by different sensitivity. Quantitative analysis of chimerism after HSCT is an important method for monitoring engraftment and allows discrimination between graft failure and relapse. This method appears to be adequate after reduced intensity conditioning (RIC) HSCT, when graft versus leukemia effect plays an important role in leukemia eradication. The use of lineage-specific chimerism analysis makes chimerism study more informative. Stable long-term complete chimerism correlates with complete hematological remission. Mixed chimerism is usually associated with relapse or graft failure. Prolonged mixed chimerism is a phenomenon in some patients after RIC with chronic lymphoproliferative disorders. This may be important to understand graft versus leukemia effect in such patients. Serial chimerism analysis is a suitable tool to evaluate efficacy of transplantation and early identification of relapse.


Keywords: allogenic hematopoietic stem cell transplantation, chimerism.

Read in PDF (RUS) pdficon


REFERENCES

  1. McCann S., Lawler M. Mixed chimerism; detection and significance following BMT. Bone Marrow Translant. 1993; 11: 91–4.
  2. Tippet P. Blood group chimeras: A review. Vox Sang. 1983; 44: 333–59.
  3. Ford C., Hamerton J., Barnes D., Loutit J. Cytological identification of radiation-chimeras. Nature 1956; 177: 452–4.
  4. Santos G., Sensenbrener P., Burke P. et al. The use of cyclophosphamide for clinical marrow transplantation. Transplant. Proc. 1972; 4: 559–64.
  5. Fehse B., Chukhlovin A., Kuhlcke K. et al. Real-time quantitative Y chromosome-specific PCR (QYCS-PCR) for monitoring hematopoietic chimerism after sex-mismatched allogeneic stem cell transplantation. J. Hematother. Stem Cell Res. 2001; 10(3): 419–25.
  6. Mattsson J. Molecular monitoring of engraftment and leukaemia relapse after allogeneic haematopoietic stem cell transplantation. Stockholm, 2001.
  7. Bader P., Niethammer D., Willasch A., Kreyenberg H., Klingebiel T. How and when should we monitor chimerism after allogeneic stem cell transplantation? Bone Marrow Transplant. 2005; 35(2): 107–19.
  8. Blazar B., Orr H., Arthur D. Restriction fragment length polymorphisms as markers of engraftment in allogeneic marrow transplantation. Blood 1985; 66: 1436–44.
  9. Min G., Hibbin J., Arthur C. et al. Use of minisatellite DNA probes for recognition and characterization of relapse after allogeneic bone marrow transplantation. Br. J. Haematol. 1988; 68: 195–201.
  10. Socie G., Lawler M., Gluckman E. et al. Studies on hematopoietic chimerism following allogeneic bone marrow transplantation in the molecular biology era. Leuk. Res. 1995; 19: 497–504.
  11. Thiede C., Bornhaeuser M., Oesschlagel U. et al. Sequential monitoring of chimerism and detection of minimal residual disease after allogeneic blood stem cell transplantation (BSCT) using multiplex PCR amplification of short tandem repeat markers. Leukemia 2001; 7: 958–65.
  12. Thiede C., Bornhauser M., Ehninger G. Strategies and clinical implications of chimerism diagnostics after allogeneic hematopoietic stem cell transplantation. Acta Haematol. 2004; 112: 16–23.
  13. Lion T., Daxberger H., Dubovsky J. et al. Analysis of chimerism within specific leukocyte subsets for detection of residual or recurrent leukemia in pediatric patients after allogeneic stem cell transplantation. Leukemia 2001; 15: 307–10.
  14. Craig-Holmes A., Shaw M. Polymorphism of human constitutive heterochromatin. Science 1971; 174: 702–4.
  15. Weber J., May P. Abundant class of human DNA polymorphisms which can be typed using the polymerase chain reaction. Am. J. Hum. Genet. 1989; 44: 388–96.
  16. Jeffreys A., Wilson V., Neumann R. et al. Amplification of human minisatellites by the polymerase chain reaction: towards DNA fingerprinting of single cells. Nucleic Acids Res. 1988; 16: 10953–71.
  17. Kreyenberg H., Holle W., Mohrle S. et al. Quantitative analysis of chimerism after allogeneic stem cell transplantation by PCR amplification of microsatellite markers and capillary electrophoresis with fluorescence detection: the Tuebingen experience. Leukemia 2003; 17: 237–40.
  18. Blau I.W., Schmidt-Hieber M., Leschinger N. et al. Engraftment kinetics and hematopoietic chimerism after reduced-intensity conditioning with fludarabine and treosulfan before allogeneic stem cell transplantation. Ann. Hematol. 2007; 86(8): 583–9.
  19. Jaksch M., Mattsson J., Uzunel M. et al. Multi-lineage mixed chimerism is common in patients with metabolic disorders after allogeneic stem cell transplantation. Bone Marrow Transplant. 2001; 27: 196.
  20. Matthes-Martin S., Lion T., Haas O.A. et al. Lineage-specific chimerism after stem cell transplantation in children following reduced intensity conditioning: potential predictive value of NK cell chimerism for late graft rejection. Leukemia 2003; 17(10): 1934–42.
  21. Baron F., Baker J.E., Storb R. et al. Kinetics of engraftment in patients with hematologic malignancies given allogeneic hematopoietic cell transplantation after non-myeloablative conditioning. Blood 2004; 104(8): 2254–62.
  22. Mohty M., Avinens O., Faucher C. et al. Predictive factors and impact of full donor T-cell chimerism after reduced intensity conditioning allogeneic stem cell transplantation. Haematologica 2007; 92(7): 1004–6.
  23. Lawler M., McCann S.R., Marsh J.C. et al. Serial chimerism analyses indicate that mixed haemopoietic chimerism influences the probability of graft rejection and disease recurrence following allogeneic stem cell transplantation (SCT) for severe aplastic anaemia (SAA): indications for routine assessment of chimerism post SCT for SAA: Severe Aplastic Working Party of the European blood and Marrow Transplant group. Br. J. Haematol. 2009; 144: 933–45