Novel Approaches to the Management of Myeloma

Article

Standard therapy for multiple myeloma, which accounts for 10% ofall hematologic malignancies, has been autologous stem cell transplantation(ASCT), alkylator-based chemotherapy, and corticosteroids. Severaladvances have been made in the treatment of multiple myelomaover the past decade, especially the arrival of new, active agents suchas thalidomide (Thalomid), bortezomib (Velcade), and lenalidomide(Revlimid). These have shown significant clinical activity as singleagents. Trials are ongoing to incorporate these new agents into thevarious stages of treatment and to combine them with other effectivetreatment modalities, including ASCT.

Standard therapy for multiple myeloma, which accounts for 10% of all hematologic malignancies, has been autologous stem cell transplantation (ASCT), alkylator-based chemotherapy, and corticosteroids. Several advances have been made in the treatment of multiple myeloma over the past decade, especially the arrival of new, active agents such as thalidomide (Thalomid), bortezomib (Velcade), and lenalidomide (Revlimid). These have shown significant clinical activity as single agents. Trials are ongoing to incorporate these new agents into the various stages of treatment and to combine them with other effective treatment modalities, including ASCT.

Multiple myeloma accounts for 10% of all hematologic malignancies.[1,2] For many years, autologous stem cell transplantation (ASCT), alkylatorbased chemotherapy, and corticosteroids had been the mainstay of therapy for the disease. Recently, thalidomide (Thalomid), bortezomib (Velcade), and lenalidomide (Revlimid) have emerged as effective single agents, demonstrating significant clinical activity in relapsed and refractory myeloma.[ 3-5] The discovery of these drugs has been accompanied by a growing awareness of the importance of bone marrow microenvironmental changes such as induction of angiogenesis, suppression of cell-mediated immunity, and expression of various adhesion molecules and cytokines in disease progression.[6,7] The present challenge is to determine how best to incorporate thalidomide, bortezomib, and lenalidomide into the therapeutic strategy for myeloma. This article will briefly summarize the efficacy data in myeloma with these three drugs, and discuss how they are being incorporated into standard clinical practice and clinical trials for the treatment of this disease. Thalidomide and Lenalidomide Thalidomide was first tested in myeloma based on the known antiangiogenic properties of the drug coupled with the knowledge that increased angiogenesis occurred in myeloma similar to solid tumors.[8,9] The first trial in the disease was conducted at the University of Arkansas and demonstrated a response rate of 25% in heavily pretreated patients with relapsed and refractory disease.[3] Since then several studies have confirmed the activity of thalidomide in relapsed myeloma, with a response rate of approximately 25% to 35% and a median response duration of 1 year.[10-13] Thalidomide has been effectively combined with dexamethasone (Thal- Dex) in the treatment of both newly diagnosed and relapsed myeloma. Response rates with Thal-Dex are approximately 65% to 70% in newly diagnosed disease and 50% in the relapsed, refractory setting.[14-20] Recent studies suggest that efficacy can be further increased with the addition of alkylating agents.[12,21,22] The usual thalidomide dose is 50 to 200 mg/d. The main disadvantage with this drug is nonhematologic toxicity, such as constipation, sedation, neuropathy, rash, and teratogenicity. Safer and more potent analogs of thalidomide are being developed; lenalidomide is one of the first analogs to enter clinical trials. Lenalidomide has shown promis-ing antimyeloma activity in two phase I trials.[5,23] In a subsequent phase II trial of 83 evaluable patients with relapsed myeloma, 24% responded to therapy. This trial included several patients who had previously failed thalidomide. Two large phase III trials in relapsed myeloma comparing lenalidomide plus dexamethasone vs dexamethasone alone have been completed. An independent data monitoring committee has recommended closure of these trials because of significantly superior time to progression with lenalidomide plus dexamethasone compared to dexamethasone alone. Detailed results are expected later this year.

Typical dosing for myeloma is 25 to 30 mg/d on days 1 through 21 of a 28-day cycle. The most common adverse effects are thrombocytopenia and neutropenia. Side effects such as sedation, constipation, and neuropathy appear to be uncommon with lenalidomide. Bortezomib Bortezomib is a boronic acid dipeptide, and the first proteasome inhibitor to enter clinical trials.[24,25] Normally, cellular proteins targeted for destruction are first tagged by the addition of ubiquitin molecules. Ubiquitinated proteins are then identified and degraded in the proteasome complex. Bortezomib is a specific inhibitor of the proteasome complex, and the resultant inhibition of this catabolic pathway leads to cell-cycle arrest and apoptosis. Based on promising preclinical and phase I data, a large phase II study of bortezomib was undertaken in relapsed and refractory myeloma. Of 193 evaluable patients, 27% achieved a significant response to therapy. In another trial, which tested two different dosing schedules, responses were seen in 33% and 50% of patients at the 1.0 mg/m2 (28 patients) and 1.3 mg/m2 (26 patients) dosing levels, respectively.[ 26] Preliminary results from a large randomized trial indicate superior time to progression and overall survival with bortezomib compared to dexamethasone in relapsed, refractory myeloma.[27] The starting dose is 1.3 mg/m2 given on days 1, 4, 8, and 11 every 21 days. The most common side effects are gastrointestinal, cytopenias, fatigue, and peripheral neuropathy. Incorporation of New Agents Into the Treatment of Myeloma The treatment of myeloma usually consists of various phases, such as induction, consolidation-typically with ASCT-maintenance, and treatment of relapsed and refractory disease. Clinical trials incorporating these new agents have been designed to address each phase of therapy. Further, trials are ongoing in specific subsets of patients, including those felt not to be transplant candidates because of advanced age or poor performance status, and those felt to have high-risk myeloma. Studies are also ongoing in patients with smoldering myeloma in an attempt to delay progression to symptomatic disease. Newly Diagnosed Myeloma Patients with newly diagnosed myeloma are treated based on whether they are candidates for ASCT, because two published randomized trials have shown that ASCT prolongs survival in myeloma.[28,29] Patients who are candidates for ASCT need to avoid alkylator-based induction to prevent stem cell damage. Four cycles of vincristine, doxorubicin (Adriamycin), and dexamethasone (VAD) had been the typical induction regimen used in this population of patients. But VAD had significant disadvantages, including the need for an indwelling intra-venous line, neurotoxicity from vincristine, cardiotoxicity from doxorubicin, and alopecia. Furthermore, vincristine and doxorubicin have negligible antimyeloma activity as single agents, and most of the efficacy of VAD is derived from dexamethasone alone. These facts, coupled with the significant activity of oral Thal-Dex in newly diagnosed myeloma, have negated the need for VAD as induction therapy. Preliminary results of a randomized trial show superior response rates with Thal-Dex compared to dexamethasone alone. But Thal-Dex is complicated by a higher incidence of grade 3/4 adverse effects than dexamethasone alone. This is especially true of deep-vein thrombosis, for which prophylaxis should be considered in all patients started on Thal- Dex. So how do we decide on the best induction therapy in standard practice outside a clinical trial setting? The main standard and clinical trial options for patients with various subcategories of newly diagnosed myeloma are summarized in Table 1. After considering the risks and benefits for patients proceeding to early transplant, dexamethasone alone may be adequate pretransplant induction if a delay of 1 to 2 months to assess response can be safely incurred, with the plan of adding thalidomide if response is inadequate. Thal-Dex can be reserved for those patients with more aggressive disease such as painful symptoms, large lytic lesions, impending cord compression, hypercalcemia, or renal failure. On the other hand, dexamethasone alone may not be adequate if an early transplant is not planned. Several ongoing trials are seeking to improve upon results with dexamethasone and Thal-Dex for these patients. Lenalidomide plus dexamethasone (Rev-Dex) is being tested in large randomized trials by the Eastern Cooperative Oncology Group (ECOG) and the Southwest Oncology Group (SWOG) in an attempt to improve efficacy and reduce toxicity. Preliminary results from a phase II Mayo Clinic trial indicate response rates greater than 80% with Rev-Dex in newly diagnosed myeloma, with fewer toxicities than previously observed with Thal-Dex.[30] This strategy is particularly appealing to patients who are proceeding to an early transplant and wish a safe and convenient oral induction regimen. Bortezomib alone appears to be active in newly diagnosed myeloma.[ 31,32] The addition of bortezomib to Thal-Dex seems to greatly improve overall and complete response rates,[33] and will appeal to patients and physicians who seek alternatives to transplant as initial therapy, relegating ASCT to a salvage role. All of these strategies are currently in the clinical trial stage and patients should be encouraged to participate in these trials as much as possible in preference to off-study therapy. For the subset of patients not considered to be transplant candidates because of advanced age or poor performance status, advances have been harder to come by. Melphalan (Alkeran) and prednisone (MP) has remained the standard initial treatment for this group for over 3 decades. Ongoing trials are seeking to improve upon this by adding new agents to the classic MP regimen. The addition of thalidomide to MP increased response rates and prolonged progression-free survival in two recent trials; however, no improvement has been seen yet in overall survival.[34,35] Studies are also ongoing with the addition of bortezomib (the MPV regimen) and lenalidomide (the MPR regimen) to MP. Improving the outcome of this group of patients is critically needed, and identifying effective ways of incorporating new agents into the initial treatment strategy is necessary to achieve this goal. Consolidation/Stem Cell Transplantation One of the main questions raised by patients today is whether the arrival of new drugs signals the end of upfront ASCT in the treatment of myeloma. Clearly, well-designed studies are needed to answer this question, and until then ASCT should still be considered in all eligible patients. Autologous stem cell transplantation can, however, be delayed and performed as salvage therapy at the time of relapse in selected patients, provided stem cells are harvested and cryopreserved early in the disease course. At least three randomized trials show that this approach is equivalent to early stem cell transplant.[ 36-38] However, at the Mayo Clinic we still feel it is preferable to proceed with up-front ASCT in eligible patients because it affords the opportunity to consider tandem ASCT, reduces issues related to advancing age and comorbidity, and avoids insurance approval issues. Clinical trials are ongoing to test the addition of new agents to improve the effectiveness of ASCT. For example, the addition of bortezomib to melphalan as the condition regimen is now being tested as a way of improving outcome following ASCT.[39] Alternatively, the ECOG is developing a phase II clinical trial to test a novel nontransplant consolidation strategy following induction therapy. Maintenance Therapy There have been several trials testing various maintenance strategies in myeloma for patients completing chemotherapy or ASCT. Most have been disappointing. Prednisone at 50 mg every other day has shown some benefit in steroid-responsive patients treated without ASCT.[40] Recently thalidomide was shown to prolong progression free survival but not overall survival following ASCT.[41] More data are needed. It would be preferable for patients to enroll in ongoing studies that are testing thalidomide (National Cancer Institute of Canada trial, Clinical Trials Network trial) or lenalidomide (Cancer and Acute Leukemia Group B trial) as maintenance therapy. Treatment of High-Risk Myeloma Patients with deletion 13 or hypodiploidy by karyotype, plasma cell labeling index > 3%, or translocations t(4;14), or t(14;16) have highrisk myeloma and tend to do poorly even with tandem ASCT. These patients should be considered for novel therapeutic strategies. Clinical trials need to be designed separately for these patients; one such trial with bortezomib as initial therapy is currently ongoing in the ECOG. Relapsed Disease All new agents discussed in this article were first identified because of their single-agent activity in relapsed and refractory myeloma. Numerous studies are currently ongoing to develop novel combination approaches. The most well-studied combinations are Thal-Dex and bortezomib plus dexamethasone. The aggressiveness of the relapse and the performance status of the patient often dictate the choice of single- agent vs combination approaches in the relapsed setting. Summary Several advances have been made in the treatment of multiple myeloma over the past decade, especially the arrival of new, active agents. Trials are ongoing to incorporate these new agents into the various stages of treatment and to combine them with other effective treatment modalities, including ASCT. However, more active new drugs are still needed.

Disclosures:

Dr. Rajkumar has received financial support for clinical trials at the Mayo Clinic from Celgene, Millennium, and Entremed.

References:

1. Kyle RA, Rajkumar SV: Multiple myeloma. N Engl J Med 351:1860-1873, 2004.
2. Kyle RA, Rajkumar SV: Plasma cell disorders, in Goldman L, Ausiello D (eds): Cecil Textbook of Medicine, 22nd ed. Philadelphia, W. B. Saunders, 2004.
3. Singhal S, Mehta J, Desikan R, et al: Antitumor activity of thalidomide in refractory multiple myeloma [see comments]. N Engl J Med 341:1565-1571, 1999.
4. Richardson PG, Barlogie B, Berenson J, et al: A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 348:2609- 2617, 2003.
5. Richardson PG, Schlossman RL, Weller E, et al: Immunomodulatory drug CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed multiple myeloma. Blood 100:3063-3067, 2002.
6. Hideshima T, Anderson KC: Molecular mechanisms of novel therapeutic approaches for multiple myeloma. Nat Rev Cancer 2:927- 937, 2002.
7. Rajkumar SV, Mesa RA, Fonseca R, et al: Bone marrow angiogenesis in 400 patients with monoclonal gammopathy of undetermined significance, multiple myeloma, and primary amyloidosis. Clin Cancer Res 8:2210-2216, 2002.
8. D'Amato RJ, Loughnan MS, Flynn E, et al: Thalidomide is an inhibitor of angiogenesis. Proc Natl Acad Sci USA 91:4082-4085, 1994.
9. Vacca A, Ribatti D, Roncali L, et al: Bone marrow angiogenesis and progression in multiple myeloma. Br J Haematol 87:503-508, 1994.
10. 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 98:492-494, 2001.
11. Rajkumar SV, Fonseca R, Dispenzieri A, et al: Thalidomide in the treatment of relapsed multiple myeloma. Mayo Clin Proc 75:897-902, 2000.
12. Dimopoulos MA, Anagnostopoulos A, Weber D: Treatment of plasma cell dyscrasias with thalidomide and its derivatives. J Clin Oncol 21:4444-4454, 2003.
13. Kumar S, Gertz MA, Dispenzieri A, et al: Response rate, durability of response, and survival after thalidomide therapy for relapsed multiple myeloma [comment]. Mayo Clin Proc 78:34-39, 2003.
14. Rajkumar SV, Hayman S, Gertz MA, et al: Combination therapy with thalidomide plus dexamethasone for newly diagnosed myeloma. J Clin Oncol 20:4319-4323, 2002.
15. Weber D, Rankin K, Gavino M, et al: Thalidomide alone or with dexamethasone for previously untreated multiple myeloma. J Clin Oncol 21:16-19, 2003.
16. Weber DM, Rankin K, Gavino M, et al: Thalidomide with dexamethasone for resistant multiple myeloma (abstract 719). Blood 96:167a, 2000.
17. Palumbo A, Giaccone L, Bertola A, et al: Low-dose thalidomide plus dexamethasone is an effective salvage therapy for advanced myeloma. Haematologica 86:399-403, 2001.
18. Dimopoulos MA, Zervas K, Kouvatseas G, et al. Thalidomide and dexamethasone combination for refractory multiple myeloma.[ comment]. Ann Oncol 12:991-995, 2001.
19. Anagnostopoulos A, Weber D, Rankin K, et al: Thalidomide and dexamethasone for resistant multiple myeloma. Br J Haematol 121:768-771, 2003.
20. Rajkumar SV, Blood E, Vesole DH, et al: Thalidomide plus dexamethasone versus dexamethasone alone in newly diagnosed multiple myeloma (E1A00): Results of a phase III trial coordinated by the Eastern Cooperative Oncology Group (abstract 205). Blood 104:2004.
21. Garcia-Sanz R, Gonzalez-Fraile MI, Sierra M, et al: The combination of thalidomide, cyclophosphamide and dexamethasone (ThaCyDex) is feasible and can be an option for relapsed/refractory multiple myeloma. Hematol J 3:43-48, 2002.
22. Srkalovic G, Elson P, Trebisky B, et al: Use of melphalan, thalidomide, and dexamethasone in treatment of refractory and relapsed multiple myeloma. Med Oncol 19:219-226, 2002.
23. Zangari M, Tricot G, Zeldis J, et al: Results of phase I study of CC-5013 for the treatment of multiple myeloma (MM) patients who relapse after high dose chemotherapy (HDCT) (abstract 3226). Blood 775a, 2001.
24. Adams J: Proteasome inhibitors as new anticancer drugs. Curr Opin Oncol 14:628-634, 2002.
25. Rajkumar SV, Richardson PG, Hideshima T, et al: Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol 23:630-639, 2005.
26. Berenson JR, Jagannath S, Barlogie B, et al: Experience with long-term therapy using the proteasome inhibitor, bortezomib, in advanced multiple myeloma (MM) (abstract 2337). Proc Am Soc Clin Oncol 22:581, 2003.
27. Richardson P, Sonneveld P, Schuster MW, et al: Bortezomib vs. dexamethasone in relapsed multiple myeloma: A phase 3 randomized study (abstract 6511). Proc Am Soc Clin Oncol 23:558, 2004.
28. 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 335:91- 97, 1996.
29. Child JA, Morgan GJ, Davies FE, et al: High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med 348:1875-1883, 2003.
30. Rajkumar SV, Hayman SR, Lacy MQ, et al: Combination therapy with CC-5013 (lenalidomide; Revlimid) plus dexamethasone (Rev/Dex) for newly diagnosed myeloma (MM) (abstract 331). Blood 104:2004.
31. Jagannath S, Durie B, Wolf JL, et al: A phase 2 study of bortezomib as first-line therapy in patients with multiple myeloma (abstract 333). Blood 104:2004.
32. Richardson PG, Chanan-Khan A, Schlossman RL, et al: Phase II trial of single agent bortezomib (Velcade) in patients with previously untreated multiple myeloma (MM) (abstract 336). Blood 104:2004.
33. Alexanian R, Wang LM, Weber DM, et al: VTD (Velcade, thalidomide, dexamethasone) as primary therapy for newly-diagnosed multiple myeloma (abstract 210). Blood 104:2004.
34. Facon T, Mary JY, Hulin C, et al: Randomized clinical trial comparing melphalanprednisone (MP), MP-thalidomide (MP-Thal) and high-dose therapy using melphalan 100 mg/m² (MEL100) for newly diagnosed myeloma patients aged 65-75 years. Interim analysis of the IFM 99-06 trial on 350 patients (abstract 206). Blood 104:2004.
35. Palumbo A, Bertola A, Musto P, et al: A prospective randomized trial of oral melphalan, prednisone, thalidomide (MPT) vs oral melphalan, prednisone (MP): An interim analysis (abstract 207). Blood 104:2004.
36. 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 92:3131-3136, 1998.
37. Facon T, Mary JY, Harousseau JL, et al: Front-line or rescue autologous bone marrow transplantation (ABMT) following a first course of high dose melphalan (HDM) in multiple myeloma (MM). Preliminary results of a prospective randomized trial (CIAM) protocol. Blood 88(suppl 1):685a, 1996.
38. Barlogie B, Kyle R, Anderson K, et al: Comparable survival in multiple myeloma (MM) with high dose therapy (HDT) employing MEL 140 mg/m2 + TBI 12 Gy autotransplants versus standard dose therapy with VBMCP and no benefit from interferon (IFN) maintenance: Results of Intergroup trial S9321 (abstract 135). Blood 102:42a, 2003.
39. Hollmig K, Stover J, Talamo G, et al: Addition of bortezomib (Velcade) to high dose melphalan (Vel-Mel) as an effective conditioning regimen with autologous stem cell support in multiple myeloma (MM) (abstract 929). Blood 104:2004.
40. Berenson JR, Crowley JJ, Grogan TM, et al: Maintenance therapy with alternate-day prednisone improves survival in multiple myeloma patients. Blood 99:3163-3168, 2002.
41. Attal M, Harousseau JL, Leyvraz L, et al: Maintenance treatment with thalidomide after autologous transplantation for myeloma: First analysis of a prospective randomized study of the Intergroupe Francophone du Myelome (IFM 99 02) (abstract 535). Blood 104:2004.

Related Videos
Frederick “Eric” Arnold, PhD
Genovefa (Zenia) Papanicolaou, MD, an infectious diseases specialist at Memorial Sloan Kettering Cancer Center
Jeffrey Chamberlain, PhD, on Exciting New Research at MDA 2024
Alan Beggs, PhD, on Challenges in Therapeutic Development for Rare Diseases
Akshay Sharma, MBBS, a bone marrow transplant physician at St. Jude Children’s Research Hospital
PJ Brooks, PhD
John DiPersio, MD, PhD, the director of the Center for Gene and Cellular Immunotherapy at Washington University School of Medicine
© 2024 MJH Life Sciences

All rights reserved.