The mm Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the mm Hub cannot guarantee the accuracy of translated content. The mm and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.
The Multiple Myeloma Hub is an independent medical education platform, sponsored by Bristol Myers Squibb, GSK, Johnson & Johnson, Pfizer, Roche and Sanofi. The levels of sponsorship listed are reflective of the amount of funding given. View funders.
Now you can support HCPs in making informed decisions for their patients
Your contribution helps us continuously deliver expertly curated content to HCPs worldwide. You will also have the opportunity to make a content suggestion for consideration and receive updates on the impact contributions are making to our content.
Find out moreCreate an account and access these new features:
Bookmark content to read later
Select your specific areas of interest
View mm content recommended for you
The survival of patients with multiple myeloma (MM) has been improving over the last two decades, and many patients can now reach an overall survival (OS) of 10 years. MM is still incurable, despite the introduction of immunotherapy, and treatment options are limited for patients who experience disease progression after treatment with first-generation novel agents such as bortezomib, lenalidomide, and thalidomide.
Carfilzomib, a second-generation irreversible proteasome inhibitor, has demonstrated efficacy in patients who were refractory to bortezomib with a good safety profile. Carfilzomib was first approved in combination with lenalidomide plus dexamethasone in the relapsed and/or refractory MM (RRMM) setting and at a different dosing regimen with dexamethasone alone.
Bendamustine, a bifunctional chemotherapeutic agent with alkylating and antimetabolite properties, can improve complete response (CR) rate, time to treatment failure when combined with prednisone, and quality of life. It is approved by the European Medicines Agency for patients with MM and severe renal impairment. Although bendamustine has safety issues with peripheral neuropathy, it showed promising efficacy in patients with advanced MM when combined with bortezomib.
There is an unmet need for new therapeutic options that are immunomodulatory drug-free for patients who have also been exposed to bortezomib. Thus, Francesca Gay et al. published a multicenter dose-escalation phase I/II study in Cancer (NCT02056756), which was conducted within the European Myeloma Network as EMN09, to evaluate the maximum tolerated dose (MTD), the safety, and the efficacy of carfilzomib, bendamustine, and dexamethasone (KBd) in patients with RRMM.1
Patients were eligible if they had
Patients with Grade >2 peripheral neuropathy or a left ventricular ejection fraction <40% were excluded.
The primary objective of the phase I part of the study was to define the MTD of KBd, while the primary objective of the phase II part was to establish the rate of very good partial responses (VGPRs). The secondary endpoints were to determine the overall response rates, progression-free survival (PFS), OS, and conduct subgroup analyses of prognostic factors.
All patients were given 20 mg dexamethasone orally on Days 1, 2, 8, 9, 15, 16, 22, and 23. After prehydratation, carfilzomib was administered intravenously for >30 minutes on Days 1, 2, 8, 9, 15, and 16 in a 28-day cycle.
From April 2014 to February 2017, 13 patients were enrolled in the phase I dose-escalation part of the study, and 50 patients were enrolled in the phase II part. Patients in this study had received a median of three previous lines of therapy before KBd. Key patient characteristics are shown in Table 1.
Table 1. Patient characteristics*
ASCT, autologous stem cell transplantation; IQR, interquartile range. |
|
Characteristic, % (unless otherwise stated) |
Total (N = 63) |
---|---|
>65 Median age (range), years |
66 (37–79) |
>65 years |
57 |
Male |
59 |
International Staging System |
|
I |
52 |
II |
30 |
III |
17 |
High-risk cytogenetics† |
48 |
Performance status |
|
0 |
41 |
≥1 |
56 |
Disease status |
|
Primary refractory |
5 |
Relapsed |
65 |
Relapsed and refractory |
30 |
Median time from diagnosis to study entry (IQR), years |
5.2 (2.7–8.2) |
Number of previous lines of therapy |
|
2 |
38 |
3 |
16 |
4 |
16 |
≥5 |
30 |
Previous therapy |
|
ASCT |
75 |
Refractory‡ |
21 |
Bortezomib |
87 |
Refractory‡ |
33 |
Immunomodulators§ |
86 |
Lenalidomide |
76 |
Refractory‡ |
60 |
The KBd regimen at dose level 0 was effective but led to three of six patients experiencing Grade 4 lymphopenia. At dose level +1, a third of patients had Grade 4 thrombocytopenia and Grade 3 febrile neutropenia. After the enrolment of four additional patients at this dose level, the MTD was defined at 36 mg/m2 (dose level +1).
Since carfilzomib at 27 mg/m2 was better tolerated and achieved similar responses to higher doses, the phase II part of the study evaluated the triplet with carfilzomib at this dose level.
In an intention-to-treat analysis, 56 out of 63 patients (88.8%) showed a decrease in M protein. KBd achieved an overall response rate of 52%, with 32% of patients at least in VGPR. Table 2 shows the best overall responses of all patients in the study.
Table 2. Best overall response*
CR, complete response; nCR, near complete response; PR, partial response; SD, stable disease; VGPR, very good partial response. |
|
Response, % |
Total (N = 63) |
---|---|
CR |
5 |
nCR |
13 |
VGPR |
14 |
≥VGPR |
32 |
PR |
21 |
≥PR |
52 |
SD |
38 |
The median PFS was 11.6 months (95% confidence interval [CI], 7.9–15.3 months) and the median OS was 30.4 months (95% CI, 20.5 months to not reached). Patients with standard-risk chromosomal aberrations had significantly better PFS compared with those who were high-risk, with a median PFS of 19.6 vs 7.9 months; hazard ratio, 0.43; 95% CI, 0.21–0.88; p = 0.021.
Despite the small sample size, the subgroup analysis showed that patients with poor prognostic indicators such as deletion 17p (n = 14) were still able to reach a median PFS of 9.4 months. Patients with standard-risk chromosomal aberrations also had a significantly improved OS vs those who were high-risk: the 18-month OS rate was 87% vs 52% (hazard ratio, 0.24; 95% CI, 0.08-0.67; p = 0.007).
There were no clinically significant differences in PFS between patients who relapsed on or were refractory to lenalidomide vs those who relapsed on or were refractory to bortezomib.
During induction, the following was observed:
During maintenance therapy, the following was observed:
Carfilzomib, at the optimal dose of 27 mg/m2, combined with bendamustine and dexamethasone demonstrated clinical efficacy in patients with advanced disease. The safety profile was manageable, with no renal toxicity observed in this study even though patients with moderate renal dysfunction were included.
Overall, in this EMN09 study, the KBd regimen achieved significant rates of ≥VGPR. Patients were able to reach responses rapidly, and the median PFS was 11.6 months.
Treatment with KBd for this advanced stage patient population compares favorably with other regimens, including bortezomib-containing combinations, thus presenting a reasonable alternative. Additionally, as bendamustine has become generic in some countries, it may come at an accessible price, which will help many countries where multiple lines of therapy are needed but not affordable, therefore alleviating a significant financial burden.
References
Please indicate your level of agreement with the following statements:
The content was clear and easy to understand
The content addressed the learning objectives
The content was relevant to my practice
I will change my clinical practice as a result of this content