Myeloid-Derived Suppressor Cells in Some Oncohematological Diseases

AV Ponomarev

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

For correspondence: Aleksandr Vasil’evich Ponomarev, graduate student, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; e-mail: kl8546@yandex.ru

For citation: Ponomarev AV. Myeloid-Derived Suppressor Cells in Some Oncohematological Diseases. Clinical oncohematology. 2017;10(1):29-38–хх (In Russ).

DOI: 10.21320/2500-2139-2017-10-1-29-38


ABSTRACT

Myeloid-derived suppressor cells are immature myeloid cells with immunosuppressive properties. The review presents characteristics of myeloid-derived suppressor cells. It includes phenotype variants, mechanisms of the suppressive effect on the immune system, and tumor recruitment mechanisms of myeloid suppressors. It provides a brief description of works which studied myeloid suppressor in oncohematological diseases including multiple myeloma, lymphomas, and leukemias.

Keywords: myeloid suppressors, myeloid-derived suppressor cells, multiple myeloma, lymphomas, leukemias.

Received: September 8, 2016

Accepted: December 3, 2016

Read in PDF (RUS)pdficon


REFERENCES

  1. Тупицына Д.Н., Ковригина А.М., Тумян Г.С. и др. Клиническое значение внутриопухолевых FOXP3+ Т-регуляторных клеток при солидных опухолях и фолликулярных лимфомах: обзор литературы и собственные данные. Клиническая онкогематология. 2012;(5)3:193–203.
    [Tupitsyna DN, Kovrigina AM, Tumian GS, et al. Different clinical meaning of intratumoral FOXP3+ T-regulatory cells in solid tumors and follicular lymphomas: literature review and own data. Klinicheskaya onkogematologiya. 2012;(5)3:193–203. (In Russ)]
  2. Кадагидзе З.Г., Черткова А.И., Славина Е.Г. NKT-клетки и противоопухолевый иммунитет. Российский биотерапевтический журнал. 2011;10(3):9–16.
    [Kadagidze ZG, Chertkova AI, Slavina EG. NKT-cells and antitumor immunity. Rossiiskii bioterapevticheskii zhurnal. 2011;10(3):9–16. (In Russ)]
  3. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev 2012;12(4):253–68. doi: 10.1038/nri3175.
  4. Gabrilovich DI, Bronte V, Chen S-H, et al. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007;67(1):425– doi: 10.1158/0008-5472.CAN-06-3037.
  5. Bowen JL, Olson JK. Innate immune CD11b+Gr-1+ cells, suppressor cells, affect the immune response during Theiler’s virus-induced demyelinating disease. J Immunol. 2009;183(11):6971–80. doi: 10.4049/jimmunol.0902193.
  6. Tsiganov EN, Verbina EM, Radaeva TV, et al. Gr-1dim CD11b+ immature myeloid-derived suppressor cells but not neutrophils are markers of lethal tuberculosis infection in mice. J Immunol. 2014;192(10):4718–27. doi: 10.4049/jimmunol.1301365.
  7. Delano MJ, Scumpia PO, Weinstein JS, et al. MyD88-dependent expansion of an immature GR-1(+)CD11b(+) population induces T cell suppression and Th2 polarization in sepsis. J Exp Med. 2007;204(6):1463–74.
  8. Гапонов М.А., Хайдуков С.В., Писарев В.М. и др. Субпопуляционная гетерогенность миелоидных иммуносупрессорных клеток у пациентов с септическими состояниями. Российский иммунологический журнал. 2015;9(18):11–14.
    [Gaponov MA, Khaidukov SV, Pisarev VM, et al. Subpopulation heterogeneity of immunosuppressive myeloid cells in patients with sepsis. Rossiiskii immunologicheskii zhurnal. 2015;9(18):11–14. (In Russ)]
  9. Makarenkova VP, Bansal V, Matta BM, et al. CD11b+/Gr-1+ myeloid suppressor cells cause T cell dysfunction after traumatic stress. J Immunol. 2006;176(4):2085–94. doi: 10.4049/jimmunol.176.4.2085.
  10. Greten TF, Manns MP, Korangy F. Myeloid derived suppressor cells in human diseases. Int. 2011;11(7):802–7. doi: 10.1016/j.intimp.2011.01.003.
  11. Diaz-Montero CM, Salem ML, Nishimura MI, et al. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin–cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58(1):49–59. doi: 10.1007/s00262-008-0523-
  12. Yazdani Y, Mohammadnia-Afrouzi M, Yousefi M, et al. Myeloid-derived suppressor cells in B cell malignancies. Tumour Biol. 2015;36(10):7339–53. doi: 10.1007/s13277-015-4004-z.
  13. Пономарев А.В. Миелоидные супрессорные клетки: общая характеристика. Иммунология. 2016;37(1):47–50. doi: 10.18821/0206-4952-2016-37-1-47-50.
    [Ponomarev AV. Myeloid suppressor cells: general characteristics. Immunologiya. 2016;37(1):47–50. doi: 10.18821/0206-4952-2016-37-1-47- (In Russ)]
  14. Gabrilovich DI, Nagaraj S. Myeloid-derived-suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–74. doi: 10.1038/nri2506.
  15. Lechner MG, Megiel C, Russell SM, et al. Functional characterization of human Cd33+ And Cd11b+ myeloid-derived suppressor cell subsets induced from peripheral blood mononuclear cells co-cultured with a diverse set of human tumor cell lines. J Transl 2011;9(1):90. doi: 10.1186/1479-5876-9-90.
  16. Rodriguez PC, Ernstoff MS, Hernandez C, et al. Arginase I–Producing Myeloid-Derived Suppressor Cells in Renal Cell Carcinoma Are a Subpopulation of Activated Granulocytes. Cancer Res. 2009;69(4):1553–60.
  17. Schmielau J, Finn OJ. Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of T-cell function in advanced cancer patients. Cancer Res. 2001;61(12):4756–60.
  18. Youn J-I, Collazo M, Shalova I, et al. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J Leuk 2012;91(1):167–81. doi: 10.1189/jlb.0311177.
  19. Youn J-I, Nagaraj S, Collazo M, et al. Subsets of Myeloid-Derived Suppressor Cells in Tumor Bearing Mice. J Immunol. 2008;181(8):5791–802. doi: 10.4049/jimmunol.181.8.5791.
  20. Corzo CA, Condamine T, Lu L, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439–53. doi: 10.1084/jem.20100587.
  21. Yang L, DeBusk LM, Fukuda K, et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004;6(4):409–21. doi: 10.1016/j.ccr.2004.08.031.
  22. Zhuang J, Zhang J, Lwin ST, et al. Osteoclasts in multiple myeloma are derived from Gr-1+CD11b+ myeloid-derived suppressor cells. PLoS One. 2012;7(11):e48871. doi: 1371/journal.pone.0048871.
  23. Choi J, Suh B, Ahn YO, et al. CD15+/CD16low human granulocytes from terminal cancer patients: granulocytic myeloid-derived suppressor cells that have suppressive function. Tumour Biol. 2012;33(1):121–9. doi: 10.1007/s13277-011-0254-
  24. Stanojevic I, Miller K, Kandolf-Sekulovic L, et al. A subpopulation that may correspond to granulocytic myeloid-derived suppressor cells reflects the clinical stage and progression of cutaneous melanoma. Int Immunol. 2016;28(2):87–97. doi: 10.1093/intimm/dxv053.
  25. Saiwai H, Kumamaru H, Ohkawa Y, et al. Ly6C+Ly6G– Myeloid-derived suppressor cells play a critical role in the resolution of acute inflammation and the subsequent tissue repair process after spinal cord injury. J Neurochem. 2013;125(1):74–88. doi: 10.1111/jnc.12135.
  26. Rodriguez PC, Augusto CO. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol 2008;222(1):180–91. doi: 10.1111/j.1600-065X.2008.00608.x.
  27. Srivastava MK, Sinha P, Clements VK, et al. Myeloid-derived suppressor cells inhibit T cell activation by depleting cystine and cysteine. Cancer Res. 2010;70(1):68–77. doi: 10.1158/0008-CAN-09-2587.
  28. Chevolet I, Speeckaert R, Schreuer M, et al. Characterization of the in vivo immune network of IDO, tryptophan metabolism, PD-L1, and CTLA-4 in circulating immune cells in melanoma. Oncoimmunology. 2015;4(3):e982382. doi: 10.4161/2162402X.2014.982382.
  29. Jitschin R, Braun M, Buttner M, et al. CLL-cells induce IDOhiCD14+HLA-DRlo myeloid-derived suppressor cells that inhibit T-cell responses and promote Tregs. Blood. 2014;124(5):750–60. doi: 10.1182/blood-2013-12-
  30. Nagaraj S, Gupta K, Pisarev V, et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13(7):828–35. doi: 10.1038/nm1609.
  31. Lu T, Ramakrishnan R, Altiok S, et al. Tumor-infiltrating myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J Clin 2011;121(10):4015–4029. doi: 10.1172/JCI45862.
  32. Hanson EM, Clements VK, Sinha P, et al. Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J. Immunol. 2009;183(2):937–44. doi: 10.4049/jimmunol.0804253.
  33. Noman MZ, Desantis G, Janji B, et al. PD-L1 is a novel direct target of HIF-1a, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–90. doi: 10.1084/jem.20131916.
  34. Filipazzi P, Valenti R, Huber V, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol. 2007;25(18):2546–53. doi: 10.1200/JCO.2006.08.5829.
  35. Sinha P, Clements VK, Bunt SK, et al. Cross-talk between myeloid-derived suppressor cells and macrophages subverts tumor immunity toward a type 2 response. J Immunol. 2007;179(2):977–83. doi: 10.4049/jimmunol.179.2.977.
  36. Li H, Han Y, Guo Q, et al. Cancer-expanded myeloid-derived suppressor cells induce anergy of NK cells through membrane-bound TGF-beta 1. J Immunol. 2009;182(1):240–9. doi: 10.4049/jimmunol.182.1.240.
  37. Liu C, Yu S, Kappes J, et al. Expansion of spleen myeloid suppressor cells represses NK cell cytotoxicity in tumor-bearing host. Blood. 2007;109(10):4336–42. doi: 10.1182/blood-2006-09-
  38. Elkabets M, Ribeiro VSG, Dinarello CA, et al. IL-1b regulates a novel myeloid-derived suppressor cell subset that impairs NK cell development and function. Eur J Immunol. 2010;40(12):3347–57. doi: 10.1002/eji.201041037.
  39. Hoechst B, Voigtlaender T, Ormandy L, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50(3):799–807. doi: 10.1002/hep.23054.
  40. Pan PY, Ma G, Weber KJ, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70(1):99–108. doi: 10.1158/0008-CAN-09-1882.
  41. Hoechst B, Gamrekelashvili J, Manns MP, et al. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood. 2011;117(24):6532–41. doi: 10.1182/blood-2010-11-
  42. Shojaei F, Wu X, Malik AK, et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25(8):911–20. doi: 10.1038/nbt1323.
  43. Connolly MK, Mallen-St Clair J, Bedrosian AS, et al. Distinct populations of metastases-enabling myeloid cells expand in the liver of mice harboring invasive and preinvasive intra-abdominal tumor. J Leuk Biol. 2010;87(4):713–25. doi: 10.1189/jlb.0909607.
  44. Yang L, Huang J, Ren X, et al. Abrogation of TGFb signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13(1):23–35. doi: 10.1016/j.ccr.2007.12.004.
  45. Giles A, Vicioso Y, Kasai M, et al. Bone marrow-derived progenitor cells develop into myeloid-derived suppressor cells at metastatic sites. J Immunother Cancer. 2013;1(Suppl 1):188. doi: 10.1186/2051-1426-1-S1-P188.
  46. Solito S, Falisi E, Diaz-Montero CM, et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood. 2011;118(8):2254–65. doi: 10.1182/blood-2010-12-
  47. Marigo I, Bosio E, Solito S, et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity. 2010;32(6):790–802. doi: 10.1016/j.immuni.2010.05.010.
  48. Highfill SL, Rodriguez PC, Zhou Q, et al. Bone marrow myeloid-derived suppressor cells (MDSCs) inhibit graft-versus-host disease (GVHD) via an arginase-1-dependent mechanism that is up-regulated by interleukin-13. Blood. 2010;116(25):5738–47. doi: 10.1182/blood-2010-06-
  49. Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol. 2010;185(4):2273–84. doi: 10.4049/jimmunol.1000901.
  50. Atretkhany KS, Nosenko MA, Gogoleva VS, et al. TNF Neutralization Results in the Delay of Transplantable Tumor Growth and Reduced MDSC Accumulation. Front Immunol. 2016;7:147. doi: 10.3389/fimmu.2016.00147.
  51. De Veirman K, Van Valckenborgh E, Lahmar Q, et al. Myeloid-derived suppressor cells as therapeutic target in hematological malignancies. Front Oncol. 2014;4:349. doi: 10.3389/fonc.2014.00349.
  52. Ramachandran I, Martner A, Pisklakova A, et al. Myeloid-derived suppressor cells regulate growth of multiple myeloma by inhibiting T cells in bone marrow. J Immunol. 2013;190(7):3815–23. doi: 10.4049/jimmunol.1203373.
  53. De Veirman K, Van Ginderachter JA, Lub S, et al. Multiple myeloma induces Mcl-1 expression and survival of myeloid-derived suppressor cells. Oncotarget. 2015;6(12):10532–47. doi: 10.18632/oncotarget.3300.
  54. Brimnes MK, Vangsted AJ, Knudsen LM, et al. Increased level of both CD4+FOXP3+ regulatory T cells and CD14+HLA-DR/low myeloid-derived suppressor cells and decreased level of dendritic cells in patients with multiple myeloma. Scand J Immunol. 2010;72(6):540–7. doi: 10.1111/j.1365-2010.02463.x.
  55. Gorgun GT, Whitehill G, Anderson JL, et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121(15):2975–87. doi: 10.1182/blood-2012-08-
  56. Gorgun GТ, Samur MK, Cowens KB, et al. Lenalidomide Enhances Immune Checkpoint Blockade-Induced Immune Response in Multiple Myeloma. Clin Cancer Res. 2015;21(20):4607–18. doi: 10.1158/1078-CCR-15-0200.
  57. Busch A, Zeh D, Janzen V, et al. Treatment with lenalidomide induces immuno-activating and counter-regulatory immunosuppressive changes in myeloma patients. Clin Exp Immunol. 2014;177(2):439–53. doi: 10.1111/cei.12343.
  58. Wang Z, Zhang L, Wang H, et al. Tumor-induced CD14+HLA-DR (-/low) myeloid-derived suppressor cells correlate with tumor progression and outcome of therapy in multiple myeloma patients. Cancer Immunol Immunother. 2015;64(3):389–99. doi: 10.1007/s00262-014-1646-
  59. De Keersmaecker B, Fostier K, Corthals J, et al. Immunomodulatory drugs improve the immune environment for dendritic cell-based immunotherapy in multiple myeloma patients after autologous stem cell transplantation. Cancer Immunol Immunother. 2014;63(10):1023–36. doi: 10.1007/s00262-014-1571-
  60. Castella B, Foglietta M, Sciancalepore P, et al. Anergic bone marrow Vg9Vd2 T cells as early and long-lasting markers of PD-1-targetable microenvironment-induced immune suppression in human myeloma. Oncoimmunology. 2015;4(11):e1047580. doi: 10.1080/2162402X.2015.1047580.
  61. Giallongo C, Tibullo D, Parrinello NL, et al. Granulocyte-like myeloid derived suppressor cells (G-MDSC) are increased in multiple myeloma and are driven by dysfunctional mesenchymal stem cells (MSC). Oncotarget. 2016;7(52):85764– doi: 10.18632/oncotarget.7969.
  62. Lee SE, Lim JY, Ryu DB, et al. Circulating immune cell phenotype can predict the outcome of lenalidomide plus low-dose dexamethasone treatment in patients with refractory/relapsed multiple myeloma. Cancer Immunol Immunother. 2016;65(8):983–94. doi: 10.1007/s00262-016-1861-
  63. Favaloro J, Liyadipitiya T, Brown R, et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk Lymphoma. 2014;55(12):2893–900. doi: 10.3109/10428194.2014.904511.
  64. Franssen LE, van de Donk NW, Emmelot ME, et al. The impact of circulating suppressor cells in multiple myeloma patients on clinical outcome of DLIs. Bone Marrow Transplant. 2015;50(6):822–8. doi: 10.1038/bmt.2015.48.
  65. Lin Y, Gustafson MP, Bulur PA, et al. Immunosuppressive CD14+HLA-DRlow/– monocytes in B-cell non-Hodgkin lymphoma. Blood. 2011;117(3):872–81. doi: 10.1182/blood-2010-05-
  66. Tadmor T, Fell R, Polliack A, et al. Absolute monocytosis at diagnosis correlates with survival in diffuse large B-cell lymphoma—possible link with monocytic myeloid-derived suppressor cells. Hematol 2013;31(2):65–71. doi: 10.1002/hon.2019.
  67. Gustafson MP, Lin Y, LaPlant B, et al. Immune monitoring using the predictive power of immune profiles. J Immunother Cancer. 2013;1(1):7. doi: 10.1186/2051-1426-1-7.
  68. Wu C, Wu X, Zhang X, et al. Prognostic significance of peripheral monocytic myeloid-derived suppressor cells and monocytes in patients newly diagnosed with diffuse large B-cell lymphoma. Int J Clin Exp Med. 2015;8(9):15173–81.
  69. Sato Y, Shimizu K, Shinga J, et al. Characterization of the myeloid-derived suppressor cell subset regulated by NK cells in malignant lymphoma. Oncoimmunology. 2015;4(3):e995541. doi: 10.1080/2162402X.2014.995541.
  70. Romano A, Parrinello NL, Vetro C, et al. Circulating myeloid-derived suppressor cells correlate with clinical outcome in Hodgkin Lymphoma patients treated up-front with a risk-adapted strategy. Br J Haematol. 2015;168(5):689–700. doi: 10.1111/bjh.13198.
  71. Marini O, Spina C, Mimiola E, et al. Identification of granulocytic myeloid-derived suppressor cells (G-MDSCs) in the peripheral blood of Hodgkin and non-Hodgkin lymphoma patients. Oncotarget. 2016;19(7):27677–88. doi: 10.18632/oncotarget.8507.
  72. Azzaoui I, Uhel F, Rossille D, et al. T-cell defect in diffuse large B-cell lymphomas involves expansion of myeloid derived suppressor cells expressing IL-10, PD-L1 and S100A12. Blood. 2016;128(8):1081–92. doi: 10.1182/blood-2015-08-
  73. Zhang H, Li ZL, Ye SB, et al. Myeloid-derived suppressor cells inhibit T cell proliferation in human extranodal NK/T cell lymphoma: a novel prognostic indicator. Cancer Immunol Immunother. 2015;64(12):1587- doi: 10.1007/s00262-015-1765-6.
  74. Christiansson L, Sоderlund S, Svensson E, et al. Increased Level of Myeloid-Derived Suppressor Cells, Programmed Death Receptor Ligand 1/Programmed Death Receptor 1, and Soluble CD25 in Sokal High Risk Chronic Myeloid Leukemia. PLoS One. 2013;8(1):e55818. doi: 10.1371/journal.pone.0055818.
  75. Giallongo C, Romano A, Parrinello NL, et al. Mesenchymal Stem Cells (MSC) Regulate Activation of Granulocyte-Like Myeloid Derived Suppressor Cells (G-MDSC) in Chronic Myeloid Leukemia Patients. PLoS One. 2016;11(7):e0158392. doi: 10.1371/journal.pone.0158392.
  76. Gustafson МP, Abraham RS, Lin Y, et al. Association of an increased frequency of CD14+HLA-DRlo/neg monocytes with decreased time to progression in chronic lymphocytic leukaemia (CLL). Br J Haematol. 2012;156(5):674–6. doi: 10.1111/j.1365-2011.08902.x.
  77. Liu J, Zhou Y, Huang Q, et al. CD14+HLA-DRlow/– expression: a novel prognostic factor in chronic lymphocytic leukemia. Oncol 2015;9(3):1167–72. doi: 10.3892/ol.2014.2808.
  78. Sun H, Li Y, Zhang ZF, et al. Increase in myeloid-derived suppressor cells (MDSCs) associated with minimal residual disease (MRD) detection in adult acute myeloid leukemia. Int J Hematol. 2015;102(5):579–86. doi: 10.1007/s12185-015-1865-
  79. Gleason MK, Ross JA, Warlick ED, et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood. 2014;123(19):3016–26. doi: 10.1182/blood-2013-10-
  80. Chen X, Eksioglu EA, Zhou J, et al. Induction of myelodysplasia by myeloid-derived suppressor cells. J Clin Invest. 2013;123(11):4595–611. doi: 10.1172/JCI67580.
  81. Kittang AO, Kordasti S, Sand KE, et al. Expansion of myeloid derived suppressor cells correlates with number of T regulatory cells and disease progression in myelodysplastic syndrome. Oncoimmunology. 2015;5(2):e1062208. doi: 10.1080/2162402X.2015.1062208.
  82. Noonan KA, Ghosh N, Rudraraju L, et al. Targeting immune suppression with PDE5 inhibition in end-stage multiple myeloma. Cancer Immunol Res. 2014;2(8):725–31. doi: 10.1158/2326-CIR-13-0213.

 

 

Quality of Life of Patients with Lymphomas at Different Time-Points after High-Dose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation

N.E. Mochkin1, D.A. Fedorenko1, V.Ya. Mel’nichenko1, T.I. Ionova2, T.P. Nikitina1,2, K.A. Kurbatova2, A.A. Novik1

1 N.I. Pirogov National Medical and Surgical Center under the Ministry of Health of the Russian Federation, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203

2 International Quality of Life Research Center, 1 office 152, Artilleriiskaya str., Saint Petersburg, Russian Federation, 191014

For correspondence: N.E. Mochkin, PhD, assistant, 70 Nizhnyaya Pervomaiskaya str., Moscow, Russian Federation, 105203; Tel: +7(495)603-72-17; e-mail: nickmed@yandex.ru

For citation: Mochkin N.E., Fedorenko D.A., Mel’nichenko V.Ya., Ionova T.I., Nikitina T.P., Kurbatova K.A., Novik A.A. Quality of Life of Patients with Lymphomas at Different Time-Points after HighDose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation. Klin. Onkogematol. 2014; 7(4): 577–582 (In Russ.).

 


ABSTRACT

The article presents results of monitoring of quality of life of 103 patients with lymphomas (non-Hodgkin’s lymphomas, n = 36; Hodgkin’s lymphomas, n = 67) at different stages after high-dose chemotherapy with autologic hematopoietic stem cell transplantation (HDC + HSCT). The majority of patients experienced improvement or stabilization of their quality of life 1 year after the HDC + aHSCT. At that, the response associated with the quality of life and clinical response to the treatment did not coincide in all cases. Obtained results confirm the importance of a comprehensive approach to assessment of the efficacy of the treatment and may be use as a principle marker of patient’s recovery at different time points after the transplantation.


Keywords: quality of life, lymphomas, high-dose chemotherapy, autologic hematopoietic stem cell transplantation.

Accepted: September 16, 2014

Read in PDF (RUS)pdficon


REFERENCES

  1. Colpo A., Hochberg E., Chen Y.B. Current status of autologous stem cell transplantation in relapsed and refractory Hodgkin’s lymphoma. Oncologist. 2012; 17: 80–90.
  2. d’Amore F., Relander T., Lauritzen G.F. et al. High-dose chemotherapy and autologous stem cell transplantation in previously untreated peripheral T-cell lymphoma — final analysis of a large prospective multicenter study (NLGT-01). Blood (ASH Annual Meeting Abstracts). 2011; 118: 331.
  3. Damon L.E., Johnson J.L., Neidzwiecki D. et al. Immunochemotherapy and autologous stem-cell transplantation for untreated patients with mantle-cell lymphoma: CALGB 59909. J. Clin. Oncol. 2009; 27: 6101–8.
  4. Freidberg J.W. Relapsed/refractory diffuse large B-cell lymphoma. Hematol. Am. Soc. Hematol. Educ. Program. 2011: 498–501.
  5. Geisler C.H., Polstad A., Laurell A. et al. Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo-purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group. Blood. 2008; 112: 2687–93.
  6. Hjermstad M.J., Kaasa S. Quality of life in adult cancer patients treated with bone marrow transplantation — a review of the literature. Eur. J. Cancer. 1995; 31A(2): 163–73.
  7. Kiss T.L., Abdolell M., Jamal N. et al. Long-term medical outcomes and quality-of-life assessment of patients with chronic myeloid leukemia followed at least 10 years after allogenic bone marrow transplantation. J. Clin. Oncol. 2002; 20(9); 2334–43.
  8. Anderson K.O., Giralt S.A., Mendoza T.R. et al. Symptom burden in patients undergoing autologous stem-cell transplantation. Bone Marrow Transplant. 2007; 39(12): 759–66.
  9. Grant M., Ferrel B., Schmidt G.M. et al. Measurement of quality of life in bone marrow transplantation survivors. Qual. Life Res. 1992; 1(6): 375–84.
  10. Новик А.А., Ионова Т.И., Афанасьев Б.В. и др. Результаты аутоло- гичной трансплантации костного мозга/стволовых кроветворных клеток у больных гемобластозами: клиническая эффективность и показатели ка- чества жизни. Вестник Межнационального центра исследования качества жизни. 2011; 17–18: 22–32. [Novik A.A., Ionova T.I., Afanas’ev B.V. et al. Results of autologic bone marrow transplantation/hematopoietic stem cells transplantation in patients with hemoblastoses: clinical efficacy and parameters of quality of life. Vestnik Mezhnatsional’nogo tsentra issledovaniya kachestva zhizni. 2011; 17–18: 22–32. (In Russ.)]
  11. Rock E.P., Kennedy D.L., Furness M.H. et al. Patient-reported outcomes supporting anticancer product approvals. J. Clin. Oncol. 2007; 25: 5094–9.
  12. Fairclough D. Patient-reported outcomes as endpoints in medical research. Sta. Meth. Med. Res. 2004; 13: 115–38.
  13. Gondek K., Sagnier P., Gichrist K., Wooley J. Current status of patientreported outcomes in industry-sponsored oncology clinical trials and product labels. J. Clin. Oncol. 2007; 25(32): 5087–93.
  14. Steven B. Patient-reported outcomes assessment in cancer trials: evaluating and enhancing the payoff to decision making. J. Clin. Oncol. 2007; 25(32): 5049–50.
  15. Watkins B. Issues and challenges with integrating patient-reported outcomes in clinical trials supported by the national cancer institute-sponsored clinical trials networks. J. Clin. Oncol. 2007; 25(32): 5051–7.
  16. Molassiotis A., Van der Akker O., Milligan D. et al. Quality of life in longterm survivors of marrow transplantation: Comparison with a matched group receiving maintenance chemotherapy. Bone Marrow Transplant. 1996; 17: 249–58.
  17. Руководство по исследованию качества жизни в медицине, 3-е изд., перераб. и доп. Под ред. Ю.Л. Шевченко. М.: Изд-во РАЕН, 2012. [Shevchenko Yu.L., ed. Rukovodstvo po issledovaniyu kachestva zhizni v meditsine (Guidelines for evaluation of the quality of life in medicine). 3rd revised edition. Moscow: RAEN Publ.; 2012.]
  18. Neitzert C.S., Ritvo P., Dancey J. et al. The psychosocial impact of bone marrow transplantation: A review of the literature. Bone Marrow Transplant. 1998; 22: 409–22.
  19. Wingard J.R. Quality of life following bone marrow transplantation. Curr. Opin. Oncol. 1998; 10: 108–11.
  20. Chao N.J., Tierney D.K., Bloom J.R. et al. Dynamic assessment of quality of life after autologous bone marrow transplantation. Blood. 1992; 80: 825–30.
  21. Cohen M.Z., Mendoza T., Neumann J. et al. Longitudinal assessment of symptoms and quality of life: Differences by ablative and nonablative blood and marrow transplantation. J. Clin. Oncol. 2004; 22(15S): 6630.
  22. Ganz P., Gotay C. Use of Patient-Reported Outcomes in Phase III Cancer Treatment Trials; Lessons Learned and Future Directions. J. Clin. Oncol. 2007; 25(32): 5063–9.
  23. Novik A., Salek S., Ionova T. Patient-reported outcomes in hematology. Guidelines. EHA SWG Quality of Life and Symptoms. Litoprint. Genoa, 2012.
  24. Hays R.D., Sherbourne C.D., Mazel R.M. User’s Manual for Medical Outcomes Study (MOS) Core measures of health-related quality of life. RAND Corporation, MR-162-RC. Available at www.rand.org.
  25. Новик А.А., Ионова Т.И. Исследование качества жизни в медицине: Учебное пособие для вузов. Под ред. Ю.Л. Шевченко. М.: ГЭОТАР-Медиа, 2004. [Novik A.A., Ionova T.I. Issledovanie kachestva zhizni v meditsine (Evaluation of the quality of life in medicine). Textbook for institutes of higher education. Shevchenko Yu.L., ed. Moscow: GEOTAR-Media Publ.; 2004.]
  26. Новик А.А., Ионова Т.И. Интегральный показатель качества жизни — новая категория в концепции исследования качества жизни. Вестник Межнационального центра исследования качества жизни. 2006; 7–8: 7–8. [Novik A.A., Ionova T.I. Integral assessment of quality of life is a new category in the concept of evaluation of quality of life. Vestnik Mezhnatsional’nogo tsentra issledovaniya kachestva zhizni. 2006; 7–8: 7–8. (In Russ.)]

Minimally invasive surgical techniques in hematological malignancies with spinal involvement

A.K. Valiev, A.V. Sokolovsky, A.S. Nered, and E.R. Musaev

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


ABSTRACT

The current statistical data show an increased number of malignancies with skeletal bones involvement, that comprise in average 1.5–2 % of all malignancies. Among them according to some authors, the most common, in decreasing order of incidence, are: multiple myeloma (35–50 %), osteosarcomas (20–30 %), chondrosarcomas (10–17 %), Ewing’s sarcoma (6–12 %), and lymphomas (3–7 %). Therefore, the issue of treating pathological fractures of the spine in multiple myeloma or lymphoma becomes more pressing, since the number of such patients is increasing. Existing surgical minimally invasive techniques enable to increase the quality of life in these groups and start special conservative treatment as soon as possible.


Keywords: spine, pathological fracture, multiple myeloma, lymphomas

Read in PDF (RUS) pdficon


REFERENCES

  1. Malawer M.M. Musculoskeletal Cancer Surgery. In: Treatment of Sarcomas and Allied Diseases. Ed. by M.M. Malawer, P.H. Sugarbaker. Washington: Kluwer Academic Publishers, 2001.
  2. Зацепин С.Т. Костная патология взрослых: Руководство для врачей. М.: Медицина, 2001. [Zatsepin S.T. Kostnaya patologiya vzroslykh: Rukovodstvo dlya vrachey (Bone disorders in adults: manual for medical practitioners). M.: Meditsyna, 2001.]
  3. Алиев М.Д. Злокачественные опухоли костей. M., 2008. [Aliyev M.D. Zlokachestvennyye opukholi kostey (Malignant bone tumors). M., 2008.]
  4. Давыдов М.И., Аксель Е.М. Статистика злокачественных новообразо- ваний в России и странах СНГ в 2009 г. Вестн. онкол. 2011; 22(3 Прил. 1). [Davydov M.I., Aksel E.M. Statistika zlokachestvennykh novoobrazovaniy v Rossii i stranah SNG v 2009 g. (Statistics of malignancies in Russia and CIScountries in 2009). Vestn. onkol. 2011; 22(3 Suppl. 1).]
  5. Kyle R.A., Rajkumar S.V. Multiple myeloma. Blood 2008; 111: 2962–72.
  6. Rehak S., Maisnar V., Malek V. et al. Diagnosis and surgical therapy of plasma cell neoplasia of spine. Neoplasma 2009; 56: 84.
  7. Mendoza S., Urrutia J., Fuentus D. Surgical treatment of solitary plasmocytoma of the spine: case series. Iowa Orthopaed. J. 2004; 24: 86–94.
  8. McDonald R.J., Trout A.T., Gray L.A. et al. Vertebroplasty in Multiple Myeloma: Outcomes in a Large Patient Series. Am. J. Neuroradiol. 2008; 29: 642–8.
  9. Garland P., Gishen P., Rahemtulla A. Percutaneous vertebroplasty to treat painful myelomatous vertebral deposits — long term efficacy outcomes. Ann. Hematol. 2010 Jul 6.
  10. Dimopoulos M.A., Moulopolos L.A., Maniatis A., Alexenian R. Solitary plasmacytoma of bone and asymptomatic multiple myeloma. Blood 2000; 96: 2037–44.
  11. Hu K., Yahalom J. Radiotherapy in the management of plasma cell tumors. Oncology 2000; 14: 101–8.
  12. Maruyama D., Watanabe T., Beppu Y. et al. Primary Bone Lymphoma: A New and Detailed Characterization of 28 Patients in a Single-Institution Study. Hematol. Stem Cell Transplant. Div. 2006; 56–67.
  13. Cortet B., Cotton., Boutry N. et al. Percutaneous vertebroplasty in patients with osteolytic metastases or multiple myeloma [see comments]. Rev. Rheum. Engl. Ed. 1997; 64(3): 177–83.
  14. Durr H.R., Muller P.E., Hiller E. et al. Malignant lymphoma of bone. Arch. Orthopaed. Trauma Surg. 2002; 122: 10–6.
  15. Lecouvet F.E., Van den Berg B.C, Maldague B.E. et al. Vertebral compression fractures in multiple myeloma. Part I. Distribution and appearance at MR imaging. Radiology 1997; 204: 195–9.
  16. Durr H.R., Wegener B., Krodel A. et al. Multiple myeloma: surgery of the spine: retrospective analysis of 27 patients. Spine 2002; 27: 320–6.
  17. Kyle R.A., Gertz M.A., Witzing T.E. et al. Review of 1027 patients with newly diagnosed multiple myeloma. Mayo Clin. Proc. 2003; 78(1): 21–33.
  18. Chahal S., Lagera J.E., Ryder J., Kleinschmidt-DeMasters B.K. Hematological neoplasms with first presentation as spinal cord compression syndromes: a 10-year retrospective series and review of the literature. Clin. Neuropathol. 2003; 22(6): 282–90.
  19. Chiodo A. Spinal cord injury caused by epidural B-cell lymphoma: report of two cases. J. Spinal Cord Med. 2007; 30(1): 70–2.
  20. Rao G., Chul S.H., Chakrabarti I.I. et al. Multiple Myeloma of the cervical spine: treatment strategies for pain and spinal instability. J. Neurosurg. Spine. 2006; 5: 140-145.
  21. Kwon A.-H., Chang U.-K., Gwak H.-S. et al. The Role of Surgery in the Treatment of Spinal Myeloma. J. Korean Neurosurg. Soc. 2005; 37: 187–92.
  22. Masala S., Fiori R., Massari F. et al. Percutaneus kyphoplasty: indications and technique. Tumori 2004; 90: 22–6.
  23. Slatkin N. Cancer-Related Pain and its Pharmacologic Management in the Patients With Bone Metastasis. J. Support Oncol. 2006; 4(Suppl. 1): 015–21.
  24. Алиев М.Д., Долгушин Б.И., Валиев А.К. и др. Чрезкожная вертебро- пластика в онкологии. М.: Издательская группа РОНЦ, 2008: 43–54. [Aliyev M.D., Dolgushin B.I., Valiyev A.K. i dr. Chrezkozhnaya vertebroplastika v onkologii (Transcutaneous vertebroplasty in oncology). M.: Izdatelskaya gruppa RONTS, 2008: 43–54.]
  25. Deramond H., Dion J.E., Chiras J. Complications in vertebroplasty. In: Percutaneous Vertebroplasty. Ed. by J.M. Mathis, H. Deramond, S.M. Belkoff. New York: Springer-Verlag, 2002: 165–73.
  26. Kaemmerlen P., Thiesse P., Jonas P. et al. Percutaneous injection of orthopedic cement in metastatic vertebral lesions [letter]. N. Engl. J. Med. 1989; 321(2): 121.
  27. Komemushi A., Tanigawa N., Kariya S. et al. Percutaneous vertebroplasty for compression fracture: analysis of vertebral body volume by CT volumetry. Acta Radiol. 2005; 46: 276–9.
  28. Валиев А.К., Мусаев Э.Р., Тепляков В.В. и др. Чрезкожная вертебро- пластика в онкологии. М.: ИНФРА-М, 2010: 69. [Valiyev A.K., Musaev E.R., Teplyakov V.V. i dr. Chrezkozhnaya vertebroplastika v onkologii (Transcutaneous vertebroplasty in oncology). M.: INFRA-M, 2010: 69.]
  29. Каллистов В.Е. Метастатические опухоли позвоночника (клиника, диагностика, лечение): Дис. ¼ канд. мед. наук. М., 1999. [Kallistov V.E. Metastaticheskiye opukholi pozvonochnika (klinika, diagnostika, lecheniye): Avtoref. dis. … kand. med. nauk (Metastatic spinal tumors (presentation, diagnosis, management)). Author’s summary of dissertation for the degree of Candidate of medical sciences. M., 1999.]
  30. Wetzel F.T., Maurer P., Thompson K. et al. Minimally Invasive Spine Surgery: A Surgical Manual. Spine 2001; 25: 382–8.
  31. Boriani S., Gasparrini A., Paderni S., Bandiera S., Cappucio M. Terapia chirurgica delle lesioni vertebralinel mieloma. Haematologica 2004; 89: 21–3.
  32. Schiff D. Spinal cord compression. Neurol. Clin. 2003; 21: 67–86.
  33. Walker M.P., Yaszemski M.J., Kim C.W. et al. Metastatic disease of the spine: evaluation and treatment. Clin. Orthop. 2003; 415: S165–75.
  34. Sharma B.S., Gupta S.K., Khosla V.K. et al. Midline and far lateral approaches to foramen magnum lesions. Neurol. India 1999; 47: 268–71.
  35. Chiras J., Deramond H. Complications des vertebroplasties. In: Echecs et Complications de la Chirurgie du Rachis. Chirurgie de Reprise. Ed. by G. Sailant, C. Laville. Paris: Sauramps Medical, 1995: 149–53.
  36. Cyteval C., Sarrabere M.P., Roux J.O. et al. Acute osteoporotic vertebral collapse: open study on percutaneous injection of acrylic surgical cement in 20 patients. Am. J. Roentgenol. 1999; 173(6): 1685–90.
  37. Li K.C., Poon P.Y. Sensitivity and specificity of MRI in detecting malignant spinal cord compression and in distinguishing malignant from benign compression fractures of vertebrae. Magn. Reson. Imaging 1988; 6: 547–56.
  38. Anselmetti G.C., Corgnier A., Debernardi F. et al. Treatment of painful compression vertebral fractures with vertebroplasty: results and complications. Radiol. Med. (Torino) 2005; 110: 262–72.
  39. Grados F., Depriester C., Cayrolle G. et al. Long-term observations of vertebral osteoporotic fractures treated by percutaneous vertebroplasty. Rheumatology (Oxford) 2000, 39: 1410–4.
  40. Uppin A.A., Hirsch J.A., Centenera L.V. et al. Occurrence of new vertebral body fracture after percutaneous vertebroplasty in patients with osteoporosis. Radiology 2003; 226: 119–24.
  41. Harrop J.S., Prpa B., Reinhardt M.K. et al. Primary and Secondary Osteoporosis’ Incidence of Subsequent Vertebral Compression Fractures After Kyphoplasty. Spine 2004; 29: 2120–5.
  42. Синельников Р.Д. Атлас анатомии человека. Т. I. М.: Медицина, 1972. [Synelnikov R.D. Atlas anatomii cheloveka (Atlas of human anatomy). T. I. M.: Meditsyna, 1972.]
  43. O’Brien J.P., Sims J.T., Evans A.J. Vertebroplasty in patients with severe vertebral compression fractures: a technical report. Am. J. Neuroradiol. 2000; 21(8): 1555–8.
  44. Dahl O.E., Garvik L.J., Lyberg T. Toxic effects of methylmetacrylate monomer on leukocytes and endothelial cells in vitro [published erratum appeared in Acta Orthop Scand. 1995; 66(4): 387]. Acta Orthop. Scand. 1994; 65(2): 147–53.
  45. Belkoff S.M., Mathis J.M., Jasper L.E. et al. The biomechanics of vertebroplasty: the effect of cement volume on mechanical behavior. Spine 2001; 26(14): 1537–41.
  46. Barr J.D., Barr M.S., Lemley T.J. et al. Percutaneous vertebroplasty for pain relief and spinal stabilization. Spine 2000; 25(8): 923–8.
  47. Heini P.F., Walchli B., Berlemann U. Percutaneous transpedicular vertebroplasty with PMMA: operative technique and early results. A prospective study for the treatment of osteoporotic compression fractures. Eur. Spine J. 2000; 9: 445–50.