All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit the International Myeloma Foundation or HealthTree for Multiple Myeloma.

The Multiple Myeloma Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your Multiple Myeloma Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The Multiple Myeloma 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 Multiple Myeloma Hub cannot guarantee the accuracy of translated content. The Multiple Myeloma Hub 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.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2017-03-30T14:12:05.000Z

Bortezomib consolidation therapy confers distinct PFS benefit in newly diagnosed MM patients

Mar 30, 2017
Share:

Bookmark this article

In a letter to the editor of Leukemia, published in March 2017, Hermann Einsele and colleagues from the Julius-Maximilians-University of Würzburg present the case for the use of bortezomib as consolidation therapy for newly diagnosed multiple myeloma (MM) patients, following autologous stem cell transplantation (ASCT). Data were analyzed from two bortezomib phase 3 trials that assessed NDMM patients after ASCT, and stratified according to age and prior treatment regimens. Studies were carried out in 47 departments across Germany, between October 2006 and May 2013. The primary endpoint was progression free survival (PFS) from the start of induction therapy. Secondary endpoints assessed were event-free survival (EFS), from start of induction to start of new chemotherapy or death, response rates, overall survival (OS), quality of life, and safety.

Key Findings

  • pts (n=371) randomly assigned to receive:
    • consolidation therapy (n= 186): four 35-day cycles of intravenous bortezomib 1.6 mg/m2 (days 1, 8, 15, and 22) or
    • observation arm: no therapy (n= 185)
    • 14 withdrew consent and are excluded from efficacy analysis
  • Cytogenetic changes were assessed by FISH
  • Unfavorable genetics (del13q, t(4:14) or del17p)= 39%; High-risk cytogenetics (t[4;14] or del17p) = 16%
  • All data points are given as: bortezomib arm vs. observation arm
  • Median PFS = 33.6 vs. 27.8 months                                                                                                    HR = 0.75; 95% CI, 0.59–0.97; P = 0.0243
  • Median PFS (sensitivity analysis) = 23.1 vs. 17.2 months                                                           adjusted HR = 0.70; (95% CI, 0.54–0.89); P = 0.0046
  • Shorter PFS for pts with unfavorable cytogenetics vs. no cytogenetic changes: adjusted HR = 1.46; (95% CI, 1.03–2.08); P = 0.034)
  • PR at randomization versus ≥ VGPR: adjusted HR = 1.35; (95% CI, 1.03–1.77); P = 0.032
  • Pts with < VGPR at randomization benefited significantly from bortezomib consolidation: HR = 0.58 (95% CI, 0.53-1.05); median PFS = 33.3 vs. 24.5 months
  • PFS improvements were observed regardless of cytogenetic status:
    • no changes: HR = 0.77; 95% CI, 0.47–1.27; P = 0.297
    • del 13q, t(4;14), del 17p: HR = 0.66; 95% CI, 0.41–1.05; P = 0.074
    • t(4;14), del 17p: HR = 0.61; 95% CI, 0.30– 1.22; P = 0.154
  • EFS (from induction to start of second-line treatment) = 37.8 vs. 34.3 months;                          adjusted HR = 0.74; 95% CI, 0.57–0.96; P = 0.024
  • pts with ≥ VGPR at the end of treatment/observation = 62% vs. 48%; P = 0.003
  • Discontinuation due to AEs = 15% (28 pts) of bortezomib-treated pts: nausea (3%), diarrhea (3%), vomiting (3%), polyneuropathy (2%), and disease progression (2%)
  • Serious AE s = 11% vs. 17% and included: herpes zoster: 3% vs. 4%, disease progression 1% vs. 4% and pneumonia 1% vs. 2%
  • Polyneuropathy, peripheral neuropathy, and peripheral sensory neuropathy = 18% vs. 6%
  • Deaths during post-treatment follow-up (30–60 months after the end of treatment visit) = 25% (47 pts) vs. 30% (56 pts)
  • No treatment-related deaths

Consolidation therapy with bortezomib (administered over a period of 5 months, 4 cycles), was well-tolerated and highly effective in delaying disease progression (6-month increase in PFS), as well as improving the quality of response in NDMM patients. Of note, improvements were seen in high-risk subsets: < VGPR after HDT/ASCT, del13q, t[4;14] or del17p, and, t(4;14), del 17p for which prognosis is generally low. In addition, a favorable response was independent of prior bortezomib treatment in the induction therapy.

  1. Einsele H. et al. Response-adapted consolidation with bortezomib after ASCT improves progression-free survival in newly diagnosed multiple myeloma.Leukemia. 2017 Apr 18. DOI: 10.1038/leu.2017.83. [Epub ahead of print]

Your opinion matters

As a result of this content, I commit to reviewing the CARTITUDE clinical program to guide my understanding of cilta-cel in clinical practice.
17 votes - 12 days left ...

Newsletter

Subscribe to get the best content related to multiple myeloma delivered to your inbox