Skip to main content
Log in

Breakthroughs in the Management of Multiple Myeloma

  • Leading Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Although multiple myeloma remains a terminal illness, the past four decades have seen a dramatic change in the outlook for a newly diagnosed patient in terms of therapies available, supportive care and insight into the pathogenesis of this disease. Among the newer agents available for treatment, thalidomide has been resurrected and discovered to be a valuable therapy for myeloma. Thalidomide appears to work, at least in part, through its anti-angiogenic properties, but much remains to be learned about its mechanism of action as well as optimal administration regimens.

With the development of increasingly more potent bisphosphonates it has become possible to diminish the painful skeletal complications of myeloma, one of the most devastating problems of this disease. The most recent generation of bisphosphonates, pamidronic acid and zoledronic acid, have provided a statistically significant decrease in the skeletal complications of myeloma when used in a prophylactic manner. These agents appear to work by inhibiting osteoclast function.

Progressive improvement in cytogenetic techniques has now demonstrated that almost all patients with myeloma have chromosomal abnormalities, some of which appear to confer varying degrees of prognostic significance. In particular, the changes in chromosome 13 are associated with an unusually poor outcome. These findings are serving as a guide toward learning more about the pathogenesis of myeloma as well as in identifying potential targets for therapy.

Stem cell transplantation has emerged as the standard treatment for the large majority of patients with myeloma following the demonstration of superior complete remission and survival, both disease-free and overall, in a French randomised trial. Unfortunately, virtually all patients will eventually relapse following autologous stem cell transplantation, prompting continuing efforts such as tandem transplants, CD34+ selection, as well as modifications in the conditioning regimen to improve outcomes. Allogeneic bone marrow transplants appear to offer a better chance for a possible cure of myeloma but have been associated with an unusually high mortality. However, this approach is being revived with the advent of the less toxic non-myeloablative transplant that has provided an 81% short-term survival in a trial combining this approach with an initial conventional autologous bone marrow transplant. Immunotherapy with dendritic cells appears now to be a feasible way to enhance innate or acquired immunity to help eliminate minimal residual disease following autologous bone marrow transplant. Unfortunately, a cure for myeloma remains elusive but the continuing advances in management may significantly prolong survival in affected patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Table I
Table II
Table III
Fig. 1
Table IV
Table V
Table VI

Similar content being viewed by others

References

  1. Osgood EE. The survival time of patients with plasmacytic myeloma. Cancer Chemother Rep 1960; 9: 1–10

    PubMed  CAS  Google Scholar 

  2. Folkman J. Seminars in medicine of the Beth Israel Hospital, Boston: clinical applications of research on angiogenesis. N Engl J Med 1995; 333: 1757–63

    PubMed  CAS  Google Scholar 

  3. Vacca A, Rubatti C, Presta M, et al. Bone marrow neovascularization, plasma cell angiogenic potential, and matrix metal-loproteinase-2 secretion parallel progression of human multiple myeloma. Blood 1999; 98: 3064–73

    Google Scholar 

  4. D’Amato RJ, Loughnan MS, Flynn E, et al. Thalidomide is an inhibitor of angiogenesis. Proc Natl Acad Sci U S A 1994; 91: 4082–5

    PubMed  Google Scholar 

  5. Singhal S, Mehta J, Desikan R, et al. Anti-tumor activity of thalidomide in refractory multiple myeloma. N Engl J Med 1999; 341: 1565–71

    PubMed  CAS  Google Scholar 

  6. Barlogie B, Desikan R, Eddlemon P, et al. Extended survival in advanced and refractory multiple myeloma after single-agent thalidomide: identification of prognostic factors in a phase 2 study of 169 patients. Blood 2001; 98(2): 492–4

    PubMed  CAS  Google Scholar 

  7. Rajkumar SV, Fonseca R, Dispenzieri A, et al. Thalidomide in the treatment of relapsed multiple myeloma. Mayo Clin Proc 2000; 75: 897–902

    PubMed  CAS  Google Scholar 

  8. Juliusson G, Celsing F, Turesson I, et al. Frequent good partial remissions from thalidomide including best response ever in patients with advanced refractory and relapsed myeloma. Br J Haematol 2000; 109: 89–96

    PubMed  CAS  Google Scholar 

  9. Barlogie B, Zangari M, Spencer T, et al. Thalidomide in the management of multiple myeloma. Semin Hematol 2001; 38(3): 250–9

    PubMed  CAS  Google Scholar 

  10. Rajkumar SV, Hayman S, Fonseca R, et al. Thalidomide plus dexamethasone (Thal/Dex) and thalidomide alone (Thal) as first-line therapy for newly diagnosed myeloma [abstract]. Blood 2000; 96: 168A

    Google Scholar 

  11. Weber DM, Fankkin K, Gavino M, et al. Thalidomide alone or with dexamethasone for previously untreated myeloma. J Clin Oncol 2003; 21(1): 16–9

    PubMed  CAS  Google Scholar 

  12. Rajkumar SV, Hayman S, Gertz MA, et al. Combination therapy with thalidomide plus dexamethasone for newly diagnosed myeloma. J Clin Oncol 2002; 20(21): 4319–23

    PubMed  CAS  Google Scholar 

  13. Geitz J, Handt S, Zwingengerger K. Thalidomide selectively modulates the density of cell surface molecules involved in the adhesion cascade. Immunopharmacology 1996; 32: 213–21

    Google Scholar 

  14. Moreira AL, Sampaio EP, Zmuidzinas A, et al. Thalidomide exerts its inhibitory action on tumor necrosis factor alpha by enhancing mRNA degradation. J Exp Med 1993; 177: 1675–80

    PubMed  CAS  Google Scholar 

  15. Parman T, Wiley MF, Wells PG. Free radical mediated oxidative DNA damage in the mechanism of thalidomide teratogenicity. Nat Med 1999; 5: 582–58

    PubMed  CAS  Google Scholar 

  16. Haslett PA, Corral LG, Albert M, et al. Thalidomide costimulates primary human T lymphocytes, preferentially inducing proliferation, cytokine production and cytotoxic responses in the CD8+ subset. J Exp Med 1998; 187(11): 1805–992

    Google Scholar 

  17. Durie BGM, Stepan DE. Efficacy of low-dose thalidomide (T) in multiple myeloma [abstract]. Blood 1999; 94 Suppl. 1: 316A

    Google Scholar 

  18. Rajkumar SV, Witzig TE. A review of angiogenesis and antiangiogenic therapy with thalidomide in multiple myeloma. Cancer Treat Rev 2000; 26: 351–62

    PubMed  CAS  Google Scholar 

  19. Fullerton PM, O’Sullivan DJ. Thalidomide neuropathy: a clinical, electrophysiological, and histological follow-up study. J Neurol Neurosurg Psychiatry 1968; 31: 543–51

    PubMed  CAS  Google Scholar 

  20. Rajkumar SV, Gertz MA, Witzig TE. Life-threatening toxic epidermal necrolysis with thalidomide therapy for myeloma. N Engl J Med 2000; 343: 972–3

    PubMed  CAS  Google Scholar 

  21. Osman K, Comenzo R, Rajkumar SV. Deep vein thrombosis and thalidomide therapy for multiple myeloma. N Engl J Med 2001; 344: 1951–2

    PubMed  CAS  Google Scholar 

  22. Coleman RE. Skeletal complications of malignancy. Cancer 1997; 80 Suppl. 8: 1588–94

    PubMed  CAS  Google Scholar 

  23. Thurlimann B. Bisphosphonates in clinical oncology: focus on pamidronate. Heidelberg: Springer-Verlag, 1999: 1–113

    Google Scholar 

  24. Evans CE, Braidman IP. Effects of two novel bisphosphonates on bone cells in vitro. J Bone Miner Res 1994; 26: 95–107

    CAS  Google Scholar 

  25. Green J, Muller K, Jaeggi DA. Preclinical pharmacology of CGP42446, a new, potent, heterocyclic bisphosphonate compound. J Bone Miner Res 1994; 19(5): 745–51

    Google Scholar 

  26. Derenne S, Amiot M, Barille S, et al. Zoledronate is a potent inhibitor of myeloma cell growth and secretion of IL-6 and MMP-1 by the tumoural environment. J Bone Miner Res 1999; 14: 2048–56

    PubMed  CAS  Google Scholar 

  27. Boissier S, Ferreras M, Peyruchaud O, et al. Bisphosphonates inhibit breast and prostate carcinoma cell invasion, an early event in the formation of bone metastases. Cancer Res 2000; 60: 2049–954

    Google Scholar 

  28. Luckman SP, Huges DE, Coxon FP, et al. Nitrogen-containing biphosphonates inhibit the mevalonate pathway and prevent post-translational prenylation of GTP-binding proteins, including Ras. J Bone Miner Res 1998; 13: 581–9

    PubMed  CAS  Google Scholar 

  29. Russell RG, Rogers MJ. Bisphosphonates: from the laboratory to the clinic and back again. Bone 1999; 25(1): 97–106

    PubMed  CAS  Google Scholar 

  30. van Beek E, Pieterman E, Cohen L, et al. Farnesyl pyrophosphate synthase is the molecular target of nitrogen-containing bisphosphonates. Biochem Biophy Res Commun 1999; 264: 108–11

    Google Scholar 

  31. Major P, Lortholary A, Hon J, et al. Zoledronic acid is superior to pamidronate in the treatment of hypercalcemia of malignancy: a pooled analysis of two randomized, controlled clinical trials. J Clin Oncol 2001; 19(2): 558–67

    PubMed  CAS  Google Scholar 

  32. Berenson J, Lichtenstein A, Porter L, et al. Efficacy of pamidronate in reducing skeletal events in patients with advanced multiple myeloma. N Engl J Med 1996; 334(8): 488–93

    PubMed  CAS  Google Scholar 

  33. Berenson J, Lichtenstein A, Porter L, et al. Long-term pamidronate treatment of advanced multiple myeloma patients reduces skeletal events. J Clin Oncol 1998; 16(2): 593–602

    PubMed  CAS  Google Scholar 

  34. Ali SM, Esteva JF, Hortobagyi G, et al. Safety and efficacy of bisphosphonates beyond 24 months in cancer patients. J Clin Oncol 2001; 19(14): 3434–7

    PubMed  CAS  Google Scholar 

  35. Philip P. Chromosomes of monoclonal gammopathies. Cancer Genet Cytogenet 1980; 2: 79–86

    Google Scholar 

  36. Dewald GW, Kyle RA, Hicks GA, et al. The clinical significance of cytogenetic studies in 100 patients with multiple myeloma, plasma cell leukemia, or amyloidosis. Blood 1985; 66(2): 380–90

    PubMed  CAS  Google Scholar 

  37. Lai JL, Zandecki M, Mary JY, et al. Improved cytogenetics in multiple myeloma: a study of 151 patients including 177 patients at diagnosis. Blood 1995; 85: 2490–7

    PubMed  CAS  Google Scholar 

  38. Zandecki M, Lai JL, Facon T. Multiple myeloma: almost all patients are cytogenetically abnormal. Br J Haematol 1996; 94(2): 217–27

    PubMed  CAS  Google Scholar 

  39. Tricot G, Sawyer JR, Jagannath S, et al. Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving llq and not with other karyotype abnormalities. Blood 1995; 86(11): 4250–6

    PubMed  CAS  Google Scholar 

  40. Königsberg R, Zojer N, Ackermann J, et al. Predictive value of interphase cytogenetics for survival of patients with multiple myeloma. J Clin Oncol 2000; 18: 804–12

    PubMed  Google Scholar 

  41. Avet-Loiseau H, Li JY, Facon T, et al. High incidence of translocations t(ll;14) (ql3;q32) and t(4;14) (pl6;q32). Cancer Res 1998; 58: 5640–5

    PubMed  CAS  Google Scholar 

  42. Avet-Loiseau H, Moreau P, Huyghe P, et al. A comparative FISH analysis identifies myeloma patient subgroups with different outcome [abstract]. Blood 2001; 98(11): 370A

    Google Scholar 

  43. Fonseca R, Coignet LJ, Dewald GW. Cytogenetic abnormalities in multiple myeloma. Hematol Oncol Clin North Am 1999 Dec; 13(6): 1169–80

    PubMed  CAS  Google Scholar 

  44. Seong C, Delasalle K, Hayes K, et al. Prognostic value of cytogenetics in multiple myeloma. Br J Haematol 1998; 101: 189–95

    PubMed  CAS  Google Scholar 

  45. Facon T, Hervé AL, Gaë G, et al. Chromosome 13 abnormalities identified by FISH analysis and serum β2-microglobulin produce a powerful myeloma staging system for patients receiving high-dose therapy. Blood 2001; 97(6): 1566–71

    PubMed  CAS  Google Scholar 

  46. Debes Marún CS, Bailey RJ, Dewald GW, et al. In multiple myeloma the combination of an elevated plasma cell labeling index (PCLI) and chromosome 13 deletion, detected by interphase FISH, can identify patients with chromosome 13 abnormalities detected by karyotype [abstract]. Blood 2001; 98(11): 156A

    Google Scholar 

  47. Nishida K, Tamura A, Nakazawa N, et al. The Ig heavy chain gene is frequently involved in chromosomal translocations in multiple myeloma and plasma cell leukemia as detected by in situ hybridization. Blood 1997; 90: 526–34

    PubMed  CAS  Google Scholar 

  48. Chesi M, Brents LA, Ely SA, et al. Activated fibroblast growth factor receptor 3 is an oncogene that contributes to tumor progression in multiple myeloma. Blood 2001; 97(3): 729–36

    PubMed  CAS  Google Scholar 

  49. Richardson P, Barlogie B, Berenson J, et al. A phase II multicenter study of the proteasome inhibitor bortezomib (Velcade™ formerly PS-341) in multiple myeloma patients (pts) with relapsed/refractory disease [abstract]. Blood 2002; 100: 104A

    Google Scholar 

  50. Richardson PG, Schlossman RL, Weiler E, et al. Immunomodulatory drug CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed myeloma. Blood 2002; 100: 3063–7

    PubMed  CAS  Google Scholar 

  51. Richardson P, Jagannath S, Schlossman R, et al. A multi-center randomized phase II study to evaluate the efficacy and safety of two CDC-5013 dose regimens when used alone or in combination with dexamethasone (Dex) for the treatment of relapsed or refractory multiple myeloma (MM) [abstract]. Blood 2002; 100: 104A

    Google Scholar 

  52. Hayashi T, Hideshima T, Akiyama M, et al. Arsenic trioxid inhibits growth of human multiple myeloma cells in the bone marrow microenvironment [abstract]. Blood 2001; 98(11): 375A

    Google Scholar 

  53. Deaglio S, Canela D, Gai G, et al. Evidence of an immunologic mechanism behind the therapeutic effect of arsenic trioxide (As203) in myeloma cells. Leuk Res 2001; 25: 227–35

    PubMed  CAS  Google Scholar 

  54. Munshi NC. Arsenic trioxide: an emerging therapy for multiple myeloma. Oncologist 2001; 6 Suppl. 2: 17–21

    PubMed  CAS  Google Scholar 

  55. Hussein MA, Mason J, Ravandi F, et al. A phase II clinical study of arsenic trioxide (ATO) in patients (pts) with relapsed or refractory multiple myeloma (MM): a preliminary report [abstract]. Blood 2001; 98(11): 378A

    Google Scholar 

  56. Bahis N, Jordan-McMurry I, Grad J, et al. Phase I results from a phase I/II study of arsenic trioxide (As203) and ascorbic acid (AA) in relapsed and chemotherapy refractory multiple myeloma [abstract]. Blood 2001; 98(11): 375A

    Google Scholar 

  57. Attal M, Harousseau JL, Stoppa AM, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma: Intergroupe Francais du Myelome. N Engl J Med 1996; 335: 91–7

    PubMed  CAS  Google Scholar 

  58. Dalton WS, Bergsagel PL, Kuehl WM, et al. Multiple myeloma. In: Hematology: American Society of Hematology Education program book. Washington, DC: American Society of Hematology, 2001: 157–77

    Google Scholar 

  59. Fermand JP, Ravaud P, Chevret S, et al. High-dose therapy and autologous peripheral blood stem cell transplantation in multiple myeloma: up-front or rescue treatment? Results of a multicenter sequential randomized clinical trial. Blood 1998; 92: 3131–6

    PubMed  CAS  Google Scholar 

  60. Majolino I, Vignetti M, Meloni G, et al. Autologous transplantation in multiple myeloma: a GITMO retrospective analysis on 290 patients: Gruppo Italiano Trapianti di Midollo Osseo. Haematologica 1999; 84: 844–52

    PubMed  CAS  Google Scholar 

  61. Tribalto M, Amadori S, Cudillo L, et al. Autologous peripheral blood stem cell transplantation as first line treatment of multiple myeloma: an Italian multicenter study. Haematologica 2000; 85: 52–8

    PubMed  CAS  Google Scholar 

  62. Blade J, Esteve J, Rives S, et al. High-dose therapy autotrans-plantation/intensification vs continued standard chemotherapy in multiple myeloma in first remission: results of a non-randomized study from a single institution. Bone Marrow Transplant 2000; 26: 845–9

    PubMed  CAS  Google Scholar 

  63. Lenhoff S, Hjorth M, Holmberg E, et al. Impact on survival of high-dose therapy with autologous stem cell support in patients younger than 60 years with newly diagnosed multiple myeloma: a population-based study. Nordic Myeloma Study Group. Blood 2000; 95: 7–11

    CAS  Google Scholar 

  64. Kyle RA, Lacy MQ. New approaches to treatment of patients with multiple myeloma. In: Rosenberg SA, editor. Principles and practice of biologic therapy of cancer. Vol. 2. New York: Lippincott Williams and Wilkins, 2001: 1–11

    Google Scholar 

  65. Vesole DH, Crowley JJ, Catchatourian R, et al. High-dose melphalan with autotransplantation for refractory multiple myeloma: results of a Southwest Oncology Group phase II trial. J Clin Oncol 1999; 17: 2173–9

    PubMed  CAS  Google Scholar 

  66. Rajkumar SV, Fonseca R, Lacy MQ, et al. Autologous stem cell transplantation for relapsed and primary refractory myeloma. Bone Marrow Transplant 1999; 23: 1267–72

    PubMed  CAS  Google Scholar 

  67. Badros A, Barlogie B, Siegel E, et al. Results of autologous stem cell transplant in multiple myeloma patients with renal failure. Br J Haematol 2001; 114: 822–9

    PubMed  CAS  Google Scholar 

  68. Badros A, Barlogie B, Siegel E, et al. Autologous stem cell transplantation in elderly multiple myeloma patients over the age of 70 years. Br J Haematol 2001; 114: 600–7

    PubMed  CAS  Google Scholar 

  69. Attal M, Harousseau JL. Randomized trial experience of the Intergroupe Francophone du Myelome. Semin Hematol 2001; 38(3): 226–30

    PubMed  CAS  Google Scholar 

  70. Harousseau JL, Attal M, Divine M, et al. Autologous stem cell transplantation after first remission induction treatment in multiple myeloma: a report of the French registry on autologous transplantation in multiple myeloma. Blood 1995; 85(11): 3077–85

    PubMed  CAS  Google Scholar 

  71. Barlogie B, Jagannath S, Desikan KR, et al. Total therapy with tandem transplants for newly diagnosed multiple myeloma. Blood 1999; 93(1): 55–65

    PubMed  CAS  Google Scholar 

  72. Cunningham D, Paz-Ares L, Milan S, et al. High-dose melphalan and autologous bone marrow transplantation as consolidation in previously untreated myeloma. J Clin Oncol 1994; 12(4): 759–63

    PubMed  CAS  Google Scholar 

  73. Lahuerta JJ, Martinez-Lopez J, Grande C, et al. Conditioning regimens in autologous stem cell transplantation for multiple myeloma: a comparative study of efficacy and toxicity from the Spanish Registry for Transplantation in Multiple Myeloma. Br J Haematol 2000; 109: 138–47

    PubMed  CAS  Google Scholar 

  74. Shimoni A, Smith TL, Aleman A, et al. Thiotepa, busulfan, cyclophosphamide (TBC) and autologous hematopoietic transplantation: an intensive regimen for the treatment of multiple myeloma. Bone Marrow Transplant 2001; 27: 821–8

    PubMed  CAS  Google Scholar 

  75. Bjorkstrand BB, Ljungman P, Svensson H, et al. Allogeneic bone marrow transplantation versus autologous stem cell transplantation in multiple myeloma: a retrospective case-matched study from the European Group for Blood and Marrow Transplantation. Blood 1996; 88: 4711–8

    PubMed  CAS  Google Scholar 

  76. Vesole DH, Tricot G, Jagannath S, et al. Autotransplants in multiple myeloma: what have we learned? Blood 1996; 88: 838–47

    PubMed  CAS  Google Scholar 

  77. Desikan R, Barlogie B, Sawyer J, et al. Results of high-dose therapy for 1000 patients with multiple myeloma: durable complete remissions and superior survival in the absence of chromosome 13 abnormalities. Blood 2000; 95: 4008–10

    PubMed  CAS  Google Scholar 

  78. Attal M, Harousseau JL. Autograft and multiple myeloma: experience of the Intergroupe Francais du Myelome. Bull Cancer 2001; 88: 888–92

    PubMed  CAS  Google Scholar 

  79. Attal M, Harousseau JL. Autologous peripheral blood progenitor cell transplantation for multiple myeloma. Baillieres Best Pract Res Clin Haematol 1999; 12: 171–91

    PubMed  CAS  Google Scholar 

  80. Lemoli RM, Fortuna A, Raspadori D, et al. Selection and transplantation of autologous hematopoietic CD34+ cells for patients with multiple myeloma. Leuk Lymphoma 1997; 26 Suppl. 1: 1–11

    PubMed  Google Scholar 

  81. Lemoli RM, Fortuna A, Motta MR, et al. Concomitant mobilization of plasma cells and hematopoietic progenitors into peripheral blood of multiple myeloma patients: positive selection and transplantation of enriched CD34+ cells to remove circulating tumor cells. Blood 1996; 87: 1625–34

    PubMed  CAS  Google Scholar 

  82. Schiller G, Vescio R, Freytes C, et al. Autologous CD34-selected blood progenitor cell transplants for patients with advanced multiple myeloma. Bone Marrow Transplant 1998; 21: 141–5

    PubMed  CAS  Google Scholar 

  83. Stewart AK, Vescio R, Schiller G, et al. Purging of autologous peripheral-blood stem cells using CD34 selection does not improve overall or progression-free survival after high-dose chemotherapy for multiple myeloma: results of a multicenter randomized controlled trial. J Clin Oncol 2001; 19: 3771–9

    PubMed  CAS  Google Scholar 

  84. Goldschmidt H, Bouko Y, Bourhis JH, et al. CD34+ selected PBPCT results in an increased infective risk without prolongation of event free survival in newly diagnosed myeloma: a randomized study from the EBMT. San Francisco (CA): American Society of Hematology, 2000: 96

    Google Scholar 

  85. Smith KJ, Hamilton ES, Waller EK, et al. CD34 selection of peripheral blood progenitor cells for autologous transplant in myeloma and amyloidosis is associated with an increased risk of post transplant varicella zoster. Orlando (FL): American Society of Hematology, 2001: 98

    Google Scholar 

  86. Bomberger C, Singh-Jairam M, Rodey G, et al. Lymphoid reconstitution after autologous PBSC transplantation with FACS-sorted CD34+ hematopoietic progenitors. Blood 1998; 91: 2588–600

    PubMed  CAS  Google Scholar 

  87. Kay NE, Leong TL, Bone N, et al. Blood levels of immune cells predict survival in myeloma patients: results of an Eastern Cooperative Oncology Group phase 3 trial for newly diagnosed multiple myeloma patients. Blood 2001; 98: 23–8

    PubMed  CAS  Google Scholar 

  88. Bjorkstrand B. European Group for Blood and Marrow Transplantation Registry studies in multiple myeloma. Semin Hematol 2001; 38: 219–25

    PubMed  CAS  Google Scholar 

  89. Gahrton G, Svensson H, Bjorkstrand B, et al. Syngeneic transplantation in multiple myeloma: a case-matched comparison with autologous and allogeneic transplantation. European Group for Blood and Marrow Transplantation. Bone Marrow Transplant 1999; 24: 741–5

    CAS  Google Scholar 

  90. Bensinger WI, Demirer T, Buckner CD, et al. Syngeneic marrow transplantation in patients with multiple myeloma. Bone Marrow Transplantation 1996; 18: 527–31

    PubMed  CAS  Google Scholar 

  91. Gahrton G. Allogeneic bone marrow transplantation in multiple myeloma. Pathol Biol (Paris) 1999; 47: 188–91

    CAS  Google Scholar 

  92. Bensinger WI. Hematopoietic cell transplantation for multiple myeloma. Cancer Control 1998; 5: 235–42

    PubMed  Google Scholar 

  93. Gahrton G, Svensson H, Cavo M, et al. Progress in allogeneic bone marrow and peripheral blood stem cell transplantation for multiple myeloma: a comparison between transplants performed 1983–93 and 1994–8 at European Group for Blood and Marrow Transplantation centres. Br J Haematol 2001; 113: 209–16

    PubMed  CAS  Google Scholar 

  94. Badros A, Barlogie B, Morris C, et al. High response rate in refractory and poor-risk multiple myeloma after allotrans-plantation using a nonmyeloablative conditioning regimen and donor lymphocyte infusions. Blood 2001; 97: 2574–9

    PubMed  CAS  Google Scholar 

  95. Alyea E, Weiler E, Schlossman R, et al. T-cell-depleted allogeneic bone marrow transplantation followed by donor lymphocyte infusion in patients with multiple myeloma: induction of graft-versus-myeloma effect. Blood 2001; 98: 934–9

    PubMed  CAS  Google Scholar 

  96. Badros A, Barlogie B, Siegel E, et al. Improved outcome of allogeneic transplantation in high-risk multiple myeloma patients after nonmyeloablative conditioning. J Clin Oncol 2002; 20(3): 1295–303

    PubMed  Google Scholar 

  97. Maloney D, Sahebi F, Stockerl-Goldstein KE, et al. Combining an allogeneic graft versus myeloma effect with high-dose autologous stem cell rescue in the treatment of multiple myeloma. Orlando (FL): American Society of Hematology, 2001: 98

    Google Scholar 

  98. Khouri IF, Keating M, Korbling M, et al. Transplant-lite: induction of graft-versus-malignancy using fludarabine-based nonablative chemotherapy and allogeneic blood progenitor-cell transplantation as treatment for lymphoid malignancies. J Clin Oncol 1998; 16: 2817–24

    PubMed  CAS  Google Scholar 

  99. Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases. Blood 1998; 91: 756–63

    PubMed  CAS  Google Scholar 

  100. Helg C, Starobinski M, Jeannet M, et al. Donor lymphocyte infusion for the treatment of relapse after allogeneic hematopoietic stem cell transplantation. Leuk Lymphoma 1998; 29: 301–13

    PubMed  CAS  Google Scholar 

  101. Lokhorst HM, Schattenberg A, Cornelissen JJ, et al. Donor lymphocyte infusions for relapsed multiple myeloma after allogeneic stem-cell transplantation: predictive factors for response and long-term outcome. J Clin Oncol 2000; 18: 3031–7

    PubMed  CAS  Google Scholar 

  102. Tricot G, Vesole DH, Jagannath S, et al. Graft-versus-myeloma effect: proof of principle. Blood 1997; 87(3): 1196–8

    Google Scholar 

  103. Lokhorst HM, Schattenberg A, Cornelissen JJ, et al. Donor leukocyte infusions are effective in relapsed multiple myeloma after allogeneic bone marrow transplantation. Blood 1997; 90(10): 4206–11

    PubMed  CAS  Google Scholar 

  104. Kroger N, Kruger W, Renges H, et al. Donor lymphocyte infusion enhances remission status in patients with persistent disease after allografting for multiple myeloma. Br J Haematol 2001; 112(2): 421–3

    PubMed  CAS  Google Scholar 

  105. Mehta J, Singhai S. Graft-versus-myeloma. Bone Marrow Transplant 1998; 22: 835–43

    PubMed  CAS  Google Scholar 

  106. Salama M, Nevill T, Marcellus D, et al. Donor leukocyte infusions for multiple myeloma. Bone Marrow Transplant 2000; 26: 1179–84

    PubMed  CAS  Google Scholar 

  107. Bellucci R, Alyea EP, Weller E, et al. Immunologic effects of prophylactic donor lymphocyte infusion after allogeneic marrow transplantation for multiple myeloma. Blood 2002; 99(12): 4610–7

    PubMed  CAS  Google Scholar 

  108. Bendandi M, Gocke CD, Kobrin CB, et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma. Nat Med 1999; 5: 1171–7

    PubMed  CAS  Google Scholar 

  109. Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med 1996; 2: 52–8

    PubMed  CAS  Google Scholar 

  110. Ruffini PA, Kwak LW. Immunotherapy of multiple myeloma. Semin Hematol 2001; 38: 260–7

    PubMed  CAS  Google Scholar 

  111. Reichardt VL, Okada CY, Liso A, et al. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma: a feasibility study. Blood 1999; 93: 2411–9

    PubMed  CAS  Google Scholar 

  112. Qing Y, Desikan R, Barlogie B, et al. Optimizing dendritic cell-based immunotherapy in multiple myeloma. Br J Hematol 2002; 117: 297–305

    Google Scholar 

  113. Schlienger K, Craighead K, Lee KP, et al. Efficient priming of protein antigen-specific human CD4(+) T cells by monocyte-derived dendritic cells. Blood 2000; 96(10): 3490–8

    PubMed  CAS  Google Scholar 

  114. Butch AW, Kelly KA, Munshi NC. Dedritic cells derived from multiple myeloma patients efficiently internalize different classes of myeloma protein. Exp Hematol 2001; 29: 85–92

    PubMed  CAS  Google Scholar 

  115. Li Y, Bendandi M, Deng Y, et al. Tumor-specific recognition of human myeloma cells by idiotype-induced CD8+ T cells. Blood 2000; 96(8): 2828–33

    PubMed  CAS  Google Scholar 

  116. Wen YJ, Min R, Tricot G, et al. Tumor lysate specific cytotoxic T lymphocytes in multiple myeloma: promising effector cells for immunotherapy. Blood 2002; 99(9): 3280–5

    PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Dr Lonial is supported by a career development award from the Lymphoma Research Foundation and is on the speakers bureau for Berlex, Celgene and Millenium. Dr Heffner is on the speakers bureau for Celgene and Millenium. The authors are most grateful to Debbie Quave and Jill Cluesmann for their excellent administrative assistance.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Leonard T. Heffher Jr.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Heffher, L.T., Lonial, S. Breakthroughs in the Management of Multiple Myeloma. Drugs 63, 1621–1636 (2003). https://doi.org/10.2165/00003495-200363160-00001

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-200363160-00001

Keywords

Navigation