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.
2019-03-01T15:14:37.000Z

Results of the phase III VCAT study: subcutaneous bortezomib, thalidomide and prednisolone consolidation

Mar 1, 2019
Share:

Bookmark this article

Consolidation therapy with thalidomide and prednisolone (TP) in patients with newly diagnosed multiple myeloma (NDMM), post-autologous stem cell transplant (ASCT) is the standard of care in Australia.1 In other studies, consolidation with bortezomib (Velcade®), a proteasome inhibitor (PI), has been shown to deepen responses and prolong progression free survival (PFS) in patients with NDMM following ASCT. Therefore, it was hypothesized that adding bortezomib to the TP consolidation regimen, may increase patient outcomes.

In this study by Noemi Horvath, Royal Adelaide Hospital, AUS, and colleagues, patients with NDMM were treated with an induction regimen of bortezomib, cyclophosphamide and dexamethasone, proceeded to ASCT, and then received consolidation therapy of bortezomib + TP (VTP), or TP alone. The results of their randomized, open-label, phase III study, VCAT (NCT01539083), were recently published in Leukemia and Lymphoma.2

Patient characteristics and study design

  • The study recruited adult patients (N = 256) with NDMM who were eligible for transplant
  • Participating centers were in Australia, Korea and China
  • Primary endpoint: proportion of post-ASCT evaluable patients achieving complete response (CR) + very good partial response (VGPR) (≥VGPR) after 12 months of consolidation
  • Secondary endpoints:
  • CR rate and stringent CR rate (sCR) after 3, 6, 9 and 12 months of consolidation, PFS from randomization, disease free survival (DFS) in patients with CR, overall survival (OS), overall response rate (ORR) to induction and ASCT, safety and tolerability, health related quality of life (HRQoL), patient-related symptoms of peripheral neuropathy (PN)
  • Median age in intent-to-treat (ITT) population (N = 256): 59 years (32–71)

Therapy pathway:

  • Three, 21-day, cycles of induction with bortezomib, cyclophosphamide and dexamethasone (VCD)
    • Subcutaneous bortezomib: 1.3 mg/m2 on days 1, 4, 8 and 11
    • Oral cyclophosphamide: 300 mg/m2 on days 1, 8 and 15
    • Oral dexamethasone: 20 mg on days 1, 2, 4, 5, 8, 9, 11 and 12
  • Peripheral blood stem cell (PBSC) mobilization and collection
  • High-dose therapy (HDT) of 200 mg/m2 of intravenous melphalan and ASCT
  •  Randomized 1:1 followed by consolidation with thalidomide and prednisolone (TP) or bortezomib + TP (VTP) (N = 203)
    • TP (N = 100): thalidomide 100mg/d for ≤12 months or until progressive disease (PD) and prednisolone: 50 mg on alternate days, indefinitely or until PD
    • VTP (N = 103): subcutaneous bortezomib: 1.3 mg/m2 every 2 weeks for 32 weeks plus TP

Key Findings

All results are given as VTP versus TP

Efficacy

  • Induction therapy (N = 254):
    • Evaluable for response: N = 243
    • ≥VGPR achieved in 18.1% of patients
    • ORR: 71.6%
  • Post-ASCT
    • ≥VGPR: 35.8%
    • ORR: 79%
    • CR: 2.5%
  • PD developed in 8 patients during induction/post-transplant
  • At randomization:
    • ≥VGPR: 42.9% vs 37.5%
    • CR: 2% vs 4.2%
  • After randomization:
    • ≥VGPR at 3 months: 69% vs 65%
    • ≥VGPR at 12 months: 85.7% vs 77.1% (primary endpoint)
      • Rate difference: 8.6% (95% CI, 2.3–19.5%, P = 0.122)
    • Twelve months following consolidation, minimal residual disease (MRD) negativity was achieved in 20.4% vs 20.8%
  • Median duration of response (DoR) in responders to consolidation was not reached in either treatment arm

Survival:

  • Median follow-up post randomization: 22.3 months vs 23.2 months
  • PFS
    • Median: 31.7 vs 32.8 months
    • Hazard ratio [HR] 1.12, 95% CI, 0.67–1.87, P = 0.6689)
    • Twelve month: 89.7% vs 86.4%
    • Twenty-four month: 65.5% vs 74.2%
  • During consolidation, 11 patients developed PD
  • OS: at data cut-off, OS data was not mature
  • DFS
    • Median (in responders): 11.2 (N = 39) vs 16.4 (N = 43) months
    • Twelve month: 44.0% vs 61.9%
    • Thirty month: 28.0% vs 33.6%

Safety

During VCD induction (N = 254):

  • Median number of cycles: 3
  • Drug-related TEAE: 85% (N = 216)
  • Grade ≥3 TEAE: 42% (N = 107)
  • Most common grade ≥3 TEAE: neutropenia (6%, N = 15)
  • PN not elsewhere classified (NEC), grade 3: 3.5% (N = 9)

During consolidation:

  • Median duration of treatment:
    • Bortezomib (VTP cohort): 6.9 months (0–8.4)
    • Thalidomide: 10.2 months (0–11.1) vs 10.1 months (0–13.8)
    • Prednisolone: 10.2 months (0–13.2) vs 10.2 months (0–13.8)
  • TEAE:
    • Drug-related TEAE: 87% (N = 90) vs 91% (N = 90)
    • Most common all-cause TEAEs: PN (62%) and upper respiratory tract infection (38%) vs PN (63%) and constipation (38%)
    • Grade ≥3 TEAE: 28% vs 36%
    • Most frequent grade ≥3 TEAE: pneumonia (5%) and neutropenia (4%) vs PN (8%) and pneumonia (6%)
  • PN:
    • Frequency of PN: 62% (N = 64) vs 63% (N = 62)
    • Grade ≥3 PN: 2% vs 8%
  • The rate of hematologic TEAEs was low in both groups
  • Serious adverse events (SAEs): 27% (N = 28) vs 22% (N = 22)
    • Drug-related SAEs: 15% (N = 15) vs 8% (N = 8)
  • Discontinuations: 85 (53 prior to randomization and 32 post-randomization)
    • Post-randomization discontinuations predominantly due to TEAEs and PD
  • No deaths due to drug-related TEAEs during consolidation
  • HRQoL: largely similar between patient groups

Patients in this study were treated with the same induction therapy; therefore, upon starting consolidation all patients were IMid-naïve but bortezomib-exposed. It is possible that the improvements in efficacy were due to the exposure to thalidomide (an IMiD), explaining the similarity in the increases of response. Investigators had hypothesized an improvement of 15%, which was not reached.

It is possible that the number of cycles of induction therapy or the route of administration of bortezomib may have influenced the ≥VGPR rate. Additionally, whilst patients were recruited from Australia, China and Korea, 79% were white, meaning the patient population was not representative of all patients with NDMM.

With regards to safety, the addition of bortezomib was well tolerated and no new safety concerns emerged; with both VTP and TP regimens well-tolerated. However, a higher proportion of patients discontinued treatment due to TEAEs in the VTP group. The rate of hematologic AEs was low.

During consolidation, the percentage of patients achieving a VGPR or better was improved in both treatment arms and despite an improvement trend in the ≥VGPR rate after 12 months in the VTP arm, statistical significance was not reached. No statistically significant differences were observed in the secondary endpoints (PFS, DFS and HRQoL). The VTP regimen investigated here is not recommended compared to TP alone for patients with NDMM who receive VCD induction and ASCT.

  1. Myeloma Australia: clinical practice guideline multiple myeloma. http://myeloma.org.au/wp-content/uploads/2017/10/MSAG-Clinical-Practice-Guideline-Myeloma-V4-March-2017.pdf [accessed 2019 Feb 25]
  2. Horvath N. et al. Phase 3 study of subcutaneous bortezomib, thalidomide, and prednisolone consolidation after subcutaneous bortezomib-based induction and autologous stem cell  transplantation in patients with previously untreated multiple myeloma: the VCAT study. Leuk and Lymph. 2019 Feb 19. DOI: 10.1080/10428194.2019.1579322

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 - 13 days left ...

Newsletter

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