This information is intended for US healthcare professionals to access current scientific information about J&J Innovative Medicine products. It is prepared by Medical Information and is not intended for promotional purposes, nor to provide medical advice.

Use of DARZALEX + DARZALEX FASPRO in Combination with Bortezomib, Lenalidomide, and Dexamethasone in Multiple Myeloma

Last Updated: 02/12/2025

Summary

  • DARZALEX for intravenous (IV) use is not approved by the regulatory agencies for use in combination with bortezomib, lenalidomide, and dexamethasone (VRd) for the treatment of multiple myeloma (MM). Janssen does not recommend the use of DARZALEX in a manner that is inconsistent with the approved labeling.
  • GRIFFIN is a 2-part, phase 2 study evaluating the safety and efficacy of DARZALEX (D) when administered in combination with VRd in patients with newly diagnosed MM (NDMM) eligible for high-dose therapy (HDT) and autologous stem cell transplant (ASCT).1-3
    • Part 1: Voorhees et al (2021)4 reported the final analysis of the safety run-in cohort (N=16) of the GRIFFIN study with a median follow-up of 40.8 months. By the end of DARZALEX in combination with bortezomib, lenalidomide, and dexamethasone (D-VRd) consolidation, 56.3% patients achieved stringent complete response (sCR), and 50% were minimal residual disease (MRD) negative (10-5 threshold). After maintenance, 93.8% patients achieved sCR, and 81.3% patients were MRD negative (10-5 threshold). Grade 3/4 treatment-emergent adverse events (TEAEs) were reported in 93.8% of patients. One death from progressive disease (PD) occurred in the patient who did not achieve sCR.
    • Part 2: Voorhees et al (2020)3 presented the primary analysis of the randomized portion of this study (n=207). Voorhees et al (2023)5 reported the final efficacy and safety results after all patients completed ≥1 year of follow-up after the end of study treatment, died, or withdrew from study participation. The median duration of follow-up was 49.6 months. In the D-VRd vs VRd arm, respectively, sCR was achieved in 67% vs 48% of patients (odds ratio [OR], 2.18; 95% confidence interval [CI], 1.22-3.89; 2-sided P=0.0079) and complete response or better (≥CR) in 83% vs 60% of patients (P=0.0005). At the end of the study, 14% (n/N=15/104) and 10% (n/N=10/103) of patients in the D-VRd and VRd arms, respectively, who were previously MRD-positive at the end of the consolidation phase, converted to MRD-negative (10-5). In the D-VRd vs VRd arm, the most common (≥10%) grade 3/4 TEAEs were neutropenia (46% vs 23%), lymphopenia (23% in both arms), leukopenia (17% vs 8%), thrombocytopenia (16% vs 9%), pneumonia (12% vs 14%), and hypophosphatemia (10% vs 11%).
    • Sborov et al (2022)6 conducted a post-hoc analysis to evaluate the risk and incidence of vascular thrombotic events (VTE) in patients receiving D-VRd vs VRd in the GRIFFIN study. At a median follow-up of 38.6 months, VTEs were reported in 10.1% (n=10) of patients in the D-VRd arm and 15.7% (n=16) of patients in the VRd arm. The most common any-grade VTEs were deep vein thrombosis (D-VRd, 2%; VRd, 6.9%), pulmonary embolism (D-VRd, 2%; VRd, 3.9%), and embolism classified as unspecified vessel type and mixed arterial and venous (D-VRd, 2%; VRd, 2.9%). Among patients experiencing VTEs, clinical response deepened over time and response rates were higher in the D-VRd vs VRd arm after 2 years of maintenance.
    • Chhabra et al (2023)7 conducted a post hoc analysis to evaluate stem cell mobilization, collection yields, and hematopoietic recovery following ASCT after frontline DARZALEX- and lenalidomide-containing quadruplet induction in patients with NDMM in the GRIFFIN and MASTER studies. In the GRIFFIN study, 175 patients (D-VRd, n=95; VRd, n=80) underwent stem cell mobilization, with a median duration of 27 days after completing D-VRd induction therapy and 24 days after completing VRd induction therapy. Among patients who underwent mobilization and stem cell collection in the D-VRd vs VRd arm, 2% vs 6% of recipients did not achieve the center-specific, minimally required cluster of differentiation (CD)34+ cell yield in the initial mobilization attempt, respectively. Among patients who underwent mobilization, plerixafor use was more prevalent in those receiving D-VRd (72%) compared with those receiving VRd (55%). The median total CD34+ cell collection after D-VRd vs VRd induction was 8.3×106/kg (range, 2.6-33.0×106/kg) vs 9.4×106/kg (range, 4.1-28.7×106/kg), respectively. The median collection duration in the D-VRd vs VRd arm was 2 days vs 1 day, respectively. Among individuals who underwent mobilization, 99% vs 98% of patients in the D-VRd vs VRd arm underwent ASCT using median CD34+ cell doses of 4.2×106/kg vs 4.8×106/kg, respectively.
  • PERSEUS (MMY3014) is an ongoing, open-label, multicenter, randomized, phase 3 study evaluating the efficacy and safety of DARZALEX FASPRO in combination with bortezomib, lenalidomide, and dexamethasone (D-VRd) vs VRd induction and consolidation followed by maintenance with lenalidomide and DARZALEX FASPRO (D-R) in D-VRd group or lenalidomide (R) in VRd group in patients with NDMM eligible for ASCT.8
    • Sonneveld et al (2023)8 reported efficacy and safety results from the PERSEUS study evaluating D-VRd vs VRd in patients with NDMM eligible for ASCT. At a median follow-up duration of 47.5 months (range, 0-54.4), 14.1% (50/355) of patients in the D-VRd arm vs 29.1% (103/354) of patients in the VRd arm experienced disease progression or death (hazard ratio [HR], 0.42; 95% CI, 0.30-0.59; P<0.0001), respectively. Overall ≥CR, 87.9% vs 70.1% (P<0.0001) and overall MRD-negativity 75.2% vs 47.5% (P<0.0001) were higher with D-VRd vs VRd. The most common grade 3/4 adverse events (AEs) in the D-VRd vs VRd arm were neutropenia (62.1% vs 51%), thrombocytopenia (29.1% vs 17.3%), diarrhea (10.5% vs 7.8%), pneumonia (10.5% vs 6.1%), and febrile neutropenia (9.4% vs 10.1%).
    • Sonneveld et al (2024)9 presented (at the 21st International Myeloma Society [IMS] Annual Meeting) the stem cell yield and ASCT results for transplant-eligible patients with NDMM from the PERSEUS study who received D-VRd vs VRd induction prior to HDT or ASCT. Of the 640 patients who underwent stem cell collection (D-VRd, n=326 vs VRd, n=314), a high percentage of patients in both groups (D-VRd, 98.2% vs VRd, 99.4%) had sufficient stem cells collected for ASCT (≥2×106/kg cells). Although the D-VRd vs VRd group had a numerically lower median number of collected CD34+ stem cells (5.52×106/kg vs 7.44×106/kg), the percentage of patients who underwent ASCT was similar in both groups (89.7% vs 87%). After completion of 6 treatment cycles, 58 patients (29 in each treatment group) received ASCT, with 100% hematopoietic reconstitution and comparable engraftment times in both groups.
    • Bertamini et al (2024)10 presented (at the 66th American Society of Hematology [ASH] Annual Meeting) results from the PERSEUS study that highlighted the significance of circulating tumor cells (CTCs) as a biomarker in transplant-eligible patients with NDMM. D-VRd vs VRd significantly improved patient outcomes, across both high and low CTC levels (P<0.0001). D-VRd vs VRd improved MRD-negativity rates (≥CR; 10–5 and 10–6 sensitivities) and sustained MRD-negativity rates (≥CR; 10-5 and 10–6 sensitivities; ≥12 months) in patients with both high and low CTC levels. D-VRd vs VRd improved progression-free survival (PFS) in patients with high cytogenetic risk and low CTC levels. However, there was no improvement in outcomes for patients with high cytogenetic risk combined with high CTC levels. Further, D-VRd vs VRd significantly improved PFS in patients with both high and low CTC levels (P<0.0001).
  • CEPHEUS (MMY3019) is a phase 3 study evaluating the efficacy and safety of DARZALEX FASPRO in combination with bortezomib, lenalidomide, and dexamethasone (D-VRd) or VRd in patients with NDMM who are transplant ineligible (TIE) or for whom transplant is not planned as initial therapy (transplant deferred). The trial has enrolled 395 patients in 13 countries.11-13
    • Usmani et al (2025)13 reported results from the phase 3 CEPHEUS study. At a median follow-up of 58.7 months (range, 0.1-64.7), the overall MRD-negativity (10-5 sensitivity with ≥CR) rate was significantly higher with D-VRd vs VRd (60.9% vs 39.4%; OR, 2.37; 95% CI, 1.58-3.55; P<0.0001). Similarly, the sustained MRD-negativity (10-5 sensitivity; ≥12 months) rate was significantly higher in the D-VRd vs VRd arm (48.7% vs 26.3%; OR, 2.63; 95% CI, 1.73-4; P<0.0001). The overall ≥CR rate was significantly higher with D-VRd vs VRd (81.2% vs 61.6%; OR, 2.73; 95% CI, 1.71-4.34; P<0.0001). Further, D-VRd significantly improved the PFS (HR, 0.57; 95% CI, 0.41-0.79; P=0.0005) with 43% reduction in the risk of progression or death vs VRd. The overall survival (OS) rate favored the D-VRd vs VRd arm (HR, 0.85; 95% CI, 0.58-1.24). Serious TEAEs occurred in 142 (72.1%) vs 131 (67.2%) of patients from the D-VRd vs VRd arm, respectively, with the most common serious TEAE being pneumonia (D-VRd, 13.7%; VRd, 12.8%). The overall safety data were consistent with the established safety profile of each individual drug.
    • Zweegman et al (2024)14 presented (at the 66th ASH Annual Meeting) an expanded analysis of MRD outcomes from the CEPHEUS study in patients with NDMM who were ineligible for a stem cell transplant (SCT) or transplant deferred. The addition of DARZALEX FASPRO to VRd improved cumulative MRD-negativity rates (both 10–5 and 10–6 sensitivities) vs VRd alone at all prespecified timepoints. At 54 months, among patients who achieved both MRD-negativity (10–6 sensitivity) and ≥CR with D-VRd, >85% were alive and progression free. Analyses of MRD-negativity (with ≥CR) rates in D-VRd vs VRd were generally consistent across prespecified subgroups.
  • PLEIADES is an ongoing, phase 2, non-randomized, open-label, multicenter study evaluating the clinical benefit of DARZALEX FASPRO administered in combination with 4 standard-of-care treatment regimens in patients with MM. Specifically with transplant-eligible NDMM DARZALEX FASPRO + VRd (D-VRd) for patients with transplant-eligible NDMM (n=67).15-18 Results specifically to the D-VRd cohort are summarized.
  • Several subgroup analyses regarding the use of DARZALEX and DARZALEX FASPRO in combination with VRd in patients with MM were identified. These analyses are listed in the References section for your information.19-25

PRODUCT LABELING

CLINICAL STUDIES

DARZALEX Phase 2 Study

GRIFFIN (MMY2004; clinicaltrials.gov identifier: NCT02874742) is an ongoing, 2-part, open-label, multicenter, phase 2, randomized, active-controlled United States (US) study evaluating the safety and efficacy of DARZALEX in combination with VRd in patients with NDMM eligible for HDT and ASCT.1-3

Study Design/Methods

  • Of note, the data presented utilized the abbreviation “RVd”, which has been replaced with “VRd” in the summary below to remain consistent throughout this scientific response.
  • Primary objective: determine if the addition of DARZALEX to VRd will increase the sCR rate by the end of the post-ASCT consolidation therapy.
  • Primary endpoint: sCR (by end of post-ASCT consolidation)
  • Secondary endpoints: MRD (10-5 via next-generation sequencing [NGS]), CR, overall response rate (ORR), very good partial response or better (≥VGPR)

Part 1: Safety Run-in Phase-Final Analysis

Voorhees et al (2021)4 reported the final analysis of the safety run-in cohort of the GRIFFIN study.  

Results

Baseline Characteristics
  • Demographics and baseline characteristics are presented in Table: Baseline Characteristics.
  • Median follow-up was 40.8 months (range, 20.6-43) after patients completed D-VRd treatment and 24 months of D-R maintenance therapy.
  • All patients in the safety run-in phase (N=16) completed induction therapy, stem cell mobilization, ASCT, consolidation, and entered maintenance therapy.
  • A total of 87.5% (n=14) of patients completed study therapy, and 2 (12.5%) discontinued the therapy because of PD (n=1) or AE (n=1; neuralgia or thrombocytopenia) during maintenance therapy.
  • Stem cell collection and neutrophil and platelet engraftment are present in Table: Stem Cell Collection and Transplantation.

Baseline Characteristics4
D-VRd
(n=16)

Age, years
   Median (range)
62.5 (46-65)
      <65 years, n (%)
14 (87.5)
      ≥65 years, n (%)
2 (12.5)
Sex, n (%)
   Male
8 (50)
   Female
8 (50)
Race, n (%)
   White
11 (68.8)
   Black or African American
4 (25)
   Asian
1 (6.3)
ECOG performance statusa, n (%)
   0
3 (18.8)
   1
10 (62.5)
   2
3 (18.8)
ISS disease stageb, n (%)
   I
12 (75)
   II
2 (12.5)
   III
2 (12.5)
Cytogenetic risk profilec, n (%)
   Standard
12 (75)
   High risk
4 (25)
Median (range) time since diagnosis of multiple myeloma, months
1.6 (0-5)
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; ECOG, Eastern Cooperative Oncology Group; ISS, International Staging System.
aECOG performance status is scored on a scale from 0 to 5, with 0 indicating no symptoms and higher scores indicating increasing disability.
bISS disease stage is based on the combination of serum β2-microglobulin and albumin levels. Higher stages indicate more advanced disease.
cCytogenetic risk was assessed by fluorescence in situ hybridization (locally tested); high risk was defined as the presence of del(17p), t(4;14), or t(14;16) in those patients with cytogenetic risk data available.


Stem Cell Collection and Transplantation4
D-VRd
(n=16)

CD34+ yield, median (range) (x 106 cells/kg)
8.05 (3.5-17.6)
CD34+ cells transplanted, median (range) (x 106 cells/kg)
4.72 (2.2-6)
Patients receiving plerixafor for mobilization, n (%)
9 (56.3)
Patients receiving cyclophosphamide, n (%)
0 (0)
Days to neutrophil (0.5 x 109/L) engraftmenta, median (maximum)
14
Days to platelet (20 x 109/L) engraftmentb, median (maximum)
13.5
Abbreviations: CD34+, cluster of differentiation 34 positive; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone.
aFor neutrophil engraftment, there were 15 evaluable patients.
bFor platelet engraftment, there were 16 evaluable patients.

Safety
  • During cycle 1, three of 16 patients developed 4 dose-limiting toxicities (DLTs) fatigue, gastroenteritis, hypotension, and pneumonitis.
    • All DLTs were grade 3 and none resulted in treatment discontinuation during induction or consolidation therapy.
  • One patient had a TEAE leading to discontinuation of study treatment.
  • Fourteen (87.5%) patients experienced any grade infections, and 5 (31.3%) patients experienced grade 3/4 infections.
    • During the maintenance phase 31.3% (n=5) of patients experienced any grade infections. (The most common being upper respiratory tract infections. One (6.3%) patient experienced a grade 3/4 infection (pneumonia and bronchitis).
  • Grade 1/2 infusion-related reactions (IRRs) occurred in 31.3% (n=5) of patients. IRRs included pruritus, chills, flushing, maculo-papular rash, and vascular access site swelling; all occurred during cycle 1 except vascular access site swelling.
  • Eleven (68.6%) patients experienced a serious AE. For the incidences of grade 3/4 TEAEs, see Table: Most Common Grade 3/4 TEAEs.

Most Common Grade 3/4 TEAEs4
Patients, n (%)
D-VRd (n=16)
Grade 3/4a
Total
15 (93.8)
Most commonly occurring
   Neutropenia
7 (43.8)
   Pneumonia
5 (31.3)
   Lymphopenia
5 (31.3)
   Thrombocytopenia
4 (25)
   Hypertension
3 (18.8)
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone, TEAE, treatment emergent adverse event.
aNo Grade 5 TEAEs were reported.

Efficacy
  • At a median follow-up of 40.8 months (range, 20.6-43), disease progression occurred in 3 patients.
  • Median time to first response was 0.77 months (range, 0.1-2.1), and median duration of response was not estimable.
  • Median time to ≥CR was 7.36 months (range, 2.8-18.5), and median duration of ≥CR was not estimable.
  • Estimated 24-month PFS and OS rate was 93.8%.
  • Estimated 36-month PFS and OS rates were 78.1% and 93.8%, respectively.
  • MRD-negativity rates at 10-5 sensitivity threshold and 10-6 sensitivity threshold, respectively:
    • By end of D-VRd induction: 18.8% (n=3) vs 0%
    • By end of D-VRd consolidation: 50% (n=8) vs 0%
    • At the last follow-up: 81.3% (n=13) vs 31.3% (n=5)
  • MRD-negativity rates of 10-5 was sustained for ≥12 months in 8 (50%) patients.
  • Response rates are presented in Table: Updated Response Rates Over Time for the Safety Run-in Cohort.

Updated Response Rates Over Time for the Safety Run-in Cohorta,4
Patients, %
By end of
D-VRd induction

By end of
D-VRd consolidation

By last follow-up
D-R Maintenance
sCR
-
56.3
93.8
CR
12.5
12.5
-
≥CR
12.5
68.8
93.8
VGPR
56.3
31.3
6.3
PR
31.3
-
-
Abbreviations: CR, complete response; D-R, DARZALEX + lenalidomide; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; PR, partial response; sCR, stringent complete response; VGPR, very good partial response.
Response data are shown for the response-evaluable population (N=16).
aPercentages do not add up to 100% due to rounding.

Part 2: Randomized Phase

Voorhees et al (2020)2,3,26 presented the primary analysis and updated analysis of the randomized portion of this study. Kaufman et al (2020)27 presented a 1-year update of safety and efficacy results at the 62nd ASH Annual Meeting & Exposition in December 2020. Laubach et al (2021)28 presented updated efficacy and safety results after 2 years of maintenance therapy in the GRIFFIN Study. Voorhees et al (2023)5 reported the final efficacy and safety results after all patients completed ≥1 year of follow-up after the end of study treatment, died, or withdrew from study participation.

Results

Baseline Characteristics
  • A total of 207 patients were randomized (D-VRd, n=104; VRd, n=103).
  • The median follow-up was 13.5 months in the primary analysis and 22.1 months in the updated analysis.
  • Ninety percent of patients in the D-VRd arm underwent ASCT compared to 76% in the VRd arm (ASCT rate lower due to early discontinuations).
  • Baseline patient demographics are summarized in Table: Patient Demographics in the Randomized Phase (ITT).

Patient Demographics in the Randomized Phase (ITT)3
Characteristic
D-VRd
(n=104)
VRd
(n=103)
Age
   Median (range), years
59 (29-70)
61 (40-70)
   ≥65 years
28 (26.9)
28 (27.2)
Male, n (%)
58 (55.8)
60 (58.3)
ECOG statusa, n (%)
n=101
n=102
   0
39 (38.6)
40 (39.2)
   1
51 (50.5)
52 (51)
   2
11 (10.9)
10 (9.8)
ISS stageb, n (%)
   I
49 (47.1)
50 (48.5)
   II
40 (38.5)
37 (35.9)
   III
14 (13.5)
14 (13.6)
Baseline creatinine clearance, n (%)
   30-50 mL/minute
9 (8.7)
9 (8.7)
   >50 mL/minute
95 (91.3)
94 (91.3)
Cytogenetic profilec, n (%)
n=98
n=97
   Standard risk
82 (83.7)
83 (85.6)
   High risk
16 (16.3)
14 (14.4)
Time since diagnosis of MM
n=103
n=102
   Median (range), months
0.7 (0-12)
0.9 (0-61)
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; ECOG, Eastern Cooperative Oncology Group; ISS, International Staging System; ITT, intention-to-treat; MM, multiple myeloma; VRd, bortezomib + lenalidomide + dexamethasone.
aECOG performance status is scored on a scale from 0 to 5, with 0 indicating no symptoms and higher scores indicating increasing disability.
bISS disease stage is based on the combination of serum-β2-microglobulin and albumin levels. Higher stages indicate more advanced disease.
cCytogenetic risk was assessed by fluorescence in situ hybridization, high risk was defined as the presence of del17p, t(4:14), or t(14:16) among patients with available cytogenetic risk data.

Efficacy
  • Primary endpoint met with D-VRd improving the sCR rate by end of consolidation (D-VRd vs VRd: 42.4% vs 32%; OR, 1.57; 95% CI, 0.87-2.82; 1-sided P=0.068). The study had 80% power to detect a 15% improvement with a 1-sided alpha of 0.1.
  • A significantly higher proportion of patients achieved an ORR following consolidation in the D-VRd group vs the VRd group (99% vs 91.8%; 2-sided P=0.0160).
  • The rate of ≥VGPR was 90.9% with D-VRd vs 73.2% with VRd.
  • The rate of ≥CR was 51.5% with D-VRd vs 42.3% with VRd.
  • The percentage of patients achieving ≥CR at the end of induction, ASCT, consolidation, and clinical cutoff in the D-VRd arm was 19.2%, 27.3%, 51.5%, and 79.8%, respectively, vs 13.4%, 19.6%, 42.3%, and 60.8% in the VRd arm.
  • MRD (10-5 via NGS) among patients achieving ≥CR greater with D-VRd vs VRd: 58.8% vs 24.4% (OR, 4.65; 95% CI, 1.76-12.28, P=0.0014).
  • In the intention-to-treat (ITT) population, 51% of patients in the D-VRd arm achieved post-ASCT MRD-negativity vs 20.4% of patients in the VRd arm, regardless of response (OR, 4.70; 95% CI, 2.38-9.28; P<0.0001). Additional MRD results are in Table: Post-Consolidation MRD-Negativity.
  • Median stem cell yield (D-VRd vs VRd): 8.2 vs 9.4 x 106 cells.

Post-Consolidation MRD-Negativity3
MRD-Negative Status (10-5), n (%); ITTa
D-VRd (n=104)
VRd (n=103)
Odds Ratio (95% CI)b
P-valuec
MRD-negative regardless of response
53/104
(51)

21/103
(20.4)

4.07
(2.18-7.59)

<0.0001
MRD-negative with CR or better
49/104
(47.1)

19/103
(18.4)

3.89
2.07-7.33)

<0.0001
In patients achieving CR or better
49/69
(62)

19/59
(32.2)

3.57
(1.72-7.44)

0.0006
MRD Evaluable Population
53/77
(68.8)

21/65
(32.3)

4.47
(2.19-9.11)

<0.0001
Abbreviations: CI, confidence interval; CR, complete response; CrCl, creatinine clearance; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intention-to-treat; MRD, minimal residual disease; NGS, next-generation sequencing; VRd, bortezomib + lenalidomide + dexamethasone.
aThe threshold of MRD-negativity was defined as 1 tumor cell per 105 white cells. MRD status is based on assessment of bone marrow aspirates by NGS in accordance with International Myeloma Working Group criteria. MRD assessments occurred in patients who had both baseline (with clone identified/calibrated) and postbaseline MRD (with negative, positive, or indeterminate result) samples taken (D-VRd, n = 71; VRd, n = 55). Patients with a missing or inconclusive assessment were considered MRD positive.  
bMantel-Haenszel estimate of the common odds ratio for stratified tables is used. The stratification factors are ISS stage (I, II, III) and CrCl (30-50 mL/min or 50 mL/min) at randomization. An odds ratio .1 indicates an advantage for the DARZALEX group.
cP values were calculated from the Fisher’s exact test.

  • At the median follow up of 22.1 months, D-VRd achieved higher sCR (62.6% vs 45.4%; OR, 1.98; 95% CI, 1.12-3.49; 2-sided P=0.0177), CR (17.2% vs 15.5%), and ≥CR (79.8% vs 60.8%, OR, 2.53; 95% CI, 1.33-4.81; 2-sided P=0.0045) vs VRd.
  • MRD-negativity rates in the D-VRd and VRd arms, respectively (10-5 sensitivity threshold):
    • D-VRd (n=104) and VRd (n=103) (ITT): MRD-negative: 51% vs 20.4% (P<0.0001); MRD-negative & ≥CR: 47.1% vs 18.4% (P<0.0001)
      • ≥CR: D-VRd (n=79) and VRd (n=59): MRD-negative: 62% vs 32.2% (P=0.0006)
      • MRD evaluable: D-VRd (n=77) and VRd (n=65): MRD-negative: 68.8% vs 32.3% (P<0.0001)
  • In the ITT population, median PFS and OS were not reached in the D-VRd and VRd arms. In the D-VRd and VRd arms, respectively (Kaplan-Meier estimates):
    • 12-month PFS rates were 96.9% and 95.3%.
    • 24-month PFS rates were 95.8% and 89.8%.
    • 12-month OS rates were 99% and 97.9%.
    • 24-month OS rates were 95.8% and 93.4%.
  • DARZALEX did not impact time to engraftment and hematopoietic reconstitution (Table: Stem Cell Collection and Transplantation).

Stem Cell Collection and Transplantation26
D-VRd
VRd
Median (range) stem cell yielda,b, x 106 CD34+ cells/kg
8.2 (3-33)
9.4 (4-29)
Median stem cells transplantedc, x 106 CD34+ cells/kg
4.2
4.8
Patients receiving plerixafor for mobilizationd, n(%)
66 (70)
45 (56)
Patients receiving cyclophosphamided, n (%)
5 (5)
4 (5)
Median (max) days to neutrophil engraftment (0.5 x 109/L)
12 (31)
12 (23)
Median (max) days to platelet engraftment (20 x 109/L)
13 (31)
12 (23)
Abbreviations: CD34+, cluster of differentiation 34 positive; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; G-CSF, granulocyte colony-stimulating factor; max, maximum; VRd, bortezomib + lenalidomide + dexamethasone.
aAmong patients who underwent peripheral blood stem cell apheresis (D-VRd, n=93; VRd, n=80).
bOne patient in the D-VRd group had a stem cell yield <3 x 106 cells/kg; no patients in either group had a stem cell yield <2 x 106 cells/kg.
cAmong patients receiving transplant (D-VRd, n=94; VRd, n=78).
dAmong patients who underwent mobilization (D-VRd, n=95; VRd, n=80). Patients underwent stem cell mobilization with G-CSF with or without plerixafor, according to institutional standards; if unsuccessful, cyclophosphamide-based mobilization was permitted.

Safety
  • In the updated safety and efficacy analysis, any grade infections occurred in 91% of patients in the D-VRd arm and 62% of patients in the VRd arm, the most common being grade 1/2 upper respiratory tract infections; grade 3/4 infections were seen in 23% of patients in the D-VRd arm vs 22% of patients in the VRd arm. See Table: Most Common TEAEs.
    • Pneumonia was reported in 13% of patients in the D-VRd arm and 15% of patients in the VRd arm. See Table: Most Common TEAEs.

Most Common TEAEsa,3,26
Event, n (%)
D-VRd
(n=99)

VRd
(n=102)

Any Grade
Grade 3/4
Any Grade
Grade 3/4
Hematologic
   Neutropenia
57 (57.6)
41 (41.4)
36 (35.3)
22 (21.6)
   Thrombocytopenia
43 (43.4)
16 (16.2)
36 (35.3)
9 (8.8)
   Leukopenia
36 (36.4)
16 (16.2)
29 (28.4)
7 (6.9)
   Anemia
35 (35.4)
9 (9.1)
33 (32.4)
6 (5.9)
   Lymphopenia
30 (30.3)
23 (23.2)
28 (27.5)
22 (21.6)
Non-hematologic
   Fatigue
68 (68.7)
6 (6.1)
62 (60.8)
6 (5.9)
   Upper respiratory tract infection
62 (62.6)
1 (1)
45 (44.1)
2 (2)
   Peripheral neuropathyb
59 (59.6)
7 (7.1)
74 (72.5)
8 (7.8)
   Diarrhea
59 (59.6)
7 (7.1)
51 (50)
4 (3.9)
   Constipation
51 (51.5)
2 (2)
40 (39.2)
1 (1)
   Cough
50 (50.5)
0 (0)
27 (26.5)
0 (0)
   Nausea
49 (49.5)
2 (2)
50 (49)
1 (1)
   Pyrexia
45 (45.5)
2 (2)
28 (27.5)
3 (2.9)
   Insomnia
42 (42.4)
2 (2)
31 (30.4)
1 (1)
   Back pain
36 (36.4)
1 (1)
34 (33.3)
4 (3.9)
   Edema peripheral
34 (34.3)
2 (2)
35 (34.3)
3 (2.9)
   Arthralgia
33 (33.3)
0 (0)
33 (32.4)
2 (2)
Infusion-related reactions
42 (42.4)
6 (6)c
-
-
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; TEAE, treatment-emergent adverse event; VRd, bortezomib + lenalidomide + dexamethasone.
aAny grade TEAEs are listed that occurred in ≥30% of patients in either group. The safety analysis population included all randomized patients who received ≥1 dose of study treatment; analysis was according to treatment received.
bIncludes patients with neuropathy peripheral and peripheral sensory neuropathy.
cNo grade 4 infusion-related reactions were reported.

Final Efficacy and Safety Analysis of Maintenance Therapy

Voorhees et al (2023)5 reported the final efficacy and safety results after all patients completed ≥1 year of follow-up after the end-of-study treatment, died, or withdrew from study participation.

Results

Patient Characteristics
  • At the time of the final analysis, all patients completed ≥1 year of follow-up after the end-of-study treatment, died, or withdrew from the study.
  • The median duration of follow-up was 49.6 months (interquartile range [IQR], 47.4-52.1).
  • By the final analysis, 25% of patients in the D-VRd arm and 51% in the VRd arm discontinued treatment. See Table: Patient Disposition.
  • The median duration of treatment in the D-VRd and VRd arms was 32.5 months (IQR, 31.1-33.4) and 27.5 months (IQR, 2.9-32.7), respectively.
    • In the D-VRd arm, among the 90 patients who received DARZALEX and lenalidomide maintenance therapy, 21% (n=19) switched from DARZALEX to DARZALEX FASPRO and received ≥1 cycle of DARZALEX FASPRO (median number, 3 [IQR, 3-5]).

Patient Disposition5
Patients, n
D-VRd (n=104)
VRd (n=103)
Treated with maintenance therapy
90
70
Completed maintenance therapy
74
48
Discontinued treatment during maintenance therapy
16
22
   AE
6
7
   PD
3
8
   Patient withdrawal
2
4
   Lost to follow-up
2
0
   Death
1
1
   Other
2
2
Discontinued treatment by final analysis
26
53
Abbreviations: AE, adverse event; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; PD, progressive disease; VRd, bortezomib + lenalidomide + dexamethasone.
Efficacy
  • At the final analysis, among response-evaluable patients in the D-VRd (n=100) vs VRd (n=98) arm, respectively, sCR was achieved in 67% vs 48% of patients (OR, 2.18; 95% CI, 1.22-3.89; 2-sided P=0.0079) and ≥CR in 83% vs 60% of patients (P=0.0005). Response data over time are summarized in Table: Summary of Response Over Time.5,29

Summary of Response Over Time5
Timepoint, %
D-VRd
VRd
sCR
CR
≥CR
VGPR
PR
SD/PD/
NE

sCR
CR
≥CR
VGPR
PR
SD/PD/
NE

End of inductiona
12
7
19
53
26
2
7
6
13
43
35
8
End of post-ASCT consolidationa
42
9
52
39
8
1
32
10
42
31
19
8
Final analysisb
67
16
83
13
3
1
48
12
60
17
14
8
Abbreviations: ASCT, autologous stem cell transplant; CR, complete response; ≥CR, complete response or better; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; IQR, interquartile range; NE, not evaluable; PD, progressive disease; PR, partial response; sCR, stringent complete response; SD, stable disease; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone.
Rates shown are the number of patients with each type of response divided by the response-evaluable population.aResponse rates were from the primary analysis cutoff (median follow-up, 13.5 months) and the response-evaluable population comprised 196 patients (D-VRd, n=99; VRd, n=97).
bResponse rates were also evaluated at the time of the final analysis (median follow-up 49·6 months; IQR 47·4-52·1), and the response-evaluable population comprised 198 patients (D-VRd, n=100; VRd, n=98).


Response Duration Among Patients in the D-VRd vs VRd Arm5
Parameter
D-VRd
VRd
Median duration to first response (ORR), months (95% CI)
0.8 (0.8-0.8)
0.8 (0.8-1)
Median duration to sCR, months (95% CI)
10.2 (8.8-13)
14.3 (9.2-21.7)
   HR (95% CI)
1.26 (0.86-1.83)
P value
0.2339
Median duration to ≥VGPR, months (95% CI)
2.2 (2.1-2.7)
3 (2.2-6.3)
Median duration to ≥CR, months (95% CI)
8.9 (7.9-9.4)
9.6 (8.4-12.2)
Median DOR
NR
NR
   Estimated 48-month DOR, % (95% CI)
89 (79.9-94.3)
71 (55.8-81.4)
Abbreviations: CI, confidence interval; ≥CR, complete response or better; DOR, duration of response; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; HR, hazard ratio; NR, not reached; ORR, overall response rate; sCR, stringent complete response; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone.

Final Analysis of Best Response and MRD-Negativity Rates at the End of Maintenance5,29
Parameter
D-VRd
VRd
P value
Responsea, n
100
98
-
   ORR, n (%)
99 (99)
90 (92)
0.016b
      ≥CR
83 (83)
59 (60)
0.0005b
      CR
16 (16)
12 (12)
-
      sCR
67 (67)
47 (48)
0.0079b
      ≥VGPR
96 (96)
76 (78)
0.0002b
      VGPR
13 (13)
17 (17)
-
      PR
3 (3)
14 (14)
-
   SD, n (%)
1 (1)
8 (8)
-
   PD, n (%)
0
0
-
MRD negative
   ITT population, n
104
103
-
      10-5 sensitivity, n (%)
67 (64)
31 (30)
<0.0001c
         OR (95% CI)
4.23 (2.35-7.62)
      10-6 sensitivity, n (%)
37 (36)
16 (16)
0.0013c
         OR (95% CI)
2.95 (1.52-5.75)
   In patients achieving ≥CR, n
83
59
-
      10-5 sensitivity, n (%)
64 (77)
28 (47)
0.0004c
      10-6 sensitivity, n (%)
35 (42)
14 (24)
0.031c
Durable MRD-negativity
   Lasting ≥12 months, n
104
103
-
      10-5 sensitivity, n (%)
46 (44)
14 (14)
<0.0001c
         OR (95% CI)
5 (2.50-9.99)
      10-6 sensitivity, n (%)
10 (10)
4 (4)
0.16c
         OR (95% CI)
2.48 (0.76-8.07)
Abbreviations: CI, confidence interval; ≥CR, complete response or better; CR, complete response; CrCl, creatinine clearance; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intention-to-treat; MM, multiple myeloma; MRD, minimal residual disease; OR, odds ratio; ORR, overall response rate; PD, progressive disease; PR, partial response; sCR, stringent complete response; SD, stable disease; ≥VGPR, very good partial response or better; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone.
The predefined per protocol final analysis occurred after all patients completed ≥1 year of long-term follow-up after the end-of-study treatment, died, or withdrew from study participation, whichever occurred first.
aResponse rate is based on the response-evaluable population, which included randomized patients who had a confirmed diagnosis of MM, had measurable disease at baseline, received ≥1 dose of study treatment, and had ≥1 postbaseline disease assessment. The response-evaluable population for the primary analysis included 99 patients in the D-VRd group and 97 patients in the VRd group.
bP value was calculated using the Cochran-Mantel-Haenszel Chi-square test stratified by ISS disease stage (I, II, or III) and baseline CrCl (30-50 mL/min or >50 mL/min) at randomization.
cP value was calculated using Fisher’s exact test.

  • By the end of the 2-year maintenance therapy, 14% (n/N=15/104) and 10% (n/N=10/103) of patients in the D-VRd and VRd arms, respectively, who were previously MRD-positive at the end of the consolidation phase, converted to MRD negative (10-5). MRD-negativity rates continuously improved over time and were consistently higher in the D-VRd vs VRd arm. See Table: Summary of MRD-Negativity Rates Over Time (ITT Population).

Summary of MRD-Negativity Rates Over Time (ITT Population)a,5,29
Timepoint, %
D-VRd
VRd
MRD-Negativity (10-5)
MRD-Negativity (10-6)
MRD-Negativity (10-5)
MRD-Negativity (10-6)
End of induction
22
1
8
0
Post-ASCT consolidation
50
11
20
3
End of study
64
36
30
16
Abbreviations: ASCT, autologous stem cell transplant; CR, complete response; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; ITT, intention-to-treat; MRD, minimal residual disease; NGS, next-generation sequencing; sCR, stringent complete response; VRd, bortezomib + lenalidomide + dexamethasone. aMRD was evaluated by NGS using the clonoSEQ assay. MRD assessments occurred at the first evidence of suspected CR or sCR, after induction (but before stem cell collection), after consolidation, and after 12 and 24 months of maintenance, regardless of response.
  • No patient in either treatment arm with sustained MRD-negativity 10-5 lasting ≥12 months became MRD-positive later.
  • The median time to MRD-negativity in the D-VRd vs VRd arm at sensitivity thresholds of 10-5 and 10-6, respectively, was 8.5 vs 34.6 months (HR, 2.70; 95% CI, 1.72-4.23; P<0.0001) and 33.9 months vs not reached (NR; HR, 1.93; 95% CI, 1.05-3.54; P=0.031).
  • Efficacy and survival outcomes are summarized in Table: Efficacy and Survival Outcomes (ITT Population).

Efficacy and Survival Outcomes (ITT Population)5,29
Parameter
D-VRd
VRd
Median PFS, months
NR
NR
   3-year PFS rate, %
89
80.7
   4-year PFS rate, %
87.2
70
   PFS HR (95% CI); P value
0.45 (0.21-0.95); 0.032
Median PFS in patients who received lenalidomide therapy as per SoC after study completion, months
NR
NR
4-year PFS rate in patients who received SoC lenalidomide therapy after study completion, %
96
80
Median PFS in patients who did not receive lenalidomide therapy as per SoC after study completion, months
NR
NR
4-year PFS rate in patients who did not receive SoC lenalidomide therapy after study completion, %
100
86
Median OS, months
NR
NR
   3-year OS rate, %
92.7
92.2
   4-year OS rate, %
92.7
92.2
   OS HR (95% CI); P value
0.90 (0.31-2.56); 0.84a
Disease progression or death, n/N (%)
11/104 (11)
18/103 (17)
   HR (95% CI)
0.45 (0.21-0.95)
P value
0.032
Abbreviations: CI, confidence interval; CrCl, creatinine clearance; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; HR, hazard ratio; ISS, International Staging System; ITT, intention-to-treat; NR, not reached; OS, overall survival; PFS, progression-free survival; SoC, standard of care; VRd, bortezomib + lenalidomide + dexamethasone.
a
HR and 95% CI are from a Cox proportional hazards model with treatment as the sole explanatory variable and stratified with ISS staging (I, II, and III) and baseline CrCl (30-50 mL/min or >50 mL/min) at randomization.
An HR <1 indicates an advantage for D-VRd. P value is based on the log-rank test stratified with ISS staging and baseline CrCl at randomization.

Safety
  • Among safety-evaluable patients in the D-VRd (n=99) vs VRd (n=102) arms, grade 3/4 TEAEs occurred in 86% (n=85) vs 79% (n=81), respectively.
  • In the D-VRd vs VRd arm, serious TEAEs occurred in 46% (n=46) vs 52% (n=53) of patients, respectively.
    • The most common serious TEAEs included pneumonia (15% vs 14%) and pyrexia (11% vs 10%).
  • TEAEs leading to treatment discontinuation were similar across treatment arms (D-VRd, 33% [n=33]; VRd, 31% [n=32]). One patient in each arm died due to TEAEs unrelated to study treatment.
  • Any-grade infections were more common in the D-VRd vs VRd arm (93% [n=92] vs 66% [n=67]). Similar incidence rates were reported across treatment arms for grade 3/4 infections (D-VRd, 29%; VRd, 26%) and infections leading to treatment discontinuation (D-VRd, 2%; VRd, 3%).
    • During maintenance therapy (cycle 7 and onwards) in the D-VRd vs VRd arm, any-grade infections occurred in 35% (n/N=31/89) vs 32% (n/N=23/71) of patients and grade 3/4 infections occurred in 18% (n=16) vs 21% (n=15) of patients.
    • In the D-VRd vs VRd arm, coronavirus disease 2019 (COVID-19) infections were reported in 5% (n=5) vs 2% (n=2) of patients, respectively. Of these, 1 patient in each arm had a grade 3 COVID-19-related event (including 1 serious event in the D-VRd arm).
  • TEAEs occurring in the safety population are summarized in Table: Most Common TEAEs in the Safety Population.
  • During maintenance therapy, second primary malignancies with first onset after the start of maintenance therapy were reported in 4 of 89 (4%) evaluable patients in the D-VRd arm and 3 of 71 (4%) evaluable patients in the VRd arm.
  • A total of 14 (D-VRd, n=7; VRd, n=7) patients died, of whom 9 (D-VRd, n=5; VRd, n=4) patients died due to PD.
  • There were 39 [39%] IRRs reported at the initial infusion, 2 [2%] IRRs at the second infusion, and 14 [14%] IRRs at the subsequent infusions.29

Most Common TEAEs in the Safety Populationa,5,29
TEAEs, n (%)
D-VRd (n=99)
VRd (n=102)
Grade 1/2
Grade 3
Grade 4
Grade 1/2
Grade 3
Grade 4
Hematologic
   Anemia
28 (28)
9 (9)
0 (0)
27 (26)
5 (5)
1 (1)
   Thrombocytopenia
28 (28)
4 (4)
12 (12)
27 (26)
4 (4)
5 (5)
   Leukopenia
22 (22)
8 (8)
9 (9)
22 (22)
6 (6)
2 (2)
   Neutropenia
17 (17)
32 (32)
14 (14)
18 (18)
21 (21)
2 (2)
   Lymphopenia
8 (8)
13 (13)
10 (10)
6 (6)
20 (20)
3 (3)
Non-hematologic
   Hypokalemia
24 (24)
3 (3)
1 (1)
24 (24)
3 (3)
0 (0)
   Hypocalcemia
17 (17)
0 (0)
0 (0)
12 (12)
2 (2)
1 (1)
   Pneumoniab
11 (11)
11 (11)
1 (1)
4 (4)
14 (14)
0 (0)
   Hyperkalemia
6 (6)
1 (1)
0 (0)
1 (1)
0 (0)
1 (1)
   Cellulitis
6 (6)
0 (0)
1 (1)
3 (3)
1 (1)
0 (0)
   Hypophosphatemia
5 (5)
9 (9)
1 (1)
6 (6)
11 (11)
0 (0)
   Hyperuricemia
4 (4)
0 (0)
0 (0)
6 (6)
0 (0)
1 (1)
   Acute kidney injury
2 (2)
2 (2)
2 (2)
4 (4)
3 (3)
0 (0)
   Atrial fibrillation
1 (1)
0 (0)
1 (1)
3 (3)
0 (0)
0 (0)
   Increased blood creatine phosphokinase
1 (1)
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
   Atrial tachycardia
1 (1)
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
   Sepsis
0 (0)
1 (1)
2 (2)
0 (0)
1 (1)
0 (0)
   Drug reaction with eosinophilia and systemic symptoms
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
   Septic shock
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
   Cerebrovascular accident
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
   Systemic inflammatory
   response syndrome
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
   Death
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
IRRsc
49 (49)
7 (7)
0 (0)
-
-
-
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; IRR, infusion-related reaction; TEAE, treatment-emergent adverse event; VRd, bortezomib + lenalidomide + dexamethasone.
aThe maximum intensity for each preferred term is listed, and TEAEs are listed for all grade 4 or 5 events and any grade 3 events occurring in ≥10% of patients in either treatment arm (corresponding grade 1-2 events are listed).
bOne grade 5 event was recorded in the D-VRd group.
cThere were no grade 4/5 IRRs. Data pertaining to IRRs are not available for the VRd arm.

Post Hoc Analysis of the Incidence of VTE in the GRIFFIN Study

Sborov et al (2022)6 conducted a post-hoc analysis to evaluate the risk and incidence of VTE in patients receiving D-VRd vs VRd in the GRIFFIN study.

Study Design/Methods

  • The risk of VTE was assessed using the SAVED risk assessment model (<2 points, low risk; >2 points, high risk), as described below:
    • Surgery within 90 days (S; +2)
    • Asian race (A; -3)
    • History of VTE (V; +3),
    • Eighty (age ≥80 years; E; +1), and
    • Dexamethasone dose (D; +2 for high, +1 for standard)
  • All patients received VTE prophylaxis with at least aspirin ≥162 mg/day. Patients with increased risk of VTE received prophylaxis with enoxaparin 40 mg/day subcutaneous (SC) or other low-molecular-weight heparin (LMWH) at an equivalent dose and frequency. Vitamin K antagonists, factor Xa inhibitors, or direct thrombin inhibitors could be used at the treating physician's discretion.

Results

Patient Characteristics

Demographic and Baseline Disease Characteristics Among Patients Who Did or Did Not Experience VTEs6
Characteristic
Patients who experienced VTEs
Patients who did not experience VTEs
Total (n=26)
D-VRd (n=10)
VRd (n=16)
Total (n=181)
D-VRd (n=94)
VRd
(n=87)

Age, years
   Median (range)
57.5
(35-70)

54
(35-70)

59.5
(47-70)

60
(29-70)

59.5
(29-70)

61
(40-70)

   <65, n (%)
17 (65.4)
7 (70)
10 (62.5)
134 (74)
69 (73.4)
65 (74.7)
   ≥65, n (%)
9 (34.6)
3 (30)
6 (37.5)
47 (26)
25 (26.6)
22 (25.3)
Male, n (%)
19 (73.1)
8 (80)
11 (68.8)
99 (54.7)
50 (53.2)
49 (56.3)
Weight, kg
n=26
n=10
n=16
n=179
n=92
n=87
   Median (range)
87.1
(62-148.5)

81.9
(63.6-141.5)

88.2
(62-148.5)

80.4
(37.4-158.6)

78.9 (48.8-158.6)
82.7 (37.4-150.1)
ECOG PS score, n (%)
n=26
n=10
n=16
n=177
n=91
n=86
   0
3 (11.5)
1 (10)
2 (12.5)
76 (42.9)
38 (41.8)
38 (44.2)
   1
17 (65.4)
7 (70)
10 (62.5)
86 (48.6)
44 (48.4)
42 (48.8)
   2
6 (23.1)
2 (20)
4 (25)
15 (8.5)
9 (9.9)
6 (7)
ISS disease stagea, n (%)
   I
13 (50)
6 (60)
7 (43.8)
86 (47.5)
43 (45.7)
43 (49.4)
   II
13 (50)
4 (40)
9 (56.3)
64 (35.4)
36 (38.3)
28 (32.2)
   III
0 (0)
0 (0)
0 (0)
28 (15.5)
14 (14.9)
14 (16.1)
   Missing
0 (0)
0 (0)
0 (0)
3 (1.7)
1 (1.1)
2 (2.3)
Type of measurable diseaseb, n (%)
   Serum and urine
4 (15.4)
1 (10)
3 (18.8)
33 (18.2)
22 (23.4)
11 (12.6)
   Free light chain
3 (11.5)
2 (20)
1 (6.3)
23 (12.7)
13 (13.8)
10 (11.5)
   Serum only
14 (53.8)
4 (40)
10 (62.5)
99 (54.7)
49 (52.1)
50 (57.5)
   Urine only
5 (19.2)
3 (30)
2 (12.5)
22 (12.2)
9 (9.6)
13 (14.9)
   Not evaluable
0 (0)
0 (0)
0 (0)
4 (2.2)
1 (1.1)
3 (3.4)
Bone marrow involvement (% plasma cells, bone marrow biopsy/aspirate)c, n (%)
   <10
3 (11.5)
1 (10)
2 (12.5)
13 (7.2)
9 (9.6)
4 (4.6)
   10-59
9 (34.6)
4 (40)
5 (31.3)
88 (48.6)
42 (44.7)
46 (52.9)
   ≥60
14 (53.8)
5 (50)
9 (56.3)
73 (40.3)
40 (42.6)
33 (37.9)
   Missing
0 (0)
0 (0)
0 (0)
7 (3.9)
3 (3.2)
4 (4.6)
Time from MM diagnosis to randomization
n=26
n=10
n=16
n=179
n=93
n=86
   Median (range), months
0.8 (0-3)
0.4 (0-3)
0.9 (0-2)
0.8 (0-61)
0.7 (0-12)
0.9 (0-61)
Cytogenetic profiled, n (%)
n=25
n=10
n=15
n=170
n=88
n=82
   Standard risk, n (%)
21 (84)
8 (80)
13 (86.7)
144 (84.7)
74 (84.1)
70 (85.4)
   High risk, n (%)
4 (16)
2 (20)
2 (13.3)
26 (15.3)
14 (15.9)
12 (14.6)
      del17p
4 (16)
2 (20)
2 (13.3)
10 (5.9)
6 (6.8)
4 (4.9)
      t(4;14)
1 (4)
0 (0)
1 (6.7)
13 (7.6)
8 (9.1)
5 (6.1)
      t(14;16)
0 (0)
0 (0)
0 (0)
4 (2.4)
1 (1.1)
3 (3.7)
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; Ig, immunoglobulin; ISS, International Staging System; MM, multiple myeloma; VRd, bortezomib + lenalidomide + dexamethasone; VTE, vascular thrombotic event.
aBased on the combination of serum β2-microglobulin and albumin.
bIncludes IgD, IgM, IgE, and biclonal.
cHighest value by biopsy or aspirate.
dCytogenetic risk was based on local fluorescence in situ hybridization or karyotype analysis. Patients with high-risk cytogenetics had a del17p, t(4;14) or t(14;16) abnormality; a patient could be counted in more than one subcategory. Patients with standard-risk cytogenetic abnormalities had an absence of high-risk cytogenetic abnormalities.

Efficacy

Response Rates in Patients With VTE6
Para-
meter, %

D-VRd (n=10)
VRd (n=16)
End of Induction
End of ASCT
End of Consolidation
After 2 Years of Maintenance
End of Induction
End of ASCT
End of Consolidation
After 2 Years of Maintenance
≥CR
20
20
50
90
25
31.3
31.3
68.8
   sCR
10
10
40
70
18.8
25
25
56.3
   CR
10
10
10
20
6.3
6.3
6.3
12.5
VGPR
50
60
50
10
50
50
56.3
18.8
PR
30
20
-
-
25
18.8
12.5
12.5
Abbreviations: ASCT, autologous stem cell transplant; CR, complete response; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; PR, partial response; sCR, stringent complete response; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone; VTE, vascular thrombotic event.

Response Rates in Patients With VTE at the First Onset of VTE6
Parameter, %
D-VRd (n=10)
VRd (n=16)
≥CR
50
31.3
   sCR
30
25
   CR
20
6.3
VGPR
30
43.8
PR
20
12.5
SD/PD/NE
-
12.5
Abbreviations: CR, complete response; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; NE, not evaluable; PD, progressive disease; PR, partial response; sCR, stringent complete response; SD, stable disease; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone; VTE, vascular thrombotic event.
Safety
  • In the D-VRd vs VRd arm, VTEs occurred in 10.1% (n=10) vs 15.7% (n=16) of patients in the safety analysis population. See Table: VTEs in the Safety Analysis Population.
  • The median time to first onset of VTE was 305 days (range, 6-810) in the D-VRd arm and 119 days (range, 21-822) in the VRd arm.
  • In the D-VRd vs VRd arm, new onset VTEs were reported at the following stages:
    • Induction (cycles 1-4): 5.1% (n=5) vs 8.8% (n=9)
    • Consolidation (cycles 5-6): 0% (n=0) vs 1.4% (n=1)
    • Maintenance:
      • Cycles 7-18: 2.2% (n=2) vs 7% (n=5)
      • Cycles 18+: 3.6% (n=3) vs 1.7% (n=1)
  • In the D-VRd vs VRd arm, the median SAVED score in patients in the ITT population was 0 (range, 0 to 3) vs 0 (range, -3 to 4) and in patients experiencing VTE was 0 (range, 0-3) vs 0.5 (range, 0-4).
  • The median number of cardiovascular comorbidities both in patients in the overall population and patients experiencing VTE was 1.
  • In the D-VRd vs VRd arm, anti-thrombosis prophylaxis at any time was received by 84.8% (n=84) vs 83.3% (n=85) of patients in the overall safety population and by 80% (n=8) vs 93.8% (n=15) of patients who developed VTE.
    • Overall, 60% (n=6) of patients in the D-VRd arm and 68.8% (n=11) of patients in the VRd arm were receiving anti-thrombosis prophylaxis during the first onset of VTE.

VTEs in the Safety Analysis Populationa,6
VTE, n (%)
D-VRd (n=99)
VRd (n=102)
Grade 1
Grade 2-4
Total
Grade 1
Grade 2-4
Total
Total number with ≥1 VTE
1 (1)
9 (9.1)
10 (10.1)
1 (1)
15 (14.7)
16 (15.7)
Embolic and thrombotic events
2 (2)
3 (3)
5 (5.1)
1 (1)
10 (9.8)
11 (10.8)
   Deep vein thrombosis
1 (1)
1 (1)
2 (2)
0 (0)
7 (6.9)
7 (6.9)
   Pulmonary embolism
0 (0)
2 (2)
2 (2)
0 (0)
4 (3.9)
4 (3.9)
   Embolism venous
0 (0)
0 (0)
0 (0)
1 (1)
0 (0)
1 (1)
   Jugular vein thrombosis
0 (0)
1 (1)
1 (1)
0 (0)
1 (1)
1 (1)
   Subclavian vein thrombosis
0 (0)
1 (1)
1 (1)
0 (0)
0 (0)
0 (0)
   Thrombophlebitis superficial
1 (1)
1 (1)
2 (2)
0 (0)
0 (0)
0 (0)
Unspecified and mixed arterial and venous
0 (0)
6 (6.1)
6 (6.1)
1 (1)
5 (4.9)
6 (5.9)
   Embolism
0 (0)
2 (2)
2 (2)
1 (1)
2 (2)
3 (2.9)
   Cerebral congestion
0 (0)
2 (2)
2 (2)
0 (0)
1 (1)
1 (1)
   Cerebrovascular accident
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
1 (1)
   Hemiparesis
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
1 (1)
   Intestinal infarction
0 (0)
1 (1)
1 (1)
0 (0)
0 (0)
0 (0)
   Vascular access site thrombosis
0 (0)
1 (1)
1 (1)
0 (0)
0 (0)
0 (0)
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; VRd, bortezomib + lenalidomide + dexamethasone; VTE, vascular thrombotic event.
aNo grade 5 VTEs were reported in either treatment arm.

Post Hoc Analysis for Stem Cell Mobilization Yields

Chhabra et al (2023)7 conducted a post hoc analysis to evaluate stem cell mobilization, collection yields, and hematopoietic recovery following ASCT after frontline DARZALEX- and lenalidomide-containing quadruplet induction in patients with NDMM from the GRIFFIN and MASTER studies.

Study Design/Methods

  • In the GRIFFIN study, patients at 35 US sites underwent stem cell mobilization with granulocyte colony-stimulating factor (G-CSF) with or without plerixafor (upfront/rescue) after 4 induction cycles (12 weeks) of D-VRd or VRd. If plerixafor-facilitated mobilization was unsuccessful, cyclophosphamide-based mobilization was permitted. After mobilization, patients received melphalan 200 mg/m2, followed by ASCT. A summary of patients from the GRIFFIN study who underwent stem cell mobilization has been presented in Table: Summary of Patients From the GRIFFIN Study Who Underwent Stem Cell Mobilization.
    • The upfront plerixafor usage strategy comprised planned use of plerixafor, irrespective of blood CD34+ count on day 4 of G-CSF administration.
    • The rescue plerixafor usage strategy comprised “just in time” use of plerixafor based on peripheral blood CD34+ count after G-CSF administration.

Summary of Patients From the GRIFFIN Study Who Underwent Stem Cell Mobilization7

n
D-VRd (n=104)
VRd (n=103)
Underwent Mobilization
95
80
   Upfront plerixafor strategy
49
31
      Received plerixafor
49
31
         Underwent ASCT
49
31
   Rescue plerixafor strategya
46
49
      Received plerixafor
19
13
         Underwent ASCT
18
13
      Received G-CSF
19
28
         Underwent ASCT
19
26
Abbreviations: ASCT, autologous stem cell transplant; CD, cluster of differentiation; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; G-CSF, granulocyte colony-stimulating factor; VRd, bortezomib + lenalidomide + dexamethasone.
aPatients who did not receive upfront plerixafor were treated using the rescue plerixafor strategy, defined as administering plerixafor only when deemed necessary and “just in time” based on preapheresis blood CD34+ count after G-CSF. Patients in the rescue plerixafor strategy group did not receive upfront plerixafor but received either rescue plerixafor or no plerixafor. Among the mobilized patients in GRIFFIN, 9 (D-VRd group, n=5; VRd group, n=4) received cyclophosphamide, and accurate information on the mobilization regimen used was not available for 16 patients (D-VRd group, n=8; VRd group, n=8).

Results

Patient Characteristics
  • Among the 207 randomized patients in the GRIFFIN study, 91% (95 of 104) in the D-VRd group and 78% (80 of 103) in the VRd group underwent stem cell mobilization. Baseline characteristics of patients who received D-VRd and VRd in the GRIFFIN study based on upfront and rescue plerixafor received are presented in Table: Baseline Characteristics of Patients Who Received D-VRd and VRd in the GRIFFIN Study.
  • Among the mobilized patients in the D-VRd vs VRd arm, 99% (94 of 95) vs 98% (78 of 80) underwent ASCT, respectively.

Baseline Characteristics of Patients Who Received D-VRd and VRd in the GRIFFIN Study7
Characteristic
D-VRd
VRd
All Patients
Received Upfront Plerixafor
Rescue Plerixafor Strategy
(n=46)a

All Patients
Received Upfront Plerixafor
Rescue Plerixafor Strategy
(n=49)a

Received Rescue Plerixafor
Received G-CSF Only
Received Rescue Plerixafor
Received G-CSF Only
Patients who underwent mobilization, n
95
49
19
19
80
31
13
28
Median age (range), years
59
(29-70)

60
(29-70)

55
(35-70)

58
(34-70)

59
(40-70)

59
(47-70)

62
(49-69)

61
(40-68)

   <65, n (%)
69 (73)
37 (76)
12 (63)
12 (63)
60 (75)
24 (77)
7 (54)
21 (75)
   ≥65, n (%)
26 (27)
12 (24)
7 (37)
7 (37)
20 (25)
7 (23)
6 (46)
7 (25)
Male, n (%)
51 (54)
28 (57)
8 (42)
12 (63)
43 (54)
14 (45)
6 (46)
17 (61)
ISS disease stageb, n (%)
   I
46 (48)
22 (45)
10 (53)
10 (53)
38 (48)
16 (52)
4 (31)
13 (46)
   II
36 (38)
19 (39)
6 (32)
8 (42)
29 (36)
9 (29)
7 (54)
11 (39)
   III
13 (14)
8 (16)
3 (16)
1 (5)
12 (15)
5 (16)
2 (15)
4 (14)
   Missing
0 (0)
0 (0)
0 (0)
0 (0)
1 (1)
1 (3)
0 (0)
0 (0)
Cytogenetic riskc, n
91
47
18
18
77
29
13
28
   Standard
   risk, n (%)

75 (82)
38 (81)
13 (72)
18 (100)
66 (86)
24 (83)
12 (92)
23 (82)
   High risk, n
   (%)

16 (18)
9 (19)
5 (28)
0 (0)
11 (14)
5 (17)
1 (8)
5 (18)
Abbreviations: D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; G-CSF, granulocyte colony-stimulating factor; ISS, International Staging System; VRd, bortezomib + lenalidomide + dexamethasone.
aAmong the mobilized patients in GRIFFIN, 9 (D-VRd, n=5; VRd, n=4) received cyclophosphamide, and no accurate information on the mobilization regimen used was available for 16 patients (D-VRd, n=8; VRd, n=8).
bISS staging is based on the combination of serum β2-microglobulin and albumin. Higher stages indicate more advanced disease.
cCytogenetic risk was assessed using fluorescence in situ hybridization (local testing); high risk was defined as the presence of del17p, t(4;14), or t(14;16) among patients with available cytogenetic risk data.

Stem Cell Mobilization
  • The median duration of stem cell collection was 2 days for patients receiving D-VRd and 1 day for patients receiving VRd. See Table: Summary of ASCT and Stem Cell Mobilization in the GRIFFIN Study.
  • Among patients who underwent mobilization, plerixafor use was more prevalent in those receiving D-VRd (72%) compared with those receiving VRd (55%).
CD34+ Stem Cell Yield
  • The median CD34+ stem cell yield in the D-VRd vs VRd arm was 8.3×106 cells/kg vs 9.4×106 cells/kg, respectively, and was numerically higher for patients who received upfront plerixafor vs who received rescue plerixafor (D-VRd, 8.8×106 cells/kg vs 7.1×106 cells/kg; P=0.10; VRd, 10.5×106 cells/kg vs 9.4×106 cells/kg; P=0.20). See Table: Summary of ASCT and Stem Cell Mobilization in the GRIFFIN Study.
Stem Cell Remobilization
Transplantation and Hematologic Recovery
  • In total, 3 mobilized patients did not undergo ASCT (at the discretion of the principal investigator and due to AE sequelae and patient withdrawal).
  • In the D-VRd vs VRd arm, 94 vs 78 patients underwent ASCT with a median transplanted CD34+ cell dose of 4.2×106/kg vs 4.8×106/kg, respectively. See Table: Summary of ASCT and Stem Cell Mobilization in the GRIFFIN Study.
  • In the D-VRd vs VRd arm, neutrophil recovery was achieved in 100% of mobilized patients after ASCT at a median duration of 12 days (range, 3-31) vs 12 days (range, 2-23), respectively.
  • Platelet recovery in the D-VRd vs VRd arm occurred at a median duration of 13 vs 12 days, respectively.

Summary of ASCT and Stem Cell Mobilization in the GRIFFIN Study7
Parameter
All Patients
Received Upfront Plerixafor
Rescue Plerixafor Strategya
Received Rescue Plerixafor
Received G-CSF Only
D-VRdb
VRd
D-VRd
VRd
D-VRd
VRd
D-VRd
VRd
Patients who underwent mobilization attempts, n
95
80
49
31
19
13
19
28
Patients who received plerixafor, n
68
44
49
31
19
13
0
0
Mobilization cycle, n (%)
   1
89 (94)
74 (93)
46 (94)
28 (90)
19 (100)
12 (92)
17 (89)
27 (96)
   2
2 (2)
3 (4)
1 (2)
1 (3)
0
1 (8)
1 (5)
1 (4)
   3
0
1 (1)
0
0
0
0
0
0
   4
0
1 (1)
0
1 (3)
0
0
0
0
   Missing
4 (4)
1 (1)
2 (4)
1 (3)
0
0
1 (5)
0
Median stem cell collection target per ASCT, ×106 CD34+ cells/kg, (range)
2.5
(2-4)

2.5
(2-5)

2.5
(2-4)

2.5
(2-5)

2
(2-4)

2.5
(2-4)

2
(2-3)

2
(2-4)

Median duration of stem cell collection, (range), days
2
(1-4)

1
(1-4)

2
(1-4)

1
(1-4)

1
(1-3)

2
(1-4)

2
(1-4)

1
(1-4)

Median stem cell yield, ×106 CD34+ cells/kg, (range)
8.3
(2.6-33)

9.4
(4.1-28.7)

8.8
(2.6-33)

10.5
(5.5-22.5)

7.1
(4.2-16.7)

9.4
(4.4-17.3)

8.3
(4.4-18.6)

7.6
(4.1-23)

Patients who collected the minimum threshold for ASCT, n (%)
89 (94)
79 (99)
45 (92)
30 (97)
19 (100)
13 (100)
18 (95)
28 (100)
Patients who collected 2x the minimum threshold for ASCT, n (%)
81 (85)
74 (93)
42 (86)
29 (94)
16 (84)
11 (85)
17 (89)
26 (93)
Median number of CD34+ cells transplanted, ×106 cells/kg, (range)
4.2
(2-27.6)

4.8
(1.1-15)

4.3
(2.5-27.6)

4.8
(1.1-15)

4
(2-8.3)

4.7
(1.7-12.2)

4.3
(2.8-9.3)

4.4
(2-13.2)

Median number of days from the end of induction to apheresis, (range)
27
(0-63)

24
(4-133)

23
(14-48)

25.5
(4-133)

26
(0-36)

23
(14-43)

33
(13-63)

22.5
(12-55)

Patients completed ASCT, n (%)
94 (99)
78 (98)
49 (100)
31 (100)
18 (95)
13 (100)
19 (100)
26 (93)
Abbreviations: ASCT, autologous stem cell transplant; CD, cluster of differentiation; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; G-CSF, granulocyte colony-stimulating factor; VRd, bortezomib + lenalidomide + dexamethasone.
aPatients who did not receive upfront plerixafor had a treatment plan called “rescue plerixafor strategy.” Rescue plerixafor use was defined as using plerixafor only when deemed necessary “just-in-time” based on preapheresis blood CD34+ cell count after G-CSF. Patients in the rescue plerixafor strategy group did not receive upfront plerixafor but received either rescue plerixafor or no plerixafor at all.
bAmong the mobilized patients in GRIFFIN, 9 (D-VRd, n=5; VRd, n=4) received cyclophosphamide, and no accurate information on the mobilization regimen used was available for 16 patients (D-VRd, n=8; VRd, n=8).

DARZALEX FASPRO in Combination With Bortezomib, Lenalidomide, and Dexamethasone

PERSEUS (MMY3014; clinicaltrials.gov identifier: NCT03710603) is an ongoing, open-label, multicenter, phase 3 study evaluating the efficacy and safety of D-VRd vs VRd induction and consolidation followed by maintenance with D-R in D-VRd group or lenalidomide in VRd arm in patients with NDMM eligible for ASCT. Sonneveld et al (2023)8 reported efficacy and safety results from the PERSEUS study evaluating D-VRd vs VRd in patients with NDMM eligible for ASCT.

Study Design/Methods

  • A total of 709 patients were randomized 1:1 to into D-VRd (n=355) vs VRd (n=354) arm.
    • Stratification was done based on the International Staging System (ISS) disease stage (I, II, or III) and standard or high cytogenetic risk (defined as the absence or presence, respectively, of a del[17p], t[4;14], or t[14;16]).
  • Dosing:
    • Induction and consolidation: Total duration of induction and consolidation treatment was 6 cycles. Each cycle was 28 days.
      • D-VRd:
        • DARZALEX FASPRO- 1800 mg once weekly (QW) in cycles 1-2 and every 2 weeks (Q2W) in cycles 3-4
        • SC bortezomib-1.3 mg/m2 on days 1, 4, 8, and 11 of each cycle
        • Oral (PO) lenalidomide-25 mg on days 1-21 of each cycle
        • PO/IV dexamethasone-40 mg on days 1-4 and days 9-12 of each cycle
      • VRd:
        • SC bortezomib-1.3 mg/m2 on days 1, 4, 8, and 11 of each cycle
        • PO lenalidomide-25 mg daily on days 1-21 of each cycle
        • PO/IV dexamethasone-40 mg on days 1-4 and days 9-12 of each cycle
    • Maintenance: Total duration of maintenance (≥24 months) was cycle 7 until PD. Each cycle was 28 days.
      • D-VRd:
        • DARZALEX FASPRO-1800 mg SC every 4 weeks
        • PO lenalidomide-10 mg until PD or unacceptable toxicity
        • Patients who achieved sustained MRD for 12 months after ≥24 months of maintenance discontinued DARZALEX FASPRO, but continued PO lenalidomide until PD or unacceptable toxicity. Once they experienced loss of MRD-negativity or CR, they restarted DARZALEX FASPRO
        • Patients who did not achieve sustained MRD for 12 months after ≥24 months of maintenance continued DARZALEX FASPRO and PO lenalidomide
      • VRd:
        • PO lenalidomide-10 mg daily until PD or unacceptable toxicity
  • Primary endpoint: PFS.
  • Key secondary endpoints: Overall ≥CR, overall MRD-negativity, and OS.
  • Other secondary endpoints: ORR, ≥VGPR, sCR, duration of MRD-negativity.

Results

Patient Characteristics
  • A total of 709 patients were randomized into the D-VRd (n=355) and VRd (n=354) arms.
  • Demographics and baseline clinical characteristics of the ITT population are presented in Table: Demographics and Baseline Clinical Characteristics of the ITT Population.
  • A total of 698 patients (D-VRd, n=351; VRd, n=347) received ≥1 dose of treatment.
  • As of the clinical data cutoff date of August 1, 2023, 322 (91.7%) vs 300 (86.5%) patients in the D-VRd vs VRd arm, who started the induction phase continued into the maintenance phase, respectively.
    • A total of 207/322 patients in the D-VRd arm who were receiving maintenance treatment discontinued DARZALEX FASPRO per protocol after receiving ≥24 months of maintenance treatment and achieving ≥CR and sustained MRD-negativity for ≥12 months.
  • A total of 315 (89.7%) vs 302 (87%) patients in the D-VRd vs VRd arm received ASCT, respectively.
  • The median duration of treatment and median relative dose intensities are presented in Table: Duration of Treatment and Relative Dose Intensities During Induction/Consolidation/Maintenance Treatment in the Safety Population.
  • The median duration of follow-up was 47.5 months (range, 0-54.4).

Demographics and Baseline Clinical Characteristics of the ITT Populationa,8
D-VRd (n=355)
VRd (n=354)
Median age (range), years
61 (32-70)
59 (31-70)
Male, n (%)
211 (59.4)
205 (57.9)
Race, n (%)
   Asian
4 (1.1)
6 (1.7)
   Black or African American
5 (1.4)
4 (1.1)
   White
330 (93)
323 (91.2)
   Other
4 (1.1)
3 (0.8)
   Missing data
12 (3.4)
18 (5.1)
ECOG PSb, n (%)
   0
221 (62.3)
230 (65)
   1
114 (32.1)
108 (30.5)
   2
19 (5.4)
16 (4.5)
   3
1 (0.3)
0 (0)
Type of measurable disease, n (%)
   IgG
204 (57.5)
185 (52.3)
   IgA
65 (18.3)
85 (24)
   Otherc
13 (3.7)
11 (3.1)
   Detected in urine only
43 (12.1)
46 (13)
   Detected in serum free light chains only
29 (8.2)
27 (7.6)
   Not evaluable
1 (0.3)
0 (0)
ISS disease staged, n
355
353
   I, n (%)
186 (52.4)
178 (50.4)
   II, n (%)
114 (32.1)
125 (35.4)
   III, n (%)
55 (15.5)
50 (14.2)
Cytogenetic risk profilee, n (%)
   Standard risk
264 (74.4)
266 (75.1)
   High risk
76 (21.4)
78 (22)
   Indeterminate
15 (4.2)
10 (2.8)
Median time since diagnosis of multiple myeloma (range), months
1.2 (0-46.5)
1.1 (0.1-184.6)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; Ig, immunoglobulin; ISS, International Staging System; ITT, intention-to-treat; VRd, bortezomib + lenalidomide + dexamethasone.
aThe ITT population was defined as all patients who underwent randomization. Informal testing showed no significant differences between the 2 treatment groups in the characteristics evaluated at baseline.
bECOG PS is scored on a scale from 0 to 5, with 0 indicating no symptoms and higher scores indicating increasing disability. One patient had an ECOG PS score of 0 at the time of randomization that worsened to an ECOG PS score of 3 at baseline.
cIncludes IgD, IgM, IgE, and biclonal.
dThe ISS disease stage is based on the combination of serum β2-microglobulin and albumin levels. Higher stages indicate more advanced disease.
eCytogenetic risk was assessed by fluorescence in situ hybridization. High risk was defined as the presence of del(17p), t(4;14), and/or t(14;16).


Duration of Treatment and Relative Dose Intensities During Induction/Consolidation/Maintenance Treatment in the Safety Populationa,b,30
Median duration
of treatment (range), months
D-VRd (n=351)
VRd (n=347)
45.7 (0.5-54.3)
42.2 (0.1-53.9)
Median relative dose intensity (range), %
Induction
Consolidation
Maintenance
Induction
Consolidation
Maintenance
   Bortezomib
(n=351)
98
(25.3-104.8)

(n=243)
97.8
(12.3-114.2)

NA
(n=347)
97.8
(40.2-110.4)

(n=236)
98.2
(9.5-106)

NA
   Lenalidomide
(n=351)
100
(28.6-122.2)

(n=271)
100
(29.5-116.7)

(n=316)
85.2
(8.5-152.8)

(n=347)
100
(36.7-105.6)

(n=260)
100
(23.3-100)

(n=300)
97.1
(39.7-150.4)

   Dexamethasone
(n=351)
100
(20.8-183.3)

(n=263)
100
(1.6-100)

NA
(n=347)
100
(35.9-121.9)

(n=250)
100
(10-125)

NA
Induction Cycles 1-2
(n=351)

Induction Cycles 3-4
(n=343)
Consolidation
(n=274)

Maintenance
(n=322)

   DARZALEX  FASPRO
100
(50-100.4)

100
(25-100)

100
(50-100)

100
(67.6-100)

NA
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; NA, not available; VRd, bortezomib + lenalidomide + dexamethasone.
aDose intensity was defined as the ratio of the total administered dose to the total planned dose.
bThe safety population included all patients who received ≥1 dose of the study treatment.

Efficacy
  • A total of 50 (14.1%) vs 103 (29.1%) patients experienced disease progression or deaths in the D-VRd vs VRd arm of the ITT population (HR, 0.42; 95% CI, 0.30-0.59; P<0.0001), respectively, crossing the prespecified stopping boundary for superiority at the first interim analysis (P=0.0126).
  • The estimated 48-month PFS rate for the D-VRd vs VRd arm was 84.3% (95% CI, 79.5-88.1) vs 67.7% (95% CI, 62.2-72.6), respectively.
  • A summary of response rates and MRD status in the ITT population is presented in Table: Summary of Response Rates and MRD Status in the ITT Population.
  • Analyses of overall ≥CR rates and overall MRD-negativity rates (at 10-5) in prespecified subgroups appeared to favor D-VRd over VRd across clinically relevant subgroups.
  • Information on PFS in prespecified subgroup analysis is summarized in Table: PFS in Prespecified Subgroups.

Summary of Response Rates and MRD Status in the ITT Populationa,8
Parameter
D-VRd (n=355)
VRd (n=354)
P Valueb
Overall response
   n
343
332
-
   % (95% CI)
96.6 (94.2-98.2)
93.8 (90.7-96.1)
-
Response, n (%)
   ≥CR
312 (87.9)
248 (70.1)
<0.0001
      sCRc
246 (69.3)
158 (44.6)
-
      CR
66 (18.6)
90 (25.4)
-
   ≥VGPR
338 (95.2)
316 (89.3)
-
      VGPR
26 (7.3)
68 (19.2)
-
   PR
5 (1.4)
16 (4.5)
-
   SD
4 (1.1)
9 (2.5)
-
   PD
2 (0.6)
1 (0.3)
-
   Response could not be evaluated
6 (1.7)
12 (3.4)
-
MRD-negativityd, n (%)
   10-5 sensitivity
267 (75.2)
168 (47.5)
<0.0001
   10-6 sensitivity
231 (65.1)
114 (32.2)
-
Sustained MRD-negativity (10-5) for ≥12 months, n (%)
230 (64.8)
105 (29.7)
-
Abbreviations: CI, confidence interval; CR, complete response; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; FLC, free light chain; IMWG, International Myeloma Working Group; ITT, intention-to-treat; MRD, minimal residual disease; NGS, next-generation sequencing; PD, progressive disease; PR, partial response; sCR, stringent complete response; SD, stable disease; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone.
aResponse rates and MRD-negativity rates at any time during the study. The responses were assessed based on the IMWG response criteria.
bP values were calculated with the use of the stratified Cochran-Mantel-Haenszel chi-squared test.
cCriteria for an sCR included the criteria for a CR plus a normal serum FLC ratio and the absence of clonal plasma cells in the bone marrow, as assessed by immunohistochemistry, immunofluorescence, or 2- to 4-color flow cytometry.
dThe MRD-negativity rate was defined as the proportion of patients who achieved both MRD-negativity and ≥CR. Sustained MRD-negativity for 12 months was defined as 2 consecutive MRD-negative results 12 months apart, without any MRD-positive results in between. The MRD status was assessed using bone marrow samples and evaluated using an NGS assay (clonoSEQ assay, version 2; Adaptive Biotechnologies) in accordance with the IMWG guidelines for assessing MRD.


PFS in Prespecified Subgroups8
Subgroups
PFS
D-VRd
VRd
D-VRd
VRd
HR (95% CI)
No. of Progression Events or Deaths/Total no.
Median PFS, months
Sex
   Male
36/211
61/205
NE
NE
0.51 (0.34-0.77)
   Female
14/144
42/149
NE
NE
0.29 (0.16-0.53)
Age
   <65 years
30/261
84/267
NE
NE
0.30 (0.20-0.46)
   ≥65 years
20/94
19/87
NE
NE
0.97 (0.52-1.81)
Race
   White
47/330
95/323
NE
NE
0.42 (0.30-0.60)
   Other
3/25
8/31
NE
NE
0.40 (0.11-1.50)
ISS staging
   I
18/186
35/178
NE
NE
0.46 (0.26-0.81)
   II
19/114
43/125
NE
NE
0.37 (0.22-0.64)
   III
13/55
25/50
NE
41.9
0.42 (0.22-0.83)
Type of MM
   IgG
28/204
58/185
NE
NE
0.36 (0.23-0.57)
   Non-IgG
13/78
31/96
NE
NE
0.46 (0.24-0.88)
Cytogenetic risk
   Standard risk
25/264
62/266
NE
NE
0.35 (0.22-0.56)
   High risk
24/76
38/78
NE
44.1
0.59 (0.36-0.99)
   Intermediate risk
1/15
3/10
NE
NE
0.16 (0.02-1.56)
ECOG PS
   0
28/221
60/230
NE
NE
0.42 (0.27-0.66)
   ≥1
22/134
43/124
NE
NE
0.41 (0.25-0.69)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; HR, hazard ratio; Ig, immunoglobulin; ISS, International Staging System; MM, multiple myeloma; NE, not estimated; PFS, progression-free survival; VRd, bortezomib + lenalidomide + dexamethasone.
Safety
  • The most common grade 3/4 AEs recorded in the D-VRd vs VRd arm were neutropenia (62.1% vs 51%), thrombocytopenia (29.1% vs 17.3%), diarrhea (10.5% vs 7.8%), pneumonia (10.5% vs 6.1%), and febrile neutropenia (9.4% vs 10.1%). See Table: Most Common AEs During Treatment in the Safety Population.
  • Grade 3/4 peripheral neuropathy occurred in 6% vs 4.9% of patients in the D-VRd vs VRd arm, respectively.
  • Serious AEs (SAEs) in the safety population are summarized in Table: SAEs in the Safety Population.
  • A second primary malignancy was observed in 37 patients (10.5%) in the D-VRd arm and 25 patients (7.2%) in the VRd arm.
  • The number of deaths recorded due to COVID-19 in the D-VRd vs VRd arm was 4 (1.1%) vs 1 (0.3%) patients, respectively.
  • A total of 25.9% vs 54.2% of patients in the D-VRd vs VRd arm discontinued treatment, respectively.
    • The most common reasons for treatment discontinuation over all phases of the study were AEs (D-VRd, 9.1%; VRd, 22.5%) and PD (D-VRd, 8.3%; VRd, 20.7%).
  • A total of 34 vs 44 patients died in the D-VRd vs VRd arm, respectively.
    • A total of 7 patients died due to COVID-19 (D-VRd, n=4; VRd, n=3).

Most Common AEs During Treatment in the Safety Populationa,8
Event, n (%)
D-VRd (n=351)
VRd (n=347)
Any Grade
Grade 3/4
Any Grade
Grade 3/4
Any AE
349 (99.4)
321 (91.5)
344 (99.1)
297 (85.6)
Hematologic AEs
   Neutropenia
243 (69.2)
218 (62.1)
204 (58.8)
177 (51)
   Thrombocytopenia
170 (48.4)
102 (29.1)
119 (34.3)
60 (17.3)
   Anemia
78 (22.2)
21 (6)
72 (20.7)
22 (6.3)
   Febrile neutropenia
34 (9.7)
33 (9.4)
38 (11)
35 (10.1)
Nonhematologic AEs
   Diarrhea
214 (61)
37 (10.5)
188 (54.2)
27 (7.8)
   Peripheral sensory neuropathy
188 (53.6)
15 (4.3)
179 (51.6)
14 (4)
   Constipation
119 (33.9)
8 (2.3)
118 (34)
6 (1.7)
   Pyrexia
111 (31.6)
8 (2.3)
109 (31.4)
9 (2.6)
   Insomnia
95 (27.1)
8 (2.3)
61 (17.6)
6 (1.7)
   Asthenia
94 (26.8)
12 (3.4)
89 (25.6)
9 (2.6)
   Cough
85 (24.2)
1 (0.3)
51 (14.7)
0 (0)
   Fatigue
84 (23.9)
10 (2.8)
92 (26.5)
18 (5.2)
   Rash
82 (23.4)
9 (2.6)
94 (27.1)
17 (4.9)
   Back pain
80 (22.8)
2 (0.6)
66 (19)
1 (0.3)
   Peripheral edema
72 (20.5)
4 (1.1)
74 (21.3)
1 (0.3)
   Nausea
71 (20.2)
2 (0.6)
58 (16.7)
2 (0.6)
   Infections
305 (86.9)
124 (35.3)
266 (76.7)
95 (27.4)
      COVID-19
123 (35)
12 (3.4)
83 (23.9)
4 (1.2)
      Upper respiratory tract
      infection

111 (31.6)
2 (0.6)
87 (25.1)
6 (1.7)
      Pneumonia
64 (18.2)
37 (10.5)
38 (11)
21 (6.1)
Second primary malignancy
37 (10.5)
NA
25 (7.2)
NA
Any IRR
21 (6)
3 (0.9)
NA
NA
Abbreviations: AE, adverse event; COVID-19, coronavirus disease 2019; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; IRR, infusion-related reaction; NA, not applicable; VRd, bortezomib + lenalidomide + dexamethasone.
aThe safety population included patients who received ≥1 dose of the study treatment. AEs of any grade that were reported in ≥20% of patients in either treatment group and grade 3/4 AEs that were reported in ≥10% of patients in either treatment group are listed.


SAEs in the Safety Populationa,30
n (%)
D-VRd (n=351)
VRd (n=347)
Total no. of patients with SAEs
200 (57)
171 (49.3)
SAEs occurring in ≥2% of patients in either treatment group
   Infections
123 (35)
95 (27.4)
      Pneumonia
40 (11.4)
21 (6.1)
      COVID-19
13 (3.7)
6 (1.7)
      COVID-19 pneumonia
11 (3.1)
5 (1.4)
      Lower respiratory tract infection
9 (2.6)
3 (0.9)
      Sepsis
7 (2)
9 (2.6)
      Upper respiratory tract infection
7 (2)
8 (2.3)
   Febrile neutropenia
16 (4.6)
16 (4.6)
   Pyrexia
13 (3.7)
16 (4.6)
   Pulmonary embolism
9 (2.6)
5 (1.4)
   Atrial fibrillation
9 (2.6)
2 (0.6)
   Diarrhea
7 (2)
9 (2.6)
Abbreviations: COVID-19, coronavirus disease 2019; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; SAE, serious adverse event; VRd, bortezomib + lenalidomide + dexamethasone.
aThe safety population included patients who received ≥1 dose of the study treatment.

  • Median CD34+ cell yield in the D-VRd vs VRd arm was 5.5x106/kg vs 7.4x106/kg, respectively.
  • The proportion of patients proceeding to ASCT in the D-VRd vs VRd arm was 89.7% vs 87%, respectively.
  • The median time to complete hematopoietic reconstitution in the D-VRd vs VRd arm was 14 days each.

Stem Cell Yield and ASCT Results

Sonneveld et al (2024)9 presented (at the 21st IMS Annual Meeting) the stem cell yield and ASCT results for transplant-eligible patients with NDMM from the PERSEUS study who received D-VRd vs VRd induction prior to HDT or ASCT.

Results

Patient Disposition

  • At the time of the data cutoff (August 1, 2023), 709 patients were randomized to receive D-VRd (n=355) vs VRd (n=354) in the PERSEUS study.
  • Of the 698 patients who received D-VRd (n=351) vs VRd (n=347), 652 patients (D-VRd, n=335 vs VRd, n=317) underwent mobilization.
    • A total of 634 patients from the D-VRd (n=326) vs VRd (n=308) group completed mobilization.
    • A total of 603 patients from the D-VRd (n=309) vs VRd (n=294) group completed the transplant.
    • Maintenance therapy is ongoing for 260 vs 159 patients from the D-VRd vs VRd group, respectively.
      • After ≥24 months of maintenance therapy, 207 patients who achieved ≥CR and sustained MRD-negativity (≤10-5 sensitivity threshold) for 12 months discontinued DARZALEX FASPRO but continued with lenalidomide maintenance therapy until disease progression or unacceptable toxicity.

Safety

  • The stem cell mobilization and harvesting details are summarized in Table: Stem Cell Mobilization and Harvesting (Safety Analysis Set).
    • A total of 652 patients from the D-VRd (n=335) vs VRd (n=317) group underwent stem cell mobilization and harvesting.
    • The median time between administration of the last induction dose and first mobilization agent was 22 days.
    • Of the 640 patients who underwent stem cell collection (D-VRd, n=326 vs VRd, n=314), a high percentage of patients in both groups (D-VRd, n=320 [98.2%] vs VRd, n=312 [99.4%]) had sufficient stem cells collected for ASCT (≥2×106/kg cells).
    • Although the D-VRd vs VRd group had numerically lower median number of CD34+ stem cells collected (5.52×106/kg vs 7.44×106/kg), the percentage of patients who underwent ASCT was similar in both groups (89.7% vs 87%).
  • The SCT details are summarized in Table: Stem Cell Transplant (Safety Analysis Set).
    • After completion of 6 treatment cycles, 58 patients (29 in each treatment group) received ASCT, with 100% hematopoietic reconstitution and comparable engraftment times (D-VRd, 14 [1-67] days vs VRd, 14 [12-137] days) in both groups.

Stem Cell Mobilization and Harvesting (Safety Analysis Set)9
Characteristic
D-VRd
(n=335)
VRd
(n=317)
PBSC mobilizing agents useda, n (%)
   Cyclophosphamide
261 (77.9)
235 (74.1)
   G-CSFb
324 (96.7)
307 (96.8)
   Plerixafor
134 (40)
72 (22.7)
Patients with CD34+ stem cell collectiona (106/kg) among patients with stem cell collectionc,d, n
326
314
   Total CD34+ stem cells collected (106/kg), mean (SD)
6.236 (3.3243)
8.317 (5.0732)
   Total CD34+ stem cells collected (106/kg), median (range)
5.52 (1-26)
7.44 (0.74-49.50)
   Total CD34+ stem cells collected (≥2×106/kg), n (%)
320 (98.2)
312 (99.4)
   Total CD34+ stem cells collected (≥5×106/kg), n (%)
193 (59.2)
242 (77.1)
Abbreviations: CD, cluster of differentiation; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; G-CSF, granulocyte colony-stimulating factor; PBSC, peripheral blood stem cell; SD, standard deviation; VRd, bortezomib + lenalidomide + dexamethasone.aPercentages were calculated with the number of patients with available data in the related subgroup as the denominator.bIncluded standardized medications of filgrastim, lenograstim, and G-CSF.cPercentages were calculated with the number of patients in each group as the denominator.dThe stem cells were collected by PBSC apheresis or bone marrow harvest.

Stem Cell Transplant (Safety Analysis Set)9
Characteristic
D-VRd
(n=315)
VRd
(n=302)
Patients with CD34+ stem cells transplanted (106/kg)a, %
89.7
87
   CD34+ stem cells transplanted (106/kg), mean (SD)
4.07 (2.628)
4.87 (3.601)
   CD34+ stem cells transplanted (106/kg), median (range)
3.25 (0.8-26)
3.98 (1-38.7)
Transplanted patients with hematopoietic reconstitutionb, n (%)
314 (99.7)
300 (99.3)
Patients with ANC ≥0.5×109/L, n
314
300
Time to achieve ANC ≥0.5×109/L, mean (SD), days
13.5 (5.47)
13 (4.88)
Time to achieve ANC ≥0.5×109/L, median (range), days
13 (1-67)
13 (1-38)
Patients who achieved sustained platelets (≥20×109/L) without transfusionc, n
314
300
  Time to achieve sustained platelets without transfusion, mean  (SD), days
14.4 (7.79)
13 (9.44)
   Time to achieve sustained platelets without transfusion, median (range), days
14 (1-94)
12 (1-137)
Patients with engraftment post-ASCTc,d, n
314
300
   Time to engraftment, mean (SD), days
15.7 (7.68)
14.9 (9.37)
Time to engraftment, median (range), days
14 (1-94)
14 (1-137)
Abbreviations: ANC, absolute neutrophil count; ASCT, autologous stem cell transplant; CD, cluster of differentiation; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; SD, standard deviation; VRd, bortezomib + lenalidomide +dexamethasone.aPercentages were calculated with the number of patients in each group as the denominator.bPercentages were calculated with the number of patients with available data in the related subgroup as the denominator.cNumber of days from the transplant date, excluding patients whose counts did not reach nadir below the set threshold.dThe date of engraftment post-ASCT is defined as the latest date of ANC ≥0.5×109/L and platelet count ≥20×109/L. Patients with hematopoietic reconstitution are included.

Significance of CTCs as a Biomarker in Transplant-Eligible Patients with NDMM - Results From the PERSEUS Study

Bertamini et al (2024)10 presented (at the 66th ASH Annual Meeting) results from the PERSEUS study that highlighted the significance of CTCs as a biomarker in transplant-eligible patients with NDMM.

Results

Patient Characteristics


Baseline Characteristics in Patients from the PERSEUS Study - CTC Subgroup10
Characteristic
D-VRd
(n=231)
VRd
(n=220)
P Value
CTC detected, n/N (%)
183/231 (79.2)
187/220 (85)
0.73
CTC, median (IQR), %
0.0104 (0.0009-0.0738)
0.0088 (0.0012-0.0746)
0.88
Median age (IQR), years
60 (53.5-65)
59 (52.8-65)
0.71
Female sex, n (%)
84 (36.4)
95 (43.2)
0.14
ISS disease stage, %
   I
53.2
50.9
0.76
   II
31.6
35
0.76
   III
15.2
14.1
0.76
High LDH, n (%)
63 (27.3)
42 (19.1)
0.04
Cytogenetic high-riska, n (%)
51 (22.1)
49 (22.3)
0.86
Abbreviations: CTC, circulating tumor cells; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; IQR, interquartile range; ISS, International Staging System; LDH, lactate dehydrogenase; VRd, bortezomib + lenalidomide + dexamethasone.aHigh-risk cytogenetics was defined by the presence of t(4;14), and/or t(14;16) and/or del17p by fluorescence in situ hybridization.

Efficacy

  • D-VRd vs VRd significantly improved patient outcomes across both high and low CTC levels (P<0.0001).10
    • D-VRd vs VRd improved MRD-negativity rates (with ≥CR; 10–5 and 10–6 sensitivities) and sustained MRD-negativity rates (with ≥CR; 10-5 and 10–6 sensitivities; ≥12 months) in patients with both high and low CTC levels and the results are presented in Table: Summary of Overall MRD-Negativity (with ≥CR) Rates.
  • Increasing levels of CTC were associated with inferior prognosis. The 4-year PFS estimates based on CTC log10 values are presented below.10
    • Log10 value of <0.001%: 93%.
    • Log10 value of 0.001-0.01%: 79%.
    • Log10 value of 0.01-0.1%: 71%.
    • Log10 value of 0.1-1%: 67%
    • Log10 value of ≥1%: 48%.
  • D-VRd vs VRd improved PFS in patients with high cytogenetic risk and low CTC levels. However, there was no improvement in outcomes for patients with high cytogenetic risk combined with high CTC levels.10 The prognostic impact of CTC and other risk factors on PFS is presented in Table: Prognostic Impact of CTC and Other Risk Factors on PFS.
    • D-VRd vs VRd significantly improved PFS in patients with both high and low CTC levels (P<0.0001).10

Summary of Overall MRD-Negativity (with ≥CR) Rates10
Patients, %
CTC Low Levela
CTC High Levelb
D-VRd
(n=195)

VRd
(n=187)

P Value
D-VRd
(n=36)

VRd
(n=33)

P Value
Overall MRD-negativity (with ≥CR)c
   10-5 sensitivity
74
58
<0.001d
69
33
<0.01d
   10-6 sensitivity
66
39
<0.001d
47
21
<0.01d
Sustained MRD-negativity (with ≥CR; ≥12 months)e
   10-5 sensitivity
64
36
<0.0001f
50
15
<0.01f
   10-6 sensitivity
42
21
<0.0001f
39
6
<0.01f
Abbreviations: CR, complete response; CTC, circulating tumor cell; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ITT, intention-to-treat; MRD, minimal residual disease; VRd, bortezomib + lenalidomide + dexamethasone.
aCTC-low is defined by CTC <0.175%.
bCTC-high is defined by CTC ≥0.175%.
cProportion of patients who achieved both MRD-negativity and ≥CR in the randomized ITT population.dP value from a Chi-square test.eTwo consecutive MRD-negative results ≥12 months apart with no MRD-positive results in between.fP value from a Fisher’s test.


Prognostic Impact of CTC and Other Risk Factors on PFS10
Risk Factor
HR (95% CI)
P Value
CTC (log10)
1.36 (1.15-1.60)
≤0.05
High-risk cytogeneticsa
2.71 (1.76-4.17)
≤0.05
   ISS stage II
1.31 (0.81-2.12)
>0.05b
   ISS stage III
2.5 (1.45-4.32)
≤0.05
Elevated LDH
1.04 (0.65-1.68)
>0.05b
Abbreviations: CI, confidence interval; CTC, circulating tumor cells; HR, hazard ratio; ISS, International Staging System; LDH, lactate dehydrogenase; PFS, progression-free survival.
aComparison with standard risk. High-risk cytogenetics is defined by the presence of t(4;14) and/or t(14;16) and/or del17p by fluorescence in situ hybridization. Absence of those is considered standard risk.
bNot significant.

CEPHEUS (MMY3019; clinicaltrials.gov identifier: NCT03652064) is an ongoing, randomized, open-label, multicenter, phase 3 study evaluating the efficacy and safety of D-VRd vs VRd in patients with NDMM who are TIE or for whom transplant is not planned as initial therapy (transplant deferred).11-13 Usmani et al (2025)13 reported results from the phase 3 CEPHEUS study.

Study Design/Methods

  • The trial has enrolled 395 patients from 13 different countries including the US.12
  • Primary endpoint: Overall MRD-negativity (≥CR) rate at 10-5 sensitivity threshold.11,12
  • Key secondary endpoints: PFS, ORR, ≥VGPR, ≥CR, PFS2, OS, and sustained MRD-negativity (10-5) rate at ≥12 months.11,12

Results

Patient Characteristics


Baseline Demographics and Clinical Characteristics of the ITT Populationa,13
Characteristic
D-VRd (n=197)
VRd (n=198)
Median age (range), years
70 (42-79)
70 (31-80)
   <65 years, n (%)
36 (18.3)
35 (17.7)
   65 to <70 years, n (%)
52 (26.4)
53 (26.8)
   ≥70 years, n (%)
109 (55.3)
110 (55.6)
Age or transplant eligibility, n (%)
   <70 years and transplant ineligible
35 (17.8)
35 (17.7)
   <70 years and transplant deferred
53 (26.9)
53 (26.8)
   ≥70 years
109 (55.3)
110 (55.6)
Maleb, n (%)
87 (44.2)
111 (56.1)
Raceb, n (%)
   White
162 (82.2)
156 (78.8)
   Black or African American
10 (5.1)
9 (4.5)
   Asian
11 (5.6)
14 (7.1)
   Native Hawaiian or other Pacific Islander
0 (0)
1 (0.5)
   Other
1 (0.5)
2 (1)
   Not reported
13 (6.6)
16 (8.1)
ECOG PSc, n (%)
   0
71 (36)
84 (42.4)
   1
103 (52.3)
100 (50.5)
   2
23 (11.7)
14 (7.1)
Frailty scored, n (%)
   0 (fit)
124 (62.9)
132 (66.7)
   1 (intermediate fitness)
73 (37.1)
66 (33.3)
Type of measurable disease, n (%)
   Detected in serum only
120 (60.9)
108 (54.5)
      IgG
89 (45.2)
76 (38.4)
      IgA
27 (13.7)
31 (15.7)
      Othere
4 (2)
1 (0.5)
   Detected in serum and urine
41 (20.8)
45 (22.7)
   Detected in urine only
20 (10.2)
24 (12.1)
   Detected in serum FLCs only
16 (8.1)
21 (10.6)
ISS disease stagef, n (%)
   I
68 (34.5)
68 (34.3)
   II
73 (37.1)
75 (37.9)
   III
56 (28.4)
55 (27.8)
Cytogenetic risk profileg, n (%)
   Standard risk
149 (75.6)
149 (75.3)
   High risk
25 (12.7)
27 (13.6)
   Indeterminateh
23 (11.7)
22 (11.1)
Median time since diagnosis of MM (range), months
1.2 (0.4-5.8)
1.3 (0.3-8)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; FLC, free light chain; Ig, immunoglobulin; ISS, International Staging System; ITT, intention-to-treat; MM, multiple myeloma; VRd, bortezomib + lenalidomide + dexamethasone.
aThe ITT population was defined as all patients who underwent randomization.
bSex and race were reported by the patient.cECOG PS is scored on a scale of 0-5, with 0 indicating no symptoms and higher scores indicating increasing disability.
dTotal additive frailty is scored on a scale of 0-5 based on age, comorbidities, and cognitive and physical conditions, with 0 indicating fit, 1 indicating intermediate fitness, and ≥2 indicating frail, per the Myeloma Geriatric Assessment score (http://www.myelomafrailtyscorecalculator.net/).
eIncludes IgD, IgM, IgE, and biclonal.fBased on the combination of serum β2-microglobulin and albumin levels. Higher stages indicate more advanced disease.
gAssessed by fluorescence in situ hybridization; high risk was defined as the presence of del(17p), t(4;14), and/or t(14;16).
hIndeterminate includes patients with missing or unevaluable samples.

Efficacy

  • The duration of treatment and relative dose intensities are summarized in Table: Duration of Treatment and Relative Dose Intensities in the Safety Population.
  • As of the clinical cutoff date of May 7, 2024, 102 (51.8%) vs 67 (34.4%) patients from the D-VRd vs VRd arm, respectively, continued their treatment.13
  • A summary of the MRD status and response rates in the ITT population is presented in Table: Summary of MRD Status and Response Rates in the ITT Population.
    • D-VRd vs VRd significantly increased the overall MRD-negativity rate (10-5 sensitivity with ≥CR; 60.9% vs 39.4%) and ≥CR rate (81.2% vs 61.6%).13
    • Patients in the D-VRd vs VRd arm showed a higher MRD-negativity rate at 10-6 sensitivity (46.2% vs 27.3%) and a significantly higher sustained (≥12 months) MRD-negativity rate at 10-5 sensitivity (48.7% vs 26.3%).13
    • The treatment effect on the overall MRD-negativity rate remained consistent across the predefined subgroups and is summarized in Table: Prespecified Subgroup Analysis of Overall MRD-Negativity Rates.
  • At a median follow-up duration of 58.7 months, the median PFS was not reached in the D-VRd arm, while it was 52.6 months in the VRd arm.13
  • D-VRd significantly improved PFS with a 43% reduction in the risk of progression or death (HR, 0.57; 95% CI, 0.41-0.79; P=0.0005).13
  • At 54 months, the estimated PFS for D-VRd vs VRd was 68.1% (95% CI, 60.8-74.3) vs 49.5% (95% CI, 41.8-56.8).13
  • The treatment effect on PFS remained consistent across the predefined subgroups and is summarized in Table: PFS in Prespecified Subgroups.
  • Data for PFS on the subsequent line of therapy are still immature. However, HR favored D-VRd over VRd (HR, 0.78; 95% CI, 0.54-1.14) and further improved when censoring for death due to COVID-19 (HR, 0.60; 95% CI, 0.40-0.93).13
  • OS for ITT population favored the D-VRd vs VRd arm (HR, 0.85; 95% CI, 0.58-1.24) and further improved when censoring for death due to COVID-19 (HR, 0.69; 95% CI, 0.45-1.05).13
  • OS data were immature, and follow-up is ongoing.13
  • A lower proportion of patients who received subsequent therapy received an anti-CD38-based therapy in the D-VRd arm (3 of 22 patients [13.6%]) vs the VRd arm (39 of 65 patients [60%]).13
  • The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 global (EORTC QLQ-C30) health status domain score improved over time in both arms, with no negative impact from the addition of DARZALEX FASPRO.13

Duration of Treatment and Relative Dose Intensities in the Safety Populationa,b,13
Parameter
D-VRd (n=197)
VRd (n=195)
Median duration of treatment (range), months
56.3 (0.1-64.6)
34.3 (0.5-63.8)
Median number of treatment cycles (range)
59 (1-71)
(1-70)
Median relative dose intensity (range)
   Bortezomib
84.5 (12.7-104.3)
81.6 (22.4-102.1)
   Lenalidomide
80.6 (2.5-248.2)
83.8 (25.7-246)
   Dexamethasone
81.5 (19.6-177)
77.9 (23.4-173.4)
DARZALEX FASPRO
      Cycles 1 and 2 (n=197)
100 (33.3-105.6)
-
      Cycles 3 to 8 (n=191)
100 (33.3-101.1)
-
      Cycle 9+ (n=175)
100 (10-100.4)
-
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; VRd, bortezomib + lenalidomide + dexamethasone.
aDose intensity was defined as the ratio of total administered dose to total planned dose.bThe safety population included all patients who received ≥1 dose of study treatment.


Summary of MRD Status and Response Rates in the ITT Population13
Parameter
D-VRd (n=197)
VRd (n=198)
OR (95% CI)
P Value
Overall MRD-negativitya, %
   10-5 sensitivity
60.9
39.4
2.37 (1.58-3.55)
<0.0001
   10-6 sensitivity
46.2
27.3
2.24 (1.48-3.40)
0.0001
Sustained MRD-negativity
(10‒5) for ≥12 months, %

48.7
26.3
2.63 (1.73-4)
<0.0001
Responseb, n
191
184
-
-
   ORR, % (95% CI)
97 (93.5-98.9)
92.9 (88.4-96.1)
-
0.0698
      sCR, n (%)
128 (65)
88 (44.4)
-
<0.0001
      CR, n (%)
32 (16.2)
34 (17.2)
-
-
      VGPR, n (%)
23 (11.7)
50 (25.3)
-
-
      PR, n (%)
8 (4.1)
12 (6.1)
-
-
   ≥CR, n (%)
160 (81.2)
122 (61.6)
2.73 (1.71-4.34)
<0.0001
   ≥VGPR, n (%)
183 (92.9)
172 (86.9)
-
0.0495
   SD, n (%)
5 (2.5)
7 (3.5)
-
-
   PD, n (%)
0 (0)
0 (0)
-
-
   Response could not be evaluated, n (%)
1 (0.5)
7 (3.5)
-
-
Abbreviations: CI, confidence interval; CR, complete response; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; IMWG, International Myeloma Working Group; ITT, intention-to-treat; MRD, minimal residual disease; OR, odds ratio; ORR, overall response rate; PD, progressive disease; PR, partial response; sCR, stringent complete response; SD, stable disease; VGPR, very good partial response; VRd, bortezomib + lenalidomide + dexamethasone.
aMRD-negativity rate was defined as the proportion of patients who achieved both MRD-negativity (10-5 threshold) and ≥CR.
bResponse rates at any time during the study. Response was assessed based on IMWG response criteria. P values were calculated using the stratified Cochran-Mantel-Haenszel chi-squared test.


Prespecified Subgroup Analysis of Overall MRD-Negativity Rates13
Subgroup
D-VRd
VRd
OR (95% CI)
Number of Patients With MRD-Negativity/
Total Number of Patients (%)

Sex
   Male
54/87 (62.1)
39/111 (35.1)
3.02 (1.69-5.41)
   Female
66/110 (60)
39/87 (44.8)
1.85 (1.04-3.26)
Age
   <70 years
59/88 (67)
36/88 (40.9)
2.94 (1.59-5.44)
   ≥70 years
61/109 (56)
42/110 (38.2)
2.06 (1.20-3.53)
Region
   Europe
69/120 (57.5)
46/116 (39.7)
2.06 (1.23-3.46)
   North America
21/37 (56.8)
13/31 (41.9)
1.82 (0.69-4.77)
   Other
30/40 (75)
19/51 (37.3)
5.05 (2.03-12.60)
Weight
   ≤65 kg
40/58 (69)
22/63 (34.9)
4.14 (1.94-8.86)
   >65-85 kg
58/101 (57.4)
31/88 (35.2)
2.48 (1.38-4.47)
   >85 kg
22/38 (57.9)
25/47 (53.2)
1.21 (0.51-2.87)
ISS staging
   I
45/68 (66.2)
30/68 (44.1)
2.48 (1.24-4.96)
   II
47/73 (64.4)
29/75 (38.7)
2.87 (1.47-5.59)
   III
28/56 (50)
19/55 (34.5)
1.89 (0.88-4.07)
Cytogenetic risk
   High risk
12/25 (48)
15/27 (55.6)
0.74 (0.25-2.20)
   Standard risk
95/149 (63.8)
57/149 (38.3)
2.84 (1.78-4.54)
   Indeterminate
13/23 (56.5)
6/22 (27.3)
3.47 (0.99-12.09)
ECOG PS score
   0
41/71 (57.7)
36/84 (42.9)
1.82 (0.96-3.45)
   ≥1
79/126 (62.7)
42/114 (36.8)
2.88 (1.71-4.87)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; MRD, minimal residual disease; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.

PFS in Prespecified Subgroups13
Subgroups
D-VRd
VRd
D-VRd
VRd
HR (95% CI)
Number of Disease Progression Events or Deaths/Total Number of Patients
Median PFS, months
Sex
   Male
24/87
53/111
NE
49.2
0.46 (0.29-0.75)
   Female
39/110
38/87
NE
NE
0.73 (0.47-1.15)
Age
   <70 years
30/88
38/88
NE
NE
0.72 (0.44-1.16)
   ≥70 years
33/109
53/110
NE
49.4
0.50 (0.33-0.78)
Region
   Europe
37/120
54/116
NE
51.1
0.54 (0.36-0.82)
   North America
10/37
13/31
NE
50.2
0.51 (0.22-1.17)
   Other
16/40
24/51
NE
NE
0.87 (0.46-1.64)
Weight
   ≤65 kg
17/58
24/63
NE
NE
0.62 (0.34-1.16)
   >65-85 kg
34/101
40/88
NE
51.1
0.65 (0.41-1.02)
   >85 kg
12/38
27/47
NE
41.9
0.46 (0.23-0.91)
ISS staging
   I
21/68
28/68
NE
60.6
0.66 (0.37-1.16)
   II
18/73
37/75
NE
45.6
0.36 (0.21-0.64)
   III
24/56
26/55
NE
49.2
0.84 (0.48-1.46)
Cytogenetic risk
   High risk
13/25
17/27
39.8
31.7
0.88 (0.42-1.84)
   Standard risk
43/149
60/149
NE
60.6
0.61 (0.41-0.91)
   Indeterminate
7/22
14/22
NE
47.9
0.33 (0.13-0.82)
ECOG PS score
   0
16/71
30/84
NE
NE
0.53 (0.29-0.97)
   ≥1
47/126
61/114
NE
43.8
0.59 (0.40-0.86)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; HR, hazard ratio; ISS, International Staging System; NE, not estimated; PFS, progression-free survival; VRd, bortezomib + lenalidomide + dexamethasone.

Safety

  • A total of 95 vs 128 patients from the D-VRd vs VRd arm, respectively, discontinued treatment.13
    • Reasons for treatment discontinuation in the D-VRd vs VRd arm, respectively, included:
      • Progressive disease: 27 (13.7%) vs 51 (26.2%) patients.
      • Adverse events (AEs): 16 vs 32 patients.
      • Deaths: 34 vs 24 patients.
        • COVID-19-related deaths: 12 (6.1%) vs 6 (3.1%) patients.
      • Refused further treatment: 15 vs 8 patients.
      • Physician decision: 3 vs 12 patients.
      • Received concurrent treatment for MM prior to disease progression: 0 vs 1 patient.
    • Treatment discontinuation due to TEAEs was observed in 7.6% vs 15.9% of patients from the D-VRd vs VRd arm, respectively.
  • The safety data were consistent with the established safety profile of each individual drug.13 TEAEs in the safety population are summarized in Table: Most Common TEAEs in the Safety Population.
    • Overall, deaths were reported in 51 vs 60 patients from the D-VRd vs VRd arm, respectively and are summarized in Table: Summary of Deaths in the ITT Population.
      • The COVID-19 pandemic impacted OS with 24 (21.6%) of all deaths in the study attributed to COVID-19 (D-VRd, n=15 vs VRd, n=9).
      • Overall, 21 COVID-19 deaths occurred during the peak of the pandemic in 2020 and 2021, with 3 additional deaths in 2022 (post-vaccine availability), and none in 2023 or 2024.
  • Regional variations were observed in pandemic impact. Brazil reported 54.2% of COVID-19 deaths (17.5% of the study population), while Poland had 16.7% of deaths (18.7% of the study population).
  • Two sensitivity analyses of OS adjusting for COVID-19 deaths showed a stronger treatment effect for D-VRd vs VRd (censoring COVID-19 deaths [HR, 0.69; 95% CI, 0.45-1.05] and considering COVID-19 deaths as a competing risk [HR for non-COVID mortality, 0.67; 95% CI, 0.44-1.03]).
    • Grade 5 non-COVID-related TEAEs occurred in 10.7% vs 7.7% of patients from the D-VRd vs VRd arm, respectively.
    • Serious TEAEs occurred in 142 (72.1%) vs 131 (67.2%) of patients from the D-VRd vs VRd arm, respectively, with the most common serious TEAE being pneumonia (D-VRd, 13.7%; VRd, 12.8%). A summary of the serious TEAEs is presented in Table: Serious TEAEs (≥2%) in the Safety Population.
    • Most grade 5 TEAEs occurred after the discontinuation of bortezomib (cycle 8) in both arms (D-VRd, 13%; VRd, 9%). When adjusted for treatment exposure, the rate of grade 5 TEAEs was similar between the two arms (D-VRd, 0.39 per 100 patient-months; VRd, 0.31 per 100 patient-months).

Most Common TEAEs in the Safety Populationa,13
TEAE, n (%)
D-VRd (n=197)
VRd (n=195)
Any Grade
Grade 3/4
Any Grade
Grade 3/4
Hematologic
   Neutropenia
110 (55.8)
87 (44.2)
76 (39)
58 (29.7)
   Thrombocytopenia
92 (46.7)
56 (28.4)
66 (33.8)
39 (20)
   Anemia
73 (37.1)
26 (13.2)
62 (31.8)
23 (11.8)
   Lymphopenia
36 (18.3)
24 (12.2)
34 (17.4)
20 (10.3)
Nonhematologic
   Diarrhea
112 (56.9)
24 (12.2)
115 (59)
18 (9.2)
   Peripheral sensory neuropathy
110 (55.8)
16 (8.1)
119 (61)
16 (8.2)
   Peripheral edema
83 (42.1)
4 (2)
76 (39)
1 (0.5)
   Constipation
75 (38.1)
4 (2)
82 (42.1)
5 (2.6)
   Insomnia
63 (32)
4 (2)
63 (32.3)
2 (1)
   Fatigue
63 (32)
18 (9.1)
60 (30.8)
16 (8.2)
   Hypokalemia
58 (29.4)
24 (12.2)
25 (12.8)
12 (6.2)
   Cataract
55 (27.9)
17 (8.6)
51 (26.2)
17 (8.7)
   Back pain
55 (27.9)
6 (3)
43 (22.1)
6 (3.1)
   Cough
53 (26.9)
1 (0.5)
38 (19.5)
2 (1)
   Asthenia
51 (25.9)
7 (3.6)
40 (20.5)
5 (2.6)
   Rash
50 (25.4)
5 (2.5)
48 (24.6)
3 (1.5)
   Nausea
49 (24.9)
0 (0)
48 (24.6)
4 (2.1)
   Pyrexia
46 (23.4)
2 (1)
30 (15.4)
1 (0.5)
   Arthralgia
45 (22.8)
3 (1.5)
39 (20)
0 (0)
   Decreased appetite
42 (21.3)
2 (1)
39 (20)
5 (2.6)
   Dizziness
41 (20.8)
1 (0.5)
41 (21)
2 (1)
   Infections
181 (91.9)
79 (40.1)
167 (85.6)
62 (31.8)
      Upper respiratory tract infection
78 (39.6)
1 (0.5)
64 (32.8)
1 (0.5)
      COVID-19
75 (38.1)
22 (11.2)
48 (24.6)
9 (4.6)
      Pneumonia
48 (24.4)
28 (14.2)
39 (20)
25 (12.8)
      Urinary tract
41 (20.8)
7 (3.6)
29 (14.9)
5 (2.6)
Second primary malignancyb
15 (7.6)
-
18 (9.2)
-
Any injection-related reaction
7 (3.6)
1 (0.5)c
-
-
Abbreviations: AE, adverse event; COVID-19, coronavirus disease 2019; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; TEAE, treatment-emergent adverse event; VRd, bortezomib + lenalidomide + dexamethasone.
aThe safety population included patients who received ≥1 dose of study treatment. AEs of any grade that were reported in ≥20% of patients in either treatment arm and grade 3/4 AEs that were reported in ≥10% of patients in either treatment arm are listed.
bOf all second primary malignancies, cutaneous malignancies were reported in 7 (3.6%) vs 7 (3.6%) patients from the D-VRd vs VRd arm, respectively.cGrade 3.


Summary of Deaths in the ITT Populationa,13
Parameter, n
D-VRd (n=197)
VRd (n=195)
Overall deaths
51
60
   PD
8
16
   AEb
37
25
      COVID-19
7
5
      COVID-19 pneumonia
5
1
      Pneumonia
3
4
      Death/sudden death
3
1
      Cardiac arrest
2
1
      General physical health deterioration
2
-
      Drug induced liver injury
1
-
      COVID-19, multiple organ dysfunction syndrome, and pulmonary embolism
1
-
      Colitis
1
-
      Sudden cardiac death
1
-
      Respiratory failure
1
-
      Acute kidney injury/failure
1
1
      Dyspnea
1
-
      Pulmonary embolism
1
-
      Pulmonary fibrosis
-
1
      Myocardial infarction
1
1
      Acute myocardial infarction, cardiogenic shock
-
1
      Multiple organ dysfunction syndrome
-
1
      Lung neoplasm malignant
-
1
      Completed suicide
-
1
      Hypovolemic shock
-
1
      Septic shock
1
1
      Sepsis
-
2
      Urinary tract infection
-
1
      Cerebrovascular accident
1
-
      Cardiopulmonary failure
1
-
      Hepatic failure
-
1
      Febrile neutropenia
1
-
      Abdominal pain
1
-
      Esophageal adenocarcinoma
1
-
   Otherb
6
19
      Unknown
3
10
      COVID-19 or COVID-19 positive/infection
1
2
      Acute hypoxic respiratory failure due to COVID-19
1
-
      COVID-19 bronchopneumonia bilat.
-
1
      Severe acute hepatitis
1
-
      No more information available
-
1
      Pneumocystosis infection
-
1
      Ischemic bowel stroke
-
1
      Acute kidney injuryc
-
1
      Cholengiocellular carcinoma intrahepatic metastasis
-
1
      Renal failure, possibility of PD
-
1
Abbreviations: AE, adverse event; COVID-19, coronavirus disease 2019; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; PD, progressive disease; VRd, bortezomib + lenalidomide + dexamethasone.
aUntil the clinical cutoff. bSubcategory terms are listed as originally entered in the database by the investigator.cPatient died following admission and was outside AE reporting window.


Serious TEAEs (≥2%) in the Safety Populationa,13
TEAE, n (%)
D-VRd (n=197)
VRd (n=195)
Serious TEAEs
142 (72.1)
131 (67.2)
   Infections
78 (39.6)
69 (35.4)
      Pneumonia
27 (13.7)
25 (12.8)
      COVID-19
22 (11.2)
16 (8.2)
      COVID-19 pneumonia
8 (4.1)
4 (2.1)
      Sepsis
7 (3.6)
4 (2.1)
      Urinary tract infection
7 (3.6)
4 (2.1)
      Septic shock
6 (3 )
1 (0.5)
      Gastroenteritis
4 (2)
4 (2.1)
      Influenza
4 (2)
1 (0.5)
   Pulmonary embolism
11 (5.6)
5 (2.6)
   Diarrhea
10 (5.1)
6 (3.1)
   Atrial fibrillation
7 (3.6)
7 (3.6)
   Acute kidney injury
6 (3)
3 (1.5)
   Asthenia
6 (3)
2 (1)
   Anemia
6 (3)
2 (1)
   Cataract
5 (2.5)
4 (2.1)
   Pvrexia
5 (2.5)
3 (1.5)
   Hypokalemia
5 (2.5)
3 (1.5)
   Hyponatremia
5 (2.5)
1 (0.5)
   Febrile neutropenia
4 (2)
4 (2.1)
   Thrombocytopenia
4 (2)
2 (1)
   Deep vein thrombosis
4 (2)
2 (1)
   Syncope
3 (1.5)
6 (3.1)
   Hypotension
3 (1.5)
4 (2.1)
   Orthostatic hypotension
2 (1)
5 (2.6)
   Dehydration
0 (0)
5 (2.6)
Abbreviations: COVID-19, coronavirus disease 2019; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; TEAE, treatment-emergent adverse event; VRd, bortezomib + lenalidomide + dexamethasone.
aThe safety population included patients who received ≥1 dose of study treatment.

Expanded Analysis of MRD Outcomes - Results From the CEPHEUS Study

Zweegman et al (2024)14 presented (at the 66th ASH Annual Meeting) an expanded analysis of MRD outcomes from the CEPHEUS study in patients with NDMM who were ineligible for SCT or for whom transplant was deferred.

Results

Patient Characteristics

  • A total of 395 patients were randomized into the D-VRd (n=197) and VRd (n=198) arms.14

Efficacy

  • A summary of the cumulative and sustained MRD-negativity (≥CR) rates is presented in Table: Summary of Cumulative and Sustained MRD-Negativity (≥CR) Rates - CEPHEUS Expanded Analysis.
    • D-VRd improved cumulative MRD-negativity rates (both 10-5 and 10-6 sensitivities) vs VRd alone at all prespecified timepoints.14
    • D-VRd almost doubled sustained MRD-negativity (≥) CR rates at 12, 24, and 36 months.14
    • Among patients who achieved both MRD-negativity (10-6 sensitivity) and ≥CR with D-VRd, >85% were alive and progression free at 54 months.14
  • D-VRd vs VRd showed PFS benefit regardless of an MRD-negativity status (10-6 sensitivity).14
    • The overall MRD-negativity rate (10-6 sensitivity) for D-VRd vs VRd was 46.2% vs 27.3%, respectively and the overall MRD-positivity rate (10-6 sensitivity) was 53.8% vs 72.7%, respectively.
    • At 54 months, the estimated PFS by MRD-negativity status (10-6 sensitivity) was 86.2% vs 79% and by MRD-positivity status was 51% vs 36.5% for D-VRd vs VRd, respectively.
  • Analyses of MRD-negativity (with ≥CR) rates in prespecified subgroups appeared to consistently favor D-VRd over VRd across these prespecified subgroups and are summarized in Table: MRD-Negativity (≥CR) Rates in Prespecified Subgroups.

Summary of Cumulative and Sustained MRD-Negativity (≥CR) Rates - CEPHEUS Expanded Analysis14
Parameter
D-VRd
(n=197)

VRd
(n=198)

OR (95% CI)
P Value
Cumulative MRD-negativity (10-5 sensitivity; ≥CR), %
   12 months
43.1
28.3
-
-
   24 months
56.9
35.9
-
-
   36 months
59.9
37.4
-
-
   48 months
60.9
38.4
-
-
Cumulative MRD-negativity (10-6 sensitivity; ≥CR), %
   12 months
22.8
11.1
-
-
   24 months
38.1
22.2
-
-
   36 months
40.6
25.3
-
-
   48 months
45.2
27.3
-
-
Sustained MRD-negativity (10-5 sensitivity; ≥CR)a, %
≥12 monthsb
49.2
27.3
2.56 (NR)
<0.0001
   ≥24 monthsc
42.1
22.7
2.47 (NR)
<0.0001
   ≥36 monthsd
29.9
15.2
2.37 (NR)
0.0005
Sustained MRD-negativity (10-6 sensitivity; ≥CR)a, %
≥12 monthsb
34
16.2
NR
NR
   ≥24 monthsc
27.9
13.6
NR
NR
   ≥36 monthsd
18.8
8.6
NR
NR
Abbreviations: CI, confidence interval; CR, complete response; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; MRD, minimal residual disease; NR, not reported; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.
aAt any time during the study.
bProportion of patients who achieved ≥CR and achieved an MRD-negative status at 2 bone marrow assessments that are 12 month apart with an allotted window of ±1 month, without an MRD-positive status in between.
cAchieving an MRD-negative status at 2 bone marrow assessments that are 24 months apart with an allotted window of ±3 months, without an MRD-positive status in between.
dAchieving an MRD-negative status at 2 bone marrow assessments that are 36 months apart with an allotted window of ±3 months, without an MRD-positive status in between.


MRD-Negativity (≥CR) Rates in Prespecified Subgroups14
Subgroups,
n/N (%)

D-VRd
VRd
OR
(95% CI)

D-VRd
VRd
OR
(95% CI)

10-5 Sensitivity
10-6 Sensitivity
Sex
   Male
54/87
(62.1)

39/111 (35.1)
3.02
(1.69-5.41)

42/87 (48.3)
28/111 (25.2)
2.77
(1.52-5.04)

   Female
66/110 (60)
39/87
(44.8)

1.85
(1.04-3.26)

49/110 (44.5)
26/87 (29.9)
1.88
(1.04-3.41)

Age
   <70 years
59/88
(67)

36/88
(40.9)

2.94
(1.59-5.44)

44/88 (50)
25/88 (28.4)
2.52
(1.35-4.70)

   ≥70 years
61/109 (56)
42/110
(38.2)

2.06
(1.20-3.53)

47/109 (43.1)
29/110 (26.4)
2.12
(1.20-3.74)

Region
   Europe
69/120 (57.5)
46/116
(39.7)

2.06
(1.23-3.46)

57/120 (47.5)
34/116 (29.3)
2.18
(1.28-3.73)

   North America
21/37
(56.8)

13/31
(41.9)

1.82
(0.69-4.77)

14/37 (37.8)
9/31
(29)

1.49
(0.54-4.13)

   Other
30/40
(75)

19/51
(37.3)

5.05
(2.03-12.60)

20/40 (50)
11/51 (21.6)
3.64
(1.46-9.04)

Weight
   ≤65 kg
40/58
(69)

22/63
(34.9)

4.14
(1.94-8.86)

31/58 (53.4)
18/63 (28.6)
2.87
(1.35-6.09)

   >65-85 kg
58/101
(57.4)

31/88
(35.2)

2.48
(1.38-4.47)

45/101 (44.6)
19/88 (21.6)
2.92
(1.54-5.54)

   >85 kg
22/38
(57.9)

25/47
(53.2)

1.21
(0.51-2.87)

15/38 (39.5)
17/47 (36.2)
1.15
(0.48-2.78)

ISS staging
   I
45/68
(66.2)

30/68
(44.1)

2.48
(1.24-4.96)

32/68 (47.1)
22/68 (32.4)
1.86
(0.93-3.73)

   II
47/73
(64.4)

29/75
(38.7)

2.87
(1.47-5.59)

37/73 (50.7)
17/75 (22.7)
3.51
(1.73-7.13)

   III
28/56
(50)

19/55
(34.5)

1.89
(0.88-4.07)

22/56 (39.3)
15/55 (27.3)
1.73
(0.78-3.84)

Cytogenetic risk
   High risk
12/25
(48)

15/27
(55.6)

0.74
(0.25-2.20)

8/25
(32)

12/27 (44.4)
0.59
(0.19-1.83)

   Standard risk
95/149
(63.8)

57/149
(38.3)

2.84
(1.78-4.54)

71/149 (47.7)
37/149 (24.8)
2.76
(1.69-4.50)

ECOG PS score
   0
41/71
(57.7)

36/84
(42.9)

1.82
(0.96-3.45)

28/71 (39.4)
27/84 (32.1)
1.37
(0.71-2.66)

   ≥1
79/126
(62.7)

42/114
(36.8)

2.88
(1.71-4.87)

63/126 (50)
27/114 (23.7)
3.22
(1.85-5.61)

Abbreviations: CI, confidence interval; CR, complete response; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; MRD, minimal residual disease; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.

DARZALEX FASPRO Phase 2 Study

PLEIADES (MMY2040; clinicaltrials.gov identifier: NCT03412565) is an ongoing, phase 2, non-randomized, open-label, multicenter study evaluating the clinical benefit of DARZALEX FASPRO administered in combination with various treatment regimens in patients with MM. Chari et al (2021)18 presented updated safety and efficacy results of the D-VRd, DARZALEX FASPRO in combination with bortezomib, melphalan, and prednisone (D-VMP), and DARZALEX FASPRO in combination with lenalidomide and dexamethasone (D-Rd) arms in the PLEIADES study.15-18 Results specifically to the D-VRd cohort are summarized below.

Study Design/Methods

  • Key inclusion criteria:
    • Age ≥18 years old; confirmed diagnosis of MM by the International Myeloma Working Group criteria; measurable, secretory disease as defined by any of the following: (serum monoclonal paraprotein [M-protein] level ≥1 g/dL; or urine M-protein level ≥200 mg/24 hours; or light chain MM for patients without measurable disease in the serum or urine: serum Ig free light chain [FLC] ≥10 mg/dL and abnormal FLC ratio).
    • Eastern Cooperative Oncology Group performance status (ECOG PS) ≤2
  • Transplant-eligible NDMM: D-VRd (21-day cycles x 4 induction cycles)
    • DARZALEX FASPRO injection: 1800 mg of DARZALEX FASPRO weekly in cycles 1-3 and day 1 of cycle 4
    • Bortezomib: 1.3 mg/m2 SC injection on days 1, 4, 8, 11 of cycles 1-4
    • Lenalidomide: 25 mg PO on days 1-14 of cycles 1-4
    • Dexamethasone: 20 mg PO/IV on days 1, 2, 8, 9, 15, 16 of cycles 1-4
  • Primary endpoints: ORR for the D-VMP and D-Rd cohorts; ≥VGPR after the 4 induction cycles for the D-VRd cohort
  • Key secondary endpoints: maximum observed serum concentrations (Cmax) and minimum observed serum concentrations (Cmin) of daratumumab, immunogenicity, percentage of participants with IRR, rate of ≥CR, ORR for the D-VRd cohort

Results: D-VRd


Baseline Demographics and Patient Characteristicsa,18
Transplant-eligible NDMM
D-VRd (n=67)
Age, years
   Median (range)
59 (33-76)
   18 to <65, n (%)
54 (80.6)
   65 to <75, n (%)
12 (17.9)
   ≥75, n (%)
1 (1.5)
Male, n (%)
48 (71.6)
Median (range) body weight, kg
77 (43-148)
Race, n (%)
   White
38 (56.7)
   Black or African American
5 (7.5)
   Asian
0 (0)
ECOG PS score, n (%)
   0
40 (59.7)
   1
26 (38.8)
   2
1 (1.5)
Median (range) number of prior lines of therapy, n
N/A
ISS stagingb,n (%)
   I
30 (44.8)
   II
23 (34.3)
   III
14 (20.9)
Cytogenetic riskc,d
   N
53
   Standard risk, n (%)
40 (75.5)
   High risk, n (%)
13 (24.5)
      t(4;14)
9 (17)
      t(14;16)
1 (1.9)
      del17p
5 (9.4)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; NDMM, newly diagnosed multiple myeloma.
aAll-treated population, defined as patients who received ≥1 dose of study treatment.
bBased on the combination of serum β2-microglobulin and albumin at screening.
cBased on fluorescence in situ hybridization or karyotyping testing conducted locally.
dHigh cytogenetic risk was defined as having ≥1 of t(4;14), t(14;16) or del17p abnormalities.


Patient Disposition and Exposurea,18
Transplant-eligible NDMM
D-VRd (n=67)
Median treatment cycles (range), n
4 (1-4)
Median duration of treatment (range), months
2.6 (0-4)
Relative dose intensity, median %
   DARZALEX FASPRO
100
   Bortezomib
97.9
   Melphalan
-
   Prednisone
-
   Lenalidomide
100
   Dexamethasone
100
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; NDMM, newly diagnosed multiple myeloma.
aThe all-treatedpopulation included all patients who received ≥1 dose of study treatment.

Efficacy
  • Median follow-up was 3.9 months for D-VRd cohort.
  • Primary endpoints were met for the D-VRd cohort with available data and response rates were similar to DARZALEX studies in GRIFFIN2.
  • In the D-VRd cohort (n=67):
    • ≥VGPR was 71.6% vs 71.7% in the GRIFFIN study.2
    • ORR was 97% vs 98%
    • PR was 25.4% vs 26.3%
    • CR was 7.5% vs 12.1%
    • sCR was 9% vs 7.1%
Pharmacokinetics  
  • Overall, trough level or trough concentration (Ctrough) and immunogenicity values were consistent with historical DARZALEX data.
  • Cmax after the 1st dose was D-VRd: 100 ± 48.5 µg/mL.
  • Sixteen patients developed rHuPh20 antibodies (all were non-neutralizing).
  • No patients developed anti-daratumumab antibodies.
Safety
  • Treatment discontinuation due to TEAEs were 2% in the D-VRd arm. A summary of TEAEs reported in each cohort is presented in Table: Safety Summary.
  • Any-grade IRRs occurred in 7.5% of patient across all cohorts.  

Safety Summary18
Transplant-eligible NDMM
D-VRd (n=67)
Any TEAE, n (%)
67 (100)
Serious
19 (28.4)
Grade 3/4
39 (58.2)
Grade 5
1 (1.5)
TEAEs leading to treatment discontinuation
1 (1.5)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; NDMM, newly diagnosed multiple myeloma; TEAE, treatment emergent adverse event.
  • Across all 3 cohorts with available data any grade IRRs occurred in 7.5% (15/199) of patients. IRRs were mild (grade 1/2), one patient had grade 3 IRR and no patients reported grade 4 IRR.
  • Median time to onset of IRRs was 4.4 hours in the D-VRd cohort.
  • Across all 3 cohorts with available data local injection site reactions occurred in 7.5% (15/199) of patient (all grade 1/2).
  • A summary of most common TEAEs is presented in Table: Most Common TEAEs (≥5% in D-VRd cohort).

Most common TEAEs (≥5% in D-VRd cohort)a,18
Event, n (%)
Transplant-eligible NDMM
D-VRd (n=67)
Neutropenia
19 (28.4)
Lymphopenia
11 (16.4)
Thrombocytopenia
10 (14.9)
Leukopenia
5 (7.5)
Anemia
3 (4.5)
Pneumonia
2 (3)
Hypertension
1 (1.5)
Hyperglycemia
1 (1.5)
Hypokalemia
0 (0)
Any-Grade IRR
6 (9)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; IRR, infusion-related reaction; NDMM, newly diagnosed multiple myeloma; TEAE, treatment-emergent adverse event. aThe all-treatedpopulation included all patients who received ≥1 dose of study treatment.

LITERATURE SEARCH

A literature search of MEDLINE®, Embase®, BIOSIS Previews®, and Derwent Drug File databases (and/or other resources, including internal/external databases) was conducted on 10 February 2025.

References

1 Voorhees P, Costa L, Reeves B, et al. Interim safety analysis of a phase 2 randomized study of daratumumab (Dara), lenalidomide (R), bortezomib (V), and dexamethasone (d; Dara-RVd). vs. RVd in patients (pts) with newly diagnosed multiple myeloma (MM) eligible for high-dose therapy (HDT) and autologous stem cell transplantation (ASCT) (GRIFFIN). Poster presented at: The Annual Meeting of the American Society of Hematology (ASH); December 9-12, 2017; Atlanta, GA.  
2 Voorhees P, Kaufman J, Laubach J, et al. Daratumumab + lenalidomide, bortezomib & dexamethasone improves depth of response in transplant-eligible newly diagnosed multiple myeloma: GRIFFIN. Oral Presentation presented at: 17th International Myeloma Workshop (IMW); September 12-15, 2019; Boston, MA.  
3 Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood. 2020;136(8):936-945.  
4 Voorhees PM, Rodriguez C, Reeves B, et al. Daratumumab plus RVd for newly diagnosed multiple myeloma: final analysis of the safety run-in cohort of GRIFFIN. Blood Adv. 2021;5(4):1092-1096.  
5 Voorhees PM, Sborov DW, Laubach J, et al. Addition of daratumumab to lenalidomide, bortezomib, and dexamethasone for transplantation-eligible patients with newly diagnosed multiple myeloma (GRIFFIN): final analysis of an open-label, randomised, phase 2 trial. Lancet Haematol. 2023;10(10):e825-e837.  
6 Sborov D, Baljevic M, Reeves B, et al. Daratumumab plus lenalidomide, bortezomib and dexamethasone in newly diagnosed multiple myeloma: analysis of vascular thrombotic events in the GRIFFIN study. Clin Lymphoma Myeloma Leuk. 2022;199(3):355-365.  
7 Chhabra S, Callander N, Watts N, et al. Stem cell mobilization yields with daratumumab- and lenalidomide-containing quadruplet induction therapy in newly diagnosed multiple myeloma: findings from the MASTER and GRIFFIN trials. Transplant Cell Ther. 2023;29(3):174.e1-174.e10.  
8 Sonneveld P, Dimopoulos MA, Boccadoro M, et al. Daratumumab, bortezomib, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 2024;390(4)(4):301-313.  
9 Sonneveld P, Dimopoulos MA, Boccadoro M, et al. Stem cell (SC) yield and transplant with daratumumab + bortezomib, lenalidomide, and dexamethasone in transplant-eligible newly diagnosed multiple myeloma patients in the phase 3 PERSEUS study. Poster presented at: 21st International Myeloma Society (IMS) Annual Meeting; September 25-28, 2024; Rio de Janeiro, Brazil.  
10 Bertamini L, Fokkema C, Rodriguez-Otero P, et al. Circulating tumor cells as a biomarker to identify high-risk transplant-eligible myeloma patients treated with bortezomib, lenalidomide, and dexamethasone with or without daratumumab during induction/consolidation, and lenalidomide with or without daratumumab during maintenance: results from the PERSEUS study. Oral Presentation presented at: The 66th American Society of Hematology (ASH) Annual Meeting and Exposition; December 7-10, 2024; San Diego, CA.  
11 Zweegman S, Usmani S, Chastian K, et al. Bortezomib, lenalidomide, and dexamethasone (VRd) ± daratumumab in patients with newly diagnosed multiple myeloma for whom transplant is not planned as initial therapy: a multicenter, randomized, phase 3 study (CEPHEUS). Poster presented at: The Annual Meeting of the American Society of Clinical Oncology (ASCO). May 31-June 4, 2019; Chicago, IL.  
12 Janssen Research & Development, LLC. A study comparing daratumumab, velcade (bortezomib), lenalidomide, and dexamethasone (D-VRd) with velcade, lenalidomide, and dexamethasone (VRd) in participants with untreated multiple myeloma and for whom hematopoietic stem cell transplant is not planned as initial therapy. In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000- [cited 2025 February 10]. Available from: https://clinicaltrials.gov/ct2/show/NCT03652064 NML Identifier: NCT03652064.  
13 Usmani SZ, Facon T, Hungria V. Daratumumab plus bortezomib, lenalidomide and dexamethasone for transplant-ineligible or transplant-deferred newly diagnosed multiple myeloma: the randomized phase 3 CEPHEUS study. doi.org/10.1038/s41591-024-03485-7. Nat Med. 2025.  
14 Zweegman S, Facon T, Hungria V, et al. Daratumumab + bortezomib, lenalidomide and dexamethasone (VRd) versus VRd alone in patients with newly diagnosed multiple myeloma ineligible for SCT or for whom SCT is not planned as initial therapy: analysis of minimal residual disease in the phase 3 CEPHEUS trial. Oral Presentation presented at: The 66th American Society of Hematology (ASH) Annual Meeting and Exposition; December 7-10, 2024; San Diego, CA.  
15 Chari A, Goldschmidt H, San-Miguel J, et al. Subcutaneous (SC) daratumumab (DARA) in combination with standard multiple myeloma (MM) treatment regimens: an open-label, multicenter phase 2 study (PLEIADES). Oral Presentation presented at: The 17th International Myeloma Workshop (IMW); September 12-15, 2019; Boston, MA.  
16 Chari A, Goldschmidt H, Yang S, et al. Subcutaneous daratumumab plus carfilzomib and dexamethasone in relapsed/refractory multiple myeloma: an open-label, multicenter, phase 2 study (PLEIADES). Poster presented at: The 17th International Myeloma Workshop (IMW); September 12-15, 2019; Boston, MA.  
17 Chari A, Miguel J, McCarthy H, et al. Subcutaneous daratumumab plus standard treatment regimens in patients with multiple myeloma across lines of therapy: PLEIADES study update. Poster presented at: Poster presented at: The 61st American Society of Hematology (ASH) Annual Meeting; December 7-10, 2019; Orlando, FL.  
18 Chari A, Rodriguez-Otero P, McCarthy H, et al. Subcutaneous daratumumab plus standard treatment regimens in patients with multiple myeloma across lines of therapy (PLEIADES): an open-label phase II study. Br J Haematol. 2021;192(5):869-878.  
19 Rodriguez C, Kaufman J, Laubach J, et al. Daratumumab + lenalidomide, bortezomib, and dexamethasone in transplant-eligible newly diagnosed multiple myeloma: a post hoc analysis of sustained minimal residual disease negativity from GRIFFIN. Poster presented at: American Society of Clinical Oncology (ASCO) Annual Meeting; June 3-7, 2022; Chicago, IL/Virtual Meeting.  
20 Nooka AK, Kaufman JL, Rodriguez C, et al. Post hoc analysis of daratumumab plus lenalidomide, bortezomib, and dexamethasone in black patients from final data of the GRIFFIN study. Br J Haematol. 2024;00:1-6.  
21 Callander NS, Silbermann R, Kaufman JL, et al. Daratumumab-based quadruplet therapy for transplant-eligible newly diagnosed multiple myeloma with high cytogenetic risk. Blood Cancer J. 2024;14(1):69.  
22 Rodriguez-Otero P, Moreau P, Dimopoulos MA, et al. Daratumumab (DARA) + bortezomib/lenalidomide/dexamethasone (VRd) with DARA-R (D-R) maintenance in transplant-eligible patients with newly diagnosed multiple myeloma (NDMM): analysis of minimal residual disease (MRD) in the PERSEUS trial. Oral Presentation presented at: American Society of Clinical Oncology (ASCO) Annual Meeting; May 31-June 4, 2024; Chicago, IL, USA.  
23 Dimopoulos MA, Sonneveld P, Rodriguez-Otero P, et al. Daratumumab (DARA)/ bortezomib/lenalidomide/ dexamethasone (D-VRd) with D-R maintenance in transplant-eligible (TE) newly diagnosed multiple myeloma (NDMM): analysis of PERSEUS based on cytogenetic risk. Poster presented at: The European Hematology Association (EHA) Hybrid Congress; June 13-16, 2024; Madrid, Spain.  
24 Dimopoulos MA, Sonneveld P, Rodriguez-Otero P, et al. Daratumumab (DARA SC)/bortezomib/lenalidomide/dexamethasone (D-VRd) with D-R maintenance in transplant-eligible (TE) newly diagnosed myeloma (NDMM): PERSEUS cytogenetic risk analysis. Oral Presentation presented at: 21st International Myeloma Society (IMS) Annual Meeting; September 25-28, 2024; Rio de Janeiro, Brazil.  
25 Rodriguez-Otero P, Voorhees PM, Boccadoro M, et al. Daratumumab plus bortezomib, lenalidomide, and dexamethasone in transplant-eligible patients with multiple myeloma: a pooled analysis of patients aged ≥65 years from both PERSEUS and GRIFFIN studies. Poster presented at: The 21st International Myeloma Society (IMS) Annual Meeting; September 25-28, 2024; Rio de Janeiro, Brazil.  
26 Voorhees P, Kaufman J, Laubach J, et al. Depth of response to daratumumab (DARA), lenalidomide, bortezomib, and dexamethasone (RVd) improves over time in patients (pts) with transplant-eligible newly diagnosed multiple myeloma (NDMM): GRIFFIN study update. Oral Presentation presented at: 61st American Society of Hematology (ASH) Annual Meeting & Exposition; December 7-10, 2019; Orlando, FL.  
27 Kaufman J, Laubach J, Sborov D, et al. Daratumumab (DARA) plus lenalidomide, bortezomib, and dexamethasone (RVd) in patients with transplant-eligible newly diagnosed multiple myeloma (NDMM): updated analysis of GRIFFIN after 12 months of maintenance therapy. Oral Presentation presented at: Virtual 62nd American Society of Hematology (ASH) Annual Meeting & Exposition; December 5-8, 2020; Virtual.  
28 Laubach J, Kaufman JL, Sborov DW, et al. Daratumumab (DARA) plus lenalidomide, bortezomib, and dexamethasone (RVd) in patients (pts) with transplant-eligible newly diagnosed multiple myeloma (NDMM): updated analysis of GRIFFIN after 24 months of maintenance. Oral Presentation presented at: 63rd American Society of Hematology (ASH) Annual Meeting & Exposition; December 11-14, 2021; Atlanta, GA/Virtual Meeting.  
29 Voorhees PM, Sborov DW, Laubach J, et al. Supplement to: Addition of daratumumab to lenalidomide, bortezomib, and dexamethasone for transplantation-eligible patients with newly diagnosed multiple myeloma (GRIFFIN): final analysis of an open-label, randomised, phase 2 trial. Lancet Haematol. 2023;10(10):e825-e837.  
30 Sonneveld P, Dimopoulos MA, Boccadoro M, et al. Supplement to: Daratumumab, bortezomib, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 2023.