Cyclophosphamide Maintenance Therapy Is a Safe and Effective Alternative In Multiple Myeloma Patients Who Undergo Autologous Stem Cell Transplantation

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5373-5373
Author(s):  
Michael T. Byrne ◽  
Jan S Moreb

Background In spite of recent improvements in survival, multiple myeloma (MM) remains an incurable disease.  Following autologous stem cell transplantation (ASCT), maintenance therapy has been shown to improve progression free survival (PFS) but data on overall survival (OS) is mixed.  The expense of therapy is significant and a substantial number of patients are unable to tolerate its toxicity.  In this study, we report our experience with the use of oral cyclophosphamide ± prednisone (CY) as an alternative for these patients.  Methods From January of 2000 to December of 2010, we retrospectively evaluated all patients with MM who underwent ASCT at our institution and separated them into four groups depending on maintenance therapy: No maintenance, interferon ± prednisone (IFN/Pd), IMIDs, and CY.  Patients who received maintenance after their first relapse were analyzed separately for tolerability, PFS, and OS.  Survival data was analyzed by logrank test and the group characteristics were compared using an unpaired t-test.    Results A total of 288 patients underwent ASCT at our institution from 2000 to 2010.  Of these patients, 8 were excluded due to insufficient follow-up.  1 patient who received bortezomib was also excluded. The median age at diagnosis was 58.0 years and the median time from diagnosis to ASCT was 8.3 months.  Collectively, this cohort included 193 Caucasians, 64 African Americans, 16 Hispanics, 4 Asian and 2 unknown race; 55.7% were men and 44.3% were women.  Following ASCT, 112 patients received no maintenance therapy, 74 of them (66.1%) had documented relapses with a median PFS of 18.2 months.  78 patients received IFN/Pd; 55 of them (70.5%) relapsed with a median PFS of 18.4 months. Alternatively, 79 patients were treated with IMIDs with 40 known relapses (50.6%) and a median PFS of 22.5 months.   CY maintenance was used in 10 patients; six patients relapsed within a median time of 20.4 months.  Collectively, maintenance therapy was associated with a significant improvement in PFS (22.5 vs. 18.2 months, p=0.037) but no difference in OS (45.5 vs. 43.9 months, p=0.29).  Among patients treated with maintenance therapy, PFS was superior in the group treated with IMIDs compared to IFN/Pd (22.5 vs. 18.4 months, p <0.001) however there was no improvement in OS (43.1 vs. 46.5 months, p=0.29). PFS was equivocal between CY and all other maintenance (20.4 vs. 22.5 months, p=0.19).  OS of first-remission CY vs. other maintenance therapy cannot be calculated due to small sample size.  34.6-38.8% of patients who received non-CY maintenance therapy had toxicity requiring dose reductions or discontinuation whereas 10% of CY patients developed toxicity (Table 1).  Following relapse and salvage therapy, 78 patients received non-CY second-remission maintenance therapy with a median PFS of 9.2 months whereas those treated with second-remission CY (n=25) had a median PFS of 11.7 months (p=0.89).  OS was comparable between the two groups (69.2 vs. 79.8 months, p=0.44).  Patients who received CY, however, had a significantly longer OS compared to those who did not receive second-remission maintenance therapy (79.8 vs. 41.2 months, p<0.001) and a trend toward improved OS compared to other forms of second remission maintenance (p=0.085).  5.0% of patients in this study developed secondary malignancies: 4 in non-maintenance group, 3 in the IFN/Pd group, 6 in the IMID group, and 1 in the CY group (Table 2).  Conclusions  CY is well tolerated with comparable efficacy to other maintenance drugs in MM, particularly in the second maintenance setting. Our data corroborates published experience that maintenance therapy improves PFS but often fails to impact OS.  Such improvement in PFS with conventional maintenance therapy is associated with a high rate of toxicity and an increase in secondary malignancies. Disclosures: No relevant conflicts of interest to declare.

2019 ◽  
Vol 19 (10) ◽  
pp. 889-898 ◽  
Author(s):  
Almuth Maria Anni Merz ◽  
Maximilian Merz ◽  
Jens Hillengass ◽  
Sarah A. Holstein ◽  
Philip McCarthy

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5232-5232
Author(s):  
Celso Mitsushi Massumoto ◽  
Edilson Pinheiro Junior ◽  
Otávio C.G. Baiocchi ◽  
Ronald Pinheiro ◽  
Adelson Alves

Abstract Introduction: autologous stem cell transplantation is a potentially curative or may augment the time to progression in Multiple Myeloma (MM) patients. The immunotherapy with rituximab may help control the minimal residual disease after high dose chemotherapy. Twenty percent (20%) of Multiple Myeloma patients express the CD20+ protein and can be target for immunotherapy. Objective: The aim of this study was to evaluate the use of rituximab after autologous stem cell transplantation for Multiple Myeloma. Patients and Methods: eight patients (4 male) with a median age of 53 (range 43–59) years diagnosed with MM. All of them had received at least one previous regimen were enrolled in the protocol study. All patients signed the consent form. Patients in relapse received a salvage regimen with C-VAD n=2 (cyclofosfamide 4 g/m2 e vincristine 0.4 mg/d (d 1–4), doxorrubicin 0.9 mg/m2 (d1-4) e dexametasone 40 mg (d1-4; 9-12; 17–22) or cyclofosfamide (1OO mg/kg, n=7) followed by stem cell harvesting. The preparative regimen was Busulfan 12 mg/kg and cyclofosfamide 120 mg/kg or Melphalan 200mg/m2. Rituximab at a dose of 375mg/m2 weekly x 4 was given every 6 months for 2 years after SCT. The clinical characteristics of the patients are shown on Table 1. Results: the median time to ANC and platelets engraftment was 11 (range 8–12) and 26 (range 17–35) days. Patients have been in CR at a median time of 11 months follow-up. Minor Rituximab-associated toxicities were seen:rigor, fever and short of breath that were controlled with acetaminophen and diphenidramine. Conclusion: the Rituximab given after autologous stem cell transplantation is safe in Multiple Myeloma patients and may prolong time to disease progression. A randomized study is required to evaluate the role of rituximab after ASCT. Table 1 - Clinical Characteristics of Patients Patients Age/gender Status Pre- BMT Status Post- BMT Salvage Tx Prep. regimen ANC/Platelets X1000 MM3/ml Follow-up (months) FRC 57/M PR PR C-VAD BU+MEL 12/28 EXPIRED MB 52/F CR1 CR1 C-VAD BU+MEL 12/60 EXPIRED AM 52/F PR PR C-VAD BU+MEL 9/26 EXPIRED IM 54/M PR CR Cyx2 BU+MEL 12/21 ALIVE GAD 50/F PR CR Cyx2 BU+MEL 12/35 ALIVE SCM 59/M CR CR Cyx2 BU+MEL 10/17 ALIVE MAD 63/F CR CR C-VAD MELPHALAN 12/25 ALIVE JFC 51/M CR CR C-VAD BU+MEL 12/18 ALIVE


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4548-4548
Author(s):  
Catherine Garnett ◽  
Chrissy Giles ◽  
Maialen Lasa ◽  
Osman Ahmed ◽  
Marco Bua ◽  
...  

Abstract Abstract 4548 High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) is currently standard treatment for younger patients with multiple myeloma (MM). In the face of almost inevitable disease relapse, there is growing evidence for second ASCT as salvage therapy in certain patient groups. However, few published data exist regarding efficacy and safety of third ASCT in relapsed disease. We retrospectively analysed the results of eight patients treated at a single UK institution who each received three separate autologous stem cell transplants for relapsed MM between May 1997 and April 2012. There were four men and four women. Median age at diagnosis was 48 years (range, 25–64 years). Paraprotein isotype was IgA in two patients and IgG in the remaining six patients. At the time of 1st transplant, seven patients were in partial response (PR) and one in complete response (CR). Conditioning melphalan dose was 200mg/m2 in all but two patients who received 140mg/m2. Three patients entered CR following 1st transplant and four patients showed PR. Median time to disease progression was 31 months (range, 11.8–52.9 months). Prior to 2nd transplant, five patients achieved very good partial response (VGPR) and three PR with induction chemotherapy. Melphalan dose was 200mg/m2 in five patients and 140mg/m2 in the remaining three. Median time to disease progression was 22.3 months (range, 10.1– 39.6 months). At the time of 3rd transplant, two patients had achieved VGPR following induction chemotherapy, one showed PR, two stable disease (SD) and three evidence of disease progression. For the 3rd transplant, melphalan dose was reduced in most cases. Median follow up post 3rd transplant was 8.3 months (range 1.1–29.3 months). One patient died of overwhelming sepsis within one month of transplantation (treatment related mortality). At the time of analysis, five patients had relapsed following 3rd ASCT, with median time to disease progression of 10.4 months (range, 2.7–23.7 months). Three of these patients died at 3.5, 17.6 and 27.1 months post 3rd transplant. The remaining two patients are alive with no evidence of disease relapse (progression free survival (PFS) time of 3.3 and 1.3 months). Overall survival (OS) for the group from diagnosis is 62% at 10 years with a median OS from diagnosis of 149 months (range 68.5 – 189.2 months) (Figure 1). Median OS for the group from 3rd transplant is 17.6 months (range, 1.1–29.3 months) (Figure 2). Median PFS is 10.4 months (range 1.1–23.7 months). These results demonstrate that third ASCT is a possible treatment strategy for patients with relapsed MM and may prolong patient survival. Figure 1: Overall survival from diagnosis for patients receiving 3rd autologous stem cell transplantation for relapsed multiple myeloma Figure 1:. Overall survival from diagnosis for patients receiving 3rd autologous stem cell transplantation for relapsed multiple myeloma Figure 2: Overall survival from time of 3rd autologous stem cell transplant Figure 2:. Overall survival from time of 3rd autologous stem cell transplant Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 23 (2) ◽  
pp. 262-268 ◽  
Author(s):  
Dharshan Sivaraj ◽  
Michael M. Green ◽  
Zhiguo Li ◽  
Anthony D. Sung ◽  
Stefanie Sarantopoulos ◽  
...  

Blood ◽  
2012 ◽  
Vol 119 (13) ◽  
pp. 3003-3015 ◽  
Author(s):  
Heinz Ludwig ◽  
Brian G. M. Durie ◽  
Philip McCarthy ◽  
Antonio Palumbo ◽  
Jésus San Miguel ◽  
...  

Abstract Maintaining results of successful induction therapy is an important goal in multiple myeloma. Here, members of the International Myeloma Working Group review the relevant data. Thalidomide maintenance therapy after autologous stem cell transplantation improved the quality of response and increased progression-free survival (PFS) significantly in all 6 studies and overall survival (OS) in 3 of them. In elderly patients, 2 trials showed a significant prolongation of PFS, but no improvement in OS. A meta-analysis revealed a significant risk reduction for PFS/event-free survival and death. The role of thalidomide maintenance after melphalan, prednisone, and thalidomide is not well established. Two trials with lenalidomide maintenance treatment after autologous stem cell transplantation and one study after conventional melphalan, prednisone, and lenalidomide induction therapy showed a significant risk reduction for PFS and an increase in OS in one of the transplant trials. Maintenance therapy with single-agent bortezomib or in combination with thalidomide or prednisone has been studied. One trial revealed a significantly increased OS with a bortezomib-based induction and bortezomib maintenance therapy compared with conventional induction and thalidomide maintenance treatment. Maintenance treatment can be associated with significant side effects, and none of the drugs evaluated is approved for maintenance therapy. Treatment decisions for individual patients must balance potential benefits and risks carefully, as a widely agreed-on standard is not established.


Sign in / Sign up

Export Citation Format

Share Document