51
|
Ludwig H, Adam Z, Hajek R, Greil R, Tóthová E, Keil F, Autzinger EM, Thaler J, Gisslinger H, Lang A, Egyed M, Womastek I, Zojer N. Light Chain–Induced Acute Renal Failure Can Be Reversed by Bortezomib-Doxorubicin-Dexamethasone in Multiple Myeloma: Results of a Phase II Study. J Clin Oncol 2010; 28:4635-41. [DOI: 10.1200/jco.2010.28.1238] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To assess the efficacy of bortezomib-doxorubicin-dexamethasone (BDD) therapy in patients with multiple myeloma with light chain–induced acute renal failure. Patients and Methods Sixty-eight patients with light chain–induced acute renal failure and glomerular filtration rate (GFR) less than 50 mL/min received bortezomib (1.0 mg/m2 on days 1, 4, 8, and 11), doxorubicin (9 mg/m2 on days 1 and 4), and dexamethasone (40 mg on days 1, 4, 8, and 11); if well tolerated after two cycles, bortezomib could be increased to 1.3 mg/m2 and doxorubicin administered on days 1, 4, 8, and 11. Results By intent-to-treat analysis a myeloma response was obtained in 72% of 18 previously and 50 not previously treated patients (complete response [CR]/near CR [nCR], 38%; very good partial response [VGPR], 15%; partial response [PR], 13%; minor response [MR], 6%). Renal response was achieved in 62% of patients (renal CR, 31%; renal PR, 7%; renal MR, 24%). Median GFR increased from 20.5 to 48.4 mL/min. GFR improvement correlated with tumor response; the greatest increase to 59.6 mL/min was seen in the group of patients with CR/nCR/VGPR. Median progression-free survival was 12.1 months. One- and 2-year survival rates were 72% and 58%, respectively. Survival did not differ between patients with and without renal response but was inferior in previously treated patients (P < .001). In multivariate analysis, baseline GFR and tumor response correlated with renal response, and pretreatment status, lactate dehydrogenase, and myeloma response correlated with survival. The most common grade 3 or 4 toxicities were infection (19.1%), thrombocytopenia (14.7%), neutropenia (14.7%), fatigue/weakness (10.3%), and polyneuropathy (8.8%). Conclusion BDD induced a high rate of myeloma and renal responses, and treatment was well tolerated.
Collapse
Affiliation(s)
- Heinz Ludwig
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Zdenek Adam
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Roman Hajek
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Richard Greil
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Elena Tóthová
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Felix Keil
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Eva Maria Autzinger
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Josef Thaler
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Heinz Gisslinger
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Alois Lang
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Miklós Egyed
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Irene Womastek
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| | - Niklas Zojer
- From the Wilhelminenspital; University Hospital Vienna, Vienna; Private Medical University Hospital Salzburg, Salzburg; Hospital Leoben, Leoben; Hospital Wels-Grieskirchen, Grieskirchen; Hospital Feldkirch, Feldkirch, Austria; Masaryk University; Faculty Hospital Brno, Brno, Czech Republic; University Hospital L. Pasteur, Kosice, Slovakia; and Kaposi Mór Teaching Hospital, Kaposvar, Hungary
| |
Collapse
|
52
|
van de Donk NWCJ, Lokhorst HM, Dimopoulos M, Cavo M, Morgan G, Einsele H, Kropff M, Schey S, Avet-Loiseau H, Ludwig H, Goldschmidt H, Sonneveld P, Johnsen HE, Bladé J, San-Miguel JF, Palumbo A. Treatment of relapsed and refractory multiple myeloma in the era of novel agents. Cancer Treat Rev 2010; 37:266-83. [PMID: 20863623 DOI: 10.1016/j.ctrv.2010.08.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 08/21/2010] [Accepted: 08/25/2010] [Indexed: 12/22/2022]
Abstract
The introduction of the Immunomodulatory drugs (IMiDs) and proteasome inhibitors, used either as a single-agent or combined with classic anti-myeloma therapies, has improved the outcome for patients with relapsed myeloma. However, there is currently no generally accepted standard treatment for relapsed/refractory myeloma patients, partly because of the absence of trials comparing the efficacy of the novel agents in relapsed/refractory myeloma. Choice of a new treatment regimen depends on both patient and disease-specific characteristics. A lenalidomide-based regimen is the first choice in patients with neuropathy, while bortezomib has the highest efficacy in patients with renal insufficiency and is not associated with increased risk of thromboembolism. A second autologous stem cell transplantation (auto-SCT) can be applied in patients with a progression-free period of ≥ 18-24 months after the first auto-SCT. In high-risk relapse such as occurring early after auto-SCT consolidation with allogeneic SCT can be considered. In this review we provide an overview of the various salvage regimens and give recommendations for treatment of patients with relapsed/refractory myeloma in different clinical settings.
Collapse
|
53
|
Shen Y, Zhou X, Wang Z, Yang G, Jiang Y, Sun C, Wang J, Tong Y, Guo H. Coagulation profiles and thromboembolic events of bortezomib plus thalidomide and dexamethasone therapy in newly diagnosed multiple myeloma. Leuk Res 2010; 35:147-51. [PMID: 20832859 DOI: 10.1016/j.leukres.2010.08.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Revised: 07/05/2010] [Accepted: 08/15/2010] [Indexed: 11/17/2022]
Abstract
Patients with multiple myeloma (MM) are at relatively high risk of developing thromboembolic event (TEE), especially during treatment with immunomodulatory agents. We characterized coagulation profiles and evaluate the incidence of TEE associated with the combination therapy of bortezomib-thalidomide-dexamethasone (VTD) in Chinese patients with newly diagnosed MM. The results indicated that the platelet count and platelet aggregation induced by the agonists were decreased after a short exposure to bortezomib in vivo. The incidence of TEE was low in VTD therapy for an overall rate of 3%. We do not recommend routine thromboprophylaxis for VTD therapy in Chinese patients with MM.
Collapse
Affiliation(s)
- Yunfeng Shen
- Department of Hematology, Wuxi People's Hospital, Nanjing Medical University, 299 Qingyang Road, Wuxi 214023, China
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Richardson PG, Weller E, Lonial S, Jakubowiak AJ, Jagannath S, Raje NS, Avigan DE, Xie W, Ghobrial IM, Schlossman RL, Mazumder A, Munshi NC, Vesole DH, Joyce R, Kaufman JL, Doss D, Warren DL, Lunde LE, Kaster S, Delaney C, Hideshima T, Mitsiades CS, Knight R, Esseltine DL, Anderson KC. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood 2010; 116:679-86. [PMID: 20385792 PMCID: PMC3324254 DOI: 10.1182/blood-2010-02-268862] [Citation(s) in RCA: 674] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 03/30/2010] [Indexed: 12/18/2022] Open
Abstract
This phase 1/2 study is the first prospective evaluation of lenalidomide-bortezomib-dexamethasone in front-line myeloma. Patients (N = 66) received 3-week cycles (n = 8) of bortezomib 1.0 or 1.3 mg/m(2) (days 1, 4, 8, 11), lenalidomide 15 to 25 mg (days 1-14), and dexamethasone 40 or 20 mg (days 1, 2, 4, 5, 8, 9, 11, 12). Responding patients proceeded to maintenance or transplantation. Phase 2 dosing was determined to be bortezomib 1.3 mg/m(2), lenalidomide 25 mg, and dexamethasone 20 mg. Most common toxicities included sensory neuropathy (80%) and fatigue (64%), with only 27%/2% and 32%/3% grade 2/3, respectively. In addition, 32% reported neuropathic pain (11%/3%, grade 2/3). Grade 3/4 hematologic toxicities included lymphopenia (14%), neutropenia (9%), and thrombocytopenia (6%). Thrombosis was rare (6% overall), and no treatment-related mortality was observed. Rate of partial response was 100% in both the phase 2 population and overall, with 74% and 67% each achieving very good partial response or better. Twenty-eight patients (42%) proceeded to undergo transplantation. With median follow-up of 21 months, estimated 18-month progression-free and overall survival for the combination treatment with/without transplantation were 75% and 97%, respectively. Lenalidomide-bortezomib-dexamethasone demonstrates favorable tolerability and is highly effective in the treatment of newly diagnosed myeloma. This study is registered at http://clinicaltrials.gov as NCT00378105.
Collapse
|
55
|
Kaufman JL, Nooka A, Vrana M, Gleason C, Heffner LT, Lonial S. Bortezomib, thalidomide, and dexamethasone as induction therapy for patients with symptomatic multiple myeloma: a retrospective study. Cancer 2010; 116:3143-51. [PMID: 20564642 DOI: 10.1002/cncr.25143] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND This single-center retrospective study determined the efficacy of bortezomib, thalidomide, and dexamethasone (BTD) as induction for patients with multiple myeloma (MM) who were eligible for autologous stem cell transplantation (ASCT). METHODS Patients with symptomatic MM who had received BTD induction before stem cell collection at Winship Cancer Institute were included. BTD induction comprised up to 8 3-week cycles of bortezomib 1.3 mg/m(2) on Days 1, 4, 8, and 11; thalidomide 100 mg daily; and dexamethasone 40 mg on Days 1 through 4 and Days 9 through 12. Stem cell mobilization involved granulocyte-colony-stimulating factor and/or cyclophosphamide. Response was assessed according to European Group for Blood and Marrow Transplantation criteria. RESULTS Review of medical records identified 44 eligible patients (34 patients who were treated in the front-line setting and 10 patients who were treated for recurrent disease) who received a median of 4 BTD cycles. The overall response rate (ORR) was 91%, which included a greater than or equal to very good partial response (> or = VGPR) rate of 57% (including 20% stringent complete responses/complete response [sCR/CR] rate). In front-line patients, the ORR was 94%, which included a 56% > or = VGPR rate (24% sCR/CR). The median CD34-positive stem cell collection was 10.67 x 10(6)/kg. The ORR after ASCT in 34 patients who were evaluable for response was 100%, including a 76% > or = VGPR rate (53% sCR/CR). Among all 44 patients, the median progression-free survival (PFS) was 27.4 months. The median overall survival (OS) was not reached after a median follow-up of 25 months, and the 2-year OS rate was 82%. There were no significant differences in PFS (27.4 months vs 23.5 months) or in 2-year survival (80% vs 90%) between patients who did and did not undergo ASCT, respectively. Twenty patients (45%) developed neuropathy, including 4 (9%) with grade 3 neuropathy episodes, and 1 patient developed deep vein thrombosis. CONCLUSIONS BTD was highly effective and well tolerated as induction for MM patients who were eligible for ASCT. Long-term outcomes appeared to be similar with or without ASCT consolidation.
Collapse
Affiliation(s)
- Jonathan L Kaufman
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | |
Collapse
|
56
|
Richardson PG, Badros AZ, Jagannath S, Tarantolo S, Wolf JL, Albitar M, Berman D, Messina M, Anderson KC. Tanespimycin with bortezomib: activity in relapsed/refractory patients with multiple myeloma. Br J Haematol 2010; 150:428-37. [PMID: 20618338 DOI: 10.1111/j.1365-2141.2010.08264.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tanespimycin (17-allylamino-17-demethoxygeldanamycin, 17-AAG) disrupts heat shock protein 90 (HSP90), a key molecular chaperone for signal transduction proteins critical to myeloma growth, survival and drug resistance. In previous studies, tanespimycin monotherapy was well tolerated and active in heavily pretreated patients with relapsed/refractory multiple myeloma (MM). Preclinical data have shown antitumour synergy between tanespimycin and bortezomib, with more pronounced intracellular accumulation of ubiquitinated proteins than either drug alone, an effect attributed to the synergistic suppression of chymotryptic activity in the 20S proteasome. HSP70 induction has been observed in all Phase 1 tanespimycin studies in which it has been measured, with several separate reports of HSP70 overexpression protecting against peripheral nerve injury. In this Phase 2, open-label multicentre study, we compared 1.3 mg/m2 bortezomib + three doses of tanespimycin: 50, 175 and 340 mg/m2 in heavily pretreated patients with relapsed and refractory MM and measured HSP70 expression and proteasome activity levels in plasma of treated patients. The study was closed prematurely for resource-based reasons, precluding dose comparison. Nonetheless, antitumour activity was observed, with promising response rates and promising severity of peripheral neuropathy.
Collapse
|
57
|
Uaprasert N, Voorhees PM, Mackman N, Key NS. Venous thromboembolism in multiple myeloma: Current perspectives in pathogenesis. Eur J Cancer 2010; 46:1790-9. [PMID: 20385482 DOI: 10.1016/j.ejca.2010.03.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 03/01/2010] [Accepted: 03/11/2010] [Indexed: 12/11/2022]
|
58
|
Dimopoulos MA, Chen C, Kastritis E, Gavriatopoulou M, Treon SP. Bortezomib as a Treatment Option in Patients With Waldenström Macroglobulinemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2010; 10:110-7. [DOI: 10.3816/clml.2010.n.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
59
|
Reece DE, Sullivan D, Lonial S, Mohrbacher AF, Chatta G, Shustik C, Burris H, Venkatakrishnan K, Neuwirth R, Riordan WJ, Karol M, von Moltke LL, Acharya M, Zannikos P, Keith Stewart A. Pharmacokinetic and pharmacodynamic study of two doses of bortezomib in patients with relapsed multiple myeloma. Cancer Chemother Pharmacol 2010; 67:57-67. [PMID: 20306195 DOI: 10.1007/s00280-010-1283-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 02/09/2010] [Indexed: 11/27/2022]
Abstract
PURPOSE Characterize bortezomib pharmacokinetics/pharmacodynamics in relapsed myeloma patients after single and repeat intravenous administration at two doses. METHODS Forty-two patients were randomized to receive bortezomib 1.0 or 1.3 mg/m(2), days 1, 4, 8, 11, for up to eight 21-day treatment cycles (n = 21, each dose group). Serial blood samples for pharmacokinetic/pharmacodynamic analysis were taken on days 1 and 11, cycles 1 and 3. Observational efficacy and safety data were collected. RESULTS Twelve patients in each dose group were evaluable for pharmacokinetics/pharmacodynamics. Plasma clearance decreased with repeat dosing (102-112 L/h for first dose; 15-32 L/h following repeat dosing), with associated increases in systemic exposure and terminal half-life. Systemic exposures of bortezomib were similar between dose groups considering the relatively narrow dose range and the observed pharmacokinetic variability, although there was no readily apparent deviation from dose-proportionality. Blood 20S proteasome inhibition profiles were similar between groups with mean maximum inhibition ranging from 70 to 84% and decreasing toward baseline over the dosing interval. Response rate (all 42 patients) was 50%, including 7% complete responses. The safety profile was consistent with the predictable and manageable profile previously established; data suggested milder toxicity in the 1.0 mg/m(2) group. CONCLUSIONS Bortezomib pharmacokinetics change with repeat dose administration, characterized by a reduction in plasma clearance and associated increase in systemic exposure. Bortezomib is pharmacodynamically active and tolerable at 1.0 and 1.3 mg/m(2) doses, with recovery toward baseline blood proteasome activity over the dosing interval following repeat dose administration, supporting the current clinical dosing regimen.
Collapse
Affiliation(s)
- Donna E Reece
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Richards T, Weber D. Advances in treatment for relapses and refractory multiple myeloma. Med Oncol 2010; 27 Suppl 1:S25-42. [PMID: 20213220 DOI: 10.1007/s12032-009-9407-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 12/22/2009] [Indexed: 12/22/2022]
Abstract
Recent advances in the treatment of multiple myeloma have resulted in improved response rates and overall survival in patients with multiple myeloma. These advances are largely due to thalidomide-, lenalidomide-, and bortezomib-based combinations that have improved response rates, not only in patients with untreated disease, but also in those with relapsed and/or refractory myeloma, in some cases producing response rates up to 85%. Eventually, however, nearly all patients relapse, emphasizing a continuing role for the introduction of investigational agents that overcome drug resistance. This article will review the current role for thalidomide, lenalidomide, and bortezomib-based combinations, as well as some preliminary findings for promising investigational agents currently in clinical trials for patients with relapsed and/or refractory disease.
Collapse
|
61
|
Ludwig H, Beksac M, Bladé J, Boccadoro M, Cavenagh J, Cavo M, Dimopoulos M, Drach J, Einsele H, Facon T, Goldschmidt H, Harousseau JL, Hess U, Ketterer N, Kropff M, Mendeleeva L, Morgan G, Palumbo A, Plesner T, San Miguel J, Shpilberg O, Sondergeld P, Sonneveld P, Zweegman S. Current multiple myeloma treatment strategies with novel agents: a European perspective. Oncologist 2010; 15:6-25. [PMID: 20086168 PMCID: PMC3227886 DOI: 10.1634/theoncologist.2009-0203] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The treatment of multiple myeloma (MM) has undergone significant developments in recent years. The availability of the novel agents thalidomide, bortezomib, and lenalidomide has expanded treatment options and has improved the outcome of patients with MM. Following the introduction of these agents in the relapsed/refractory setting, they are also undergoing investigation in the initial treatment of MM. A number of phase III trials have demonstrated the efficacy of novel agent combinations in the transplant and nontransplant settings, and based on these results standard induction regimens are being challenged and replaced. In the transplant setting, a number of newer induction regimens are now available that have been shown to be superior to the vincristine, doxorubicin, and dexamethasone regimen. Similarly, in the front-line treatment of patients not eligible for transplantation, regimens incorporating novel agents have been found to be superior to the traditional melphalan plus prednisone regimen. Importantly, some of the novel agents appear to be active in patients with high-risk disease, such as adverse cytogenetic features, and certain comorbidities, such as renal impairment. This review presents an overview of the most recent data with these novel agents and summarizes European treatment practices incorporating the novel agents.
Collapse
Affiliation(s)
- Heinz Ludwig
- Department of Medicine, Wilhelminenspital, Montleartstr. 37, 1160 Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Lonial S. Relapsed multiple myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2010; 2010:303-309. [PMID: 21239810 DOI: 10.1182/asheducation-2010.1.303] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Advances in treatment options for patients with multiple myeloma have made a significant impact on the overall survival of patients and have helped achieve levels of response and duration of remission previously not achievable with standard chemotherapy-based approaches. These improvements are due, in large part, to the development of the novel agents thalidomide, bortezomib, and lenalidomide, each of which has substantial single-agent activity. In addition, a large number of second-generation agents are also in clinical development, such that the repertoire of available treatment options continues to expand. To better interpret clinical trials performed in the relapsed setting, it is important that definitions of relapse categories are used to help better pinpoint the specific benefit for a given therapy, especially in the combination therapy setting as it aids in determining if ongoing work should be continued or abandoned for a given new agent. Insights from preclinical modeling and in vitro work have identified several new combinations, new targets and second- or third-generation versions of existing targets that hold great promise in the setting of relapsed myeloma. Combinations of thalidomide, bortezomib, and lenalidomide with conventional agents or among each other have resulted in enhanced response rates and efficacy. Clinical trials of agents such as carfilzomib, pomalidomide, vorinostat, panobinostat, and elotuzomab are just a few of the many exciting new compounds that are being tested in phase 1 and phase 2 clinical trials for relapsed patients. Further clinical and translational testing are critical to better understanding how best to combine these new agents, as well as identifying patient populations that may best benefit from treatment with these developing new agents.
Collapse
Affiliation(s)
- Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
63
|
Abstract
PURPOSE OF REVIEW The present review updates treatment of neutropenia from articles published from January 2008 through April 2009. RECENT FINDINGS Chemotherapy-induced neutropenia occurs most commonly in the first cycle of treatment. Older patients, patients with multiple comorbidities, and those receiving more myelotoxic drugs are prone to develop neutropenia and its complications. Current guidelines recommend the prophylactic use of the myeloid growth factors for the first cycle of chemotherapy for patients with more than a 20% risk of febrile neutropenia. Meta analysis from randomized trials shows that granulocyte colony-stimulating factor prophylaxis is associated with patients receiving more intensive chemotherapy, having better survival, but also having a higher risk of secondary acute myeloid leukemia. Antibiotics are standard treatment of febrile neutropenia and are increasingly used for prophylaxis in 'low-risk' patients. SUMMARY The myeloid growth factor granulocyte colony-stimulating factor has radically changed our approach to the prevention of febrile neutropenia. Antibiotics remain the mainstay of treatment of febrile neutropenia.
Collapse
|
64
|
Richardson PG, Weller E, Jagannath S, Avigan DE, Alsina M, Schlossman RL, Mazumder A, Munshi NC, Ghobrial IM, Doss D, Warren DL, Lunde LE, McKenney M, Delaney C, Mitsiades CS, Hideshima T, Dalton W, Knight R, Esseltine DL, Anderson KC. Multicenter, phase I, dose-escalation trial of lenalidomide plus bortezomib for relapsed and relapsed/refractory multiple myeloma. J Clin Oncol 2009; 27:5713-9. [PMID: 19786667 PMCID: PMC2799050 DOI: 10.1200/jco.2009.22.2679] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Accepted: 06/24/2009] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Lenalidomide and bortezomib are active in relapsed and relapsed/refractory multiple myeloma (MM). In preclinical studies, lenalidomide sensitized MM cells to bortezomib and dexamethasone. This phase I, dose-escalation study (ie, NCT00153933) evaluated safety and determined the maximum-tolerated dose (MTD) of lenalidomide plus bortezomib in patients with relapsed or with relapsed and refractory MM. PATIENTS AND METHODS Patients received lenalidomide 5, 10, or 15 mg/d on days 1 through 14 and received bortezomib 1.0 or 1.3 mg/m(2) on days 1, 4, 8, and 11 of 21-day cycles. Dexamethasone (20mg or 40 mg on days 1, 2, 4, 5, 8, 9, 11, and 12) was added for progressive disease after two cycles. Primary end points were safety and MTD determination. RESULTS Thirty-eight patients were enrolled across six dose cohorts. The MTD was lenalidomide 15 mg/d plus bortezomib 1.0 mg/m(2). Dose-limiting toxicities (n = 1 for each) were grade 3 hyponatremia and herpes zoster reactivation and grade 4 neutropenia. The most common treatment-related, grades 3 to 4 toxicities included reversible neutropenia, thrombocytopenia, anemia, and leukopenia. Among 36 response-evaluable patients, 61% (90% CI, 46% to 75%) achieved minimal response or better. Among 18 patients who had dexamethasone added, 83% (90% CI, 62% to 95%) achieved stable disease or better. Median overall survival was 37 months. CONCLUSION Lenalidomide plus bortezomib was well tolerated and showed promising activity with durable responses in patients with relapsed and relapsed/refractory MM, including patients previously treated with lenalidomide, bortezomib, and/or thalidomide. The combination of lenalidomide, bortezomib, and dexamethasone is being investigated in a phase II study in this setting and in newly diagnosed MM.
Collapse
|
65
|
Zangari M, Fink LM, Elice F, Zhan F, Adcock DM, Tricot GJ. Thrombotic events in patients with cancer receiving antiangiogenesis agents. J Clin Oncol 2009; 27:4865-73. [PMID: 19704059 DOI: 10.1200/jco.2009.22.3875] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tumor-associated neoangiogenesis has recently become a suitable target for antineoplastic drug development. In this overview, we discuss specific drug-associated hemostatic complications, the already known pathogenetic mechanisms involved, and the effect of varying antithrombotic strategies. Multiple agents with angiogenic inhibitory capacity (thalidomide, lenalidomide, bevacizumab, sunitinib, sorafenib, and sirolimus) have obtained US Food and Drug Administration approval, and many others have entered clinical trials. Arterial and venous thromboembolism and hemorrhage have emerged as significant toxicities associated with the use of angiogenesis inhibitors. We present a detailed analysis of the literature on thrombotic complication of antiangiogenic drugs. Close attention to hemostatic complications during antiangiogenic treatment is warranted. Further studies are required to better understand the pathophysiologic mechanisms involved and to define a safe prophylactic strategy.
Collapse
Affiliation(s)
- Maurizio Zangari
- University of Utah, Division of Hematology, Blood/Marrow Transplant and Myeloma Program, Salt Lake City, UT, USA.
| | | | | | | | | | | |
Collapse
|
66
|
Jagannath S, Durie BGM, Wolf JL, Camacho ES, Irwin D, Lutzky J, McKinley M, Potts P, Gabayan AE, Mazumder A, Crowley J, Vescio R. Extended follow-up of a phase 2 trial of bortezomib alone and in combination with dexamethasone for the frontline treatment of multiple myeloma. Br J Haematol 2009; 146:619-26. [PMID: 19622094 DOI: 10.1111/j.1365-2141.2009.07803.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
High-quality response to multiple myeloma (MM) therapy can be predictive for improved outcomes. Novel agents may improve the depth of responses and therefore prolong survival. We report on the extended follow-up of a phase II study in frontline MM of bortezomib alone and in combination with dexamethasone. Forty-nine previously untreated, symptomatic MM patients received bortezomib 1.3 mg/m(2), days 1, 4, 8, 11, for up to six 3-week cycles. High-dose dexamethasone was added for patients not reaching either a partial response after cycle 2 or a complete response (CR) after cycle 4. The overall response rate in 48 evaluable patients was 90%, with 42% achieving at least a very good partial response, of which 19% were CR/near CR. Thirty-six patients received high-dose dexamethasone with 28 (77%) showing improved response. Twenty-seven patients have undergone successful stem-cell transplantation (SCT). After median follow-up of 49 months, 15 patients have died; median overall survival has still not been reached, with an estimated survival at 4 years of 67%. Overall survival with and without SCT was not different (P = 0.54). Grade 3/4 adverse events included neutropenia (10%), sensory neuropathy (6% grade 3), neuropathic pain (4% grade 3), and diarrhoea (4% grade 3). Bortezomib +/- dexamethasone is an effective and well-tolerated induction regimen for the frontline treatment of MM.
Collapse
Affiliation(s)
- Sundar Jagannath
- St. Vincent's Comprehensive Cancer Center, New York, NY 10011, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Abstract
BACKGROUND Current treatments for autoantibody-mediated diseases (i.e., systemic lupus erythematosus) and alloantibodies (in transplant) are minimally effective. Although they deplete naïve B cells, plasmablasts, and transiently reduce antibody concentrations, they are minimally effective against long-lived, antibody-producing plasma cells. In transplantation, plasma cells produce antibodies directed against human leukocyte antigen (HLA) antigens causing poor allograft survival. We report the first clinical experience with a plasma cell depleting therapy, bortezomib, to abrogate anti-HLA antibodies in transplantation (outside of rejection) in an attempt to improve long-term allograft survival. METHODS Eleven patients with anti-HLA alloantibodies were treated with bortezomib. All patients underwent plasmapheresis to aid in removal of antibodies and to determine the effect of bortezomib. Serial measurements of anti-HLA antibody levels were conducted weekly by single antigen bead on Luminex platform. RESULTS Bortezomib treatment elicited substantial reduction in both donor-specific antibody (DSA) and non-DSA levels. Antibodies were directed against DSA in 8 of 11 cases. Mean time to antibody appearance was 2 months posttransplant. Within 22 days (median) from treatment initiation, 9 of 11 patients' antibody levels dropped to less than 1000 mean fluorescence intensity. Of two patients without successful depletion, all had peak mean fluorescence intensity more than 10,000. At a mean follow-up of approximately 4 months posttreatment, all patients have stable graft function. Minimal transient side effects were noticed with bortezomib in the form of gastrointestinal toxicity, thrombocytopenia, and paresthesias. CONCLUSIONS Bortezomib therapy effectively abrogates anti-HLA antibodies. Hence, removal of antibodies, by proteasome inhibition, represents a new treatment strategy for transplantation and may have benefit in autoimmune-related disease.
Collapse
|
68
|
Shimizu K, Itoh J. A possible link between Trousseau's syndrome and tissue factor producing plasma cells. Am J Hematol 2009; 84:382-5. [PMID: 19425064 DOI: 10.1002/ajh.21419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
69
|
|
70
|
|
71
|
Lonial S, Cavenagh J. Emerging combination treatment strategies containing novel agents in newly diagnosed multiple myeloma. Br J Haematol 2009; 145:681-708. [PMID: 19344388 DOI: 10.1111/j.1365-2141.2009.07649.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Treatment strategies for multiple myeloma have changed substantially over the past 10 years following the introduction of bortezomib and the immunomodulatory drugs thalidomide and lenalidomide. In the front-line setting, combination regimens incorporating these novel agents are demonstrating substantial activity, which is translating into improved outcomes compared with previous standards of care. Response rates and depth of response that were previously only seen with high-dose therapy plus stem-cell transplantation (HDT-SCT) can now be achieved with new induction regimens utilizing these novel agents. This has raised the need for trials that will determine the clinical benefit of early SCT in patients that have already achieved a high quality of response. Here, we review the improvements in response and outcome that are seen with these novel-agent regimens, both as induction therapy prior to HDT-SCT and in non-transplant patients, and highlight the latest data from key studies of various novel combinations, including regimens featuring bortezomib plus thalidomide or lenalidomide. We also review data on response and outcomes in patients with poor prognostic characteristics that indicate that the adverse impact typically seen with these factors may be overcome using novel-agent therapy.
Collapse
Affiliation(s)
- Sagar Lonial
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | | |
Collapse
|
72
|
Incidence and prophylaxis of venous thromboembolic events in multiple myeloma patients receiving immunomodulatory therapy. Thromb Res 2009; 123:679-86. [PMID: 18992924 DOI: 10.1016/j.thromres.2008.09.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Revised: 09/21/2008] [Accepted: 09/28/2008] [Indexed: 12/21/2022]
|
73
|
Davies AM, Chansky K, Lara PN, Gumerlock PH, Crowley J, Albain KS, Vogel SJ, Gandara DR. Bortezomib plus gemcitabine/carboplatin as first-line treatment of advanced non-small cell lung cancer: a phase II Southwest Oncology Group Study (S0339). J Thorac Oncol 2009; 4:87-92. [PMID: 19096312 PMCID: PMC3024911 DOI: 10.1097/jto.0b013e3181915052] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Bortezomib is a small-molecule proteasome inhibitor with single-agent activity in patients with non-small cell lung carcinoma (NSCLC) and synergy with gemcitabine in preclinical studies. This phase II study of bortezomib in combination with gemcitabine/carboplatin was conducted in chemotherapy-naive advanced NSCLC patients to assess efficacy and safety. METHODS Patients with selected stage IIIB/IV NSCLC, performance status 0-1, and no history of brain metastasis received up to six 21-day cycles of gemcitabine 1000 mg/m, days 1 and 8, carboplatin area under curve 5.0, day 1, and bortezomib 1.0 mg/m, days 1, 4, 8, and 11. RESULTS One-hundred-fourteen patients (52% adenocarcinoma, 85% stage IV) received a median of 3.6 treatment cycles. Median follow-up was >3 years. Median overall survival was 11 months; 1-year and 2-year survival rates were 47% and 19%, respectively. Median progression-free survival was 5 months; 1-year progression-free survival rate was 7%. Response rate was 23%, and disease control rate (responses + stable disease) was 68%. The most common grade 3/4 toxicities were thrombocytopenia (63%) and neutropenia (52%). One patient experienced febrile neutropenia. Grade 3/4 neuropathy occurred in 4%, and a further 6% experienced grade 2 sensory neuropathy. CONCLUSIONS Bortezomib plus gemcitabine/carboplatin resulted in a notable survival benefit in patients with advanced NSCLC, with the anticipated primary toxicity of myelosuppression. Further studies designed to investigate the role of bortezomib in advanced NSCLC are warranted.
Collapse
Affiliation(s)
- Angela M Davies
- Department of Hematology/Oncology, University of California, Davis Cancer Center, Sacramento, California, USA.
| | | | | | | | | | | | | | | |
Collapse
|