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.

DARZALEX FASPRO® (daratumumab and hyaluronidase-fihj)

Medical Information

Use of DARZALEX + DARZALEX FASPRO in High-Risk Multiple Myeloma in Clinical Trials

Last Updated: 02/19/2025

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).
  • DARZALEX FASPRO is approved for MM in combination with bortezomib, lenalidomide, and dexamethasone for induction and consolidation in newly diagnosed patients who are eligible for autologous stem cell transplant.
  • Janssen does not recommend the use of DARZALEX or DARZALEX FASPRO in a manner that is inconsistent with the approved labeling.
  • This response includes information on high-risk subgroups from studies that include patients with newly diagnosed multiple myeloma (NDMM) and relapsed/refractory multiple myeloma (RRMM).
  • 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 daratumumab (D-R) in DVRd group or lenalidomide (R) in VRd group in patients with NDMM eligible for autologous stem cell transplant (ASCT).1
    • Dimopoulos et al (2024)2 presented (at the European Hematology Association [EHA] Hybrid Congress) an expanded sub-group analysis of PERSEUS clinical outcomes (progression-free survival [PFS], overall minimal residual disease [MRD] negativity, and sustained MRD negativity) based on the presence of high-risk cytogenetic abnormalities (HRCAs), including gain(1q21) and amp(1q21). At a median follow-up of 47.5 months, hazard ratio (HR) point estimates for PFS favored D-VRd vs VRd for revised standard (HR, 0.29; 95% confidence interval [CI], 0.15-0.56; P=0.0001) and revised high cytogenetic risk (HR, 0.53; 95% CI, 0.35-0.81; P=0.0027).
    • Dimopoulos et al (2024)3 presented (at the 21st International Myeloma Society [IMS] Annual Meeting) an expanded subgroup analysis of PERSEUS clinical outcomes (PFS, overall MRD-negativity, and sustained MRD-negativity) based on the second revised International Staging System (R2-ISS) and the presence of HRCAs, including gain(1q21) and amp(1q21). At a median follow-up of 47.5 months, HR point estimates for PFS favored D-VRd vs VRd for revised standard risk (HR, 0.29; 95% CI, 0.15-0.56; P=0.0001) and revised high risk (HR, 0.53; 95% CI, 0.35-0.81; P=0.0027). D-VRd increased PFS irrespective of the R2-ISS disease stage with a pronounced effect for R2-ISS stages II (HR, 0.29; 95% CI, 0.14-0.57) and III (HR, 0.46; 95% CI, 0.29-0.75). The overall and sustained (≥12 months) MRD-negativity (at 10-5 and 10-6 threshold) with ≥CR rates based on the R2-ISS disease stage favored D-VRd followed by D-R maintenance over VRd followed by R maintenance.
    • Bertamini et al (2024)4 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 NDMM patients. 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.
  • 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.5-7
  • AURIGA (MMY3021) is an ongoing, open-label, active-controlled, multicenter, randomized phase 3 study evaluating the conversion rate to MRD-negativity after maintenance treatment with DARZALEX FASPRO in combination with lenalidomide (D-R) vs R alone in patients with NDMM who are anti-cluster of differentiation (CD) 38 naïve, have a very good partial response or better (≥VGPR), and are MRD-positive following ASCT after standard-of-care induction/consolidation. A total of 200 patients were randomized (D-R, n=99; R, n=101).8-10
    • Badros et al (2024)8,9,11 reported primary results from the phase 3 AURIGA study. Efficacy data by key subgroups is noted in Table: Subgroup Analysis (ISS Staging, Cytogenetic Risk, and Revised Cytogenetic Risk at Diagnosis) of MRD-Negativity (10- 5) Conversion Rate From Baseline to 12 Months of Maintenance Treatment in the ITT Population.8
    • Foster et al (2024)12 presented (at the 66th ASH Annual Meeting) a post hoc analysis of the phase 3 AURIGA study that evaluated clinically relevant subgroups at a median follow-up of 32.3 months. The clinically relevant subgroups included elderly and Black patients; patients with high-risk disease per the International Staging System (ISS) disease staging; and patients with high cytogenetic risk per standard, revised, and IMS 2024 high-risk criteria. The subgroup analysis of MRD-negativity (10-5 sensitivity) conversion rates by 12 months of maintenance appeared to consistently favor D-R vs lenalidomide (R) maintenance alone, regardless of age, race, ISS disease stage, or response upon study entry. D-R vs R maintenance improved MRD-negativity (10-5 sensitivity) conversion rates by 12 months in patients with ultra high-risk disease, standard-risk disease, and patients with modified IMS 2024 high-risk disease. D-R vs R maintenance showed a PFS benefit across various definitions of high cytogenetic risk, in addition to patients with standard-risk disease and regardless of the number of HRCAs. D-R vs R maintenance improved PFS among patients with gain/amp(1q21), irrespective of other HRCAs (HR, 0.46; 95% CI, 0.13-1.59).
  • MAIA is a phase 3 study that evaluated the safety and efficacy of DARZALEX + lenalidomide and dexamethasone (D-Rd) compared to Rd alone in transplant-ineligible NDMM patients.13 Kumar et al (2022)14 presented (at the 64th ASH Annual Meeting and Exposition) updated efficacy and safety results (median follow-up, 64.5 months), including updated overall survival (OS) results (median follow-up, 73.6 months) from MAIA. Efficacy data by key subgroups is noted in Table: OS in ISS Staging and Cytogenetic Risk Subgroups (MAIA). Moreau et al (2022)15 presented (at the 64th ASH Annual Meeting and Exposition) the efficacy and safety results in clinically important subgroups of patients from the MAIA study, including ISS stage III disease and cytogenetic risk. Efficacy data by key subgroups are noted in Tables: PFS in the ISS Stage III and Cytogenetic Risk Subgroups (ITT Population; MAIA), ORR in the ISS Stage III and Cytogenetic Risk Subgroups (ITT Population; MAIA), MRD-Negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups (ITT Population; MAIA), and Sustained MRD-Negativity Rates in the ISS Stage and Cytogenetic Risk Subgroups Lasting ≥12 months (ITT Population; MAIA).
  • ALCYONE is a phase 3 study which assessed the safety and efficacy of bortezomib, melphalan, and prednisone (VMP) alone and DARZALEX + VMP (D-VMP) in patients with NDMM who were ineligible for high-dose therapy (HDT) with ASCT.16 Mateos et al (2022)17 presented (at the 64th Annual Meeting and Exposition) updated efficacy and safety results from ALCYONE at a median follow-up of 78.8 months. Efficacy data by cytogenetic risk status are noted in Table: OS in ISS Staging and Cytogenetic Risk Subgroups (ALCYONE).17
  • Jakubowiak et al (2022)18 conducted a pooled analysis of patient-level data from the MAIA and ALCYONE studies, analyzing PFS and best response in transplant-ineligible patients with NDMM and HRCAs. The median PFS was 21.2 months in the pooled DARZALEX cohort vs 19.3 months in the pooled control cohort (adjusted HR, 0.59; 95% CI, 0.41-0.85; P=0.0046), representing a 41% reduction in risk of disease progression or death. Complete response or better (≥CR) was achieved in 41.6% (n=42) of patients in the pooled DARZALEX cohort vs 22.5% (n=20) of patients in the pooled control cohort (adjusted odds ratio [OR], 2.63; 95% CI, 1.34-5.16; P=0.0051).
  • OCTANS is a phase 3, multicenter, randomized, active-controlled, open-label study evaluating the efficacy and safety of D-VMP vs VMP alone in Asian patients with NDMM ineligible for high-dose chemotherapy with ASCT.19,20
    • Hou et al (2021)21 presented (at the 63rd ASH Annual Meeting & Exposition) clinical benefit results in patient subgroups within the pooled population of Asian patients from the OCTANS study and global patients from the ALCYONE study with NDMM ineligible for ASCT. Among patients with high cytogenetic risk, although median PFS improved with D-VMP (19.5 months) vs VMP (18.1 months), PFS benefit was less pronounced than that observed for patients with standard cytogenetic risk (median PFS: D-VMP, not reached [NR]; VMP, 17.4 months).
    • Fu et al (2023)22 evaluated PFS by MRD status in a pooled population of Asian and global patients from the OCTANS and ALCYONE studies. In a pooled analysis of OCTANS and ALCYONE, D-VMP vs VMP alone was associated with prolonged PFS across most of the high-risk subgroups evaluated. PFS in the high-risk subgroups of the OCTANS and ALCYONE studies is noted in Table: PFS by High-Risk Subgroups in Pooled Data from the OCTANS and ALCYONE Studies.
  • CASSIOPEIA is a 2-part, phase 3 study evaluating the safety and efficacy of DARZALEX + bortezomib, thalidomide, and dexamethasone (D-VTd) in transplant eligible patients with NDMM.23 Moreau et al (2021)24 reported results from Part 2 of the CASSIOPEIA study. Efficacy data by key subgroups is noted in Table: PFS in the Prespecified ISS and Cytogenetic Risk Subgroups (CASSIOPEIA Part 2). Sonneveld et al (2019)25 presented a subgroup analysis of patients with high-risk NDMM in Part 1 of the CASSIOPEIA study based on cytogenetic risk status and ISS stage. Efficacy data from the subgroup analyses of the study are presented in Table: Post-consolidation Response and MRD Status for High-Risk Patients (CASSIOPEIA Part 1).
  • GRIFFIN is a phase 2, 2-part study evaluating the safety and efficacy of DARZALEX when administered in combination with VRd (D-VRd) in patients with NDMM eligible for HDT and ASCT.26-28 Chari et al (2024)29,30 reported the final efficacy and safety analysis results of clinically relevant subgroups from the GRIFFIN study, including ISS stage III disease and cytogenetic risk. Efficacy data by key subgroups are noted in Tables: Final Analysis of MRD-Negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN), Final Analysis of sCR in the ISS Stage III and Cytogenetic Subgroups (GRIFFIN), Final Analysis of MRD-negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups with a Best Response of ≥CR (GRIFFIN), and Final Analysis of PFS in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN).
  • MASTER (ClinicalTrials.gov identifier: NCT03224507) evaluated the efficacy and safety of DARZALEX + carfilzomib + lenalidomide + dexamethasone (D-KRd) induction followed by autologous hematopoietic cell transplantation (AHCT) and MRD-adapted consolidation therapy in patients with NDMM.31 Costa et al (2023)31 reported the results from the final analysis of the MASTER study at a median follow-up of 42.2 months. The overall MRDnegative remission rate (next-generation sequencing [NGS]; <105 threshold) in MRD-evaluable patients at any point in treatment was 81%. The 3-year PFS rate was 88% for patients with standard-risk (0 HRCAs), 79% for patients with high-risk (1 HRCA), and 50% for patients with ultra high-risk (≥2 HRCAs) cytogenetic abnormalities. The 3-year OS rate was 94% (95% CI, 88-98) for patients with 0 HRCA, 92% (95% CI, 86-96) for patients with 1 HRCA, and 75% (95% CI, 63-85) for patients with ≥2 HRCAs. Costa et al (2023)32 reported additional details on PFS and OS for different study populations and at difference timepoints according to MRD status.
  • Callander et al (2024)33 reported the results of a post hoc analysis evaluating the clinical efficacy of DARZALEX-based quadruplet therapies, including D-KRd and D-VRd, in transplant-eligible patients with NDMM and HRCAs from the MASTER and GRIFFIN studies, respectively. In the MASTER study, in patients with 0, 1, and ≥2 HRCAs, respectively, ≥CR was achieved by 90.6%, 89.1%, and 70.8% of patients; the estimated 24-month PFS rate was 92.4%, 95.7%, and 65.5%; and MRD-negativity (105) was reported in 80%, 86.4%, and 83.3% of patients. In the GRIFFIN study, in patients with 0, 1, and ≥2 HRCAs, respectively, ≥CR was achieved by 90.9%, 78.8%, and 61.5% of patients; the 24-month PFS rate was 96.7%, 93.8%, and 64.2%; and MRD-negativity (10-5) was reported in 76.1%, 55.9%, and 61.5% of patients. The total number of deaths reported in MASTER and GRIFFIN studies were 3 and 8, respectively.
  • CASTOR is a phase 3 study assessing the safety and efficacy of bortezomib and dexamethasone (Vd) alone and DARZALEX + Vd (D-Vd) in patients with RRMM.34 Sonneveld et al (2022)35 reported updated efficacy and safety results from CASTOR at a median follow-up of 72.6 months. Efficacy data by key subgroups is noted in the Table: OS in ISS Staging and Cytogenetic Risk Pre-Specified Subgroups (ITT Population; CASTOR). Weisel et al (2020)36 reported the updated efficacy and safety data based on cytogenetic risk status of DVd from CASTOR in patients with RRMM. Overall response rate (ORR), very good partial response or better (≥VGPR), and ≥CR were higher with D-Vd vs Vd, regardless of cytogenetic risk status. Efficacy data for cytogenetic risk subgroups are noted in Table: Response and MRD-negative Rates in Patients with Standard and High Cytogenetic Risk (CASTOR).
  • POLLUX is a phase 3 study assessing the safety and efficacy of Rd vs D-Rd in patients with RRMM.37 Dimopoulos et al (2022)38 presented (at the 3rd Annual Meeting of the EMN) updated efficacy and safety results from POLLUX at a median follow-up of 79.7 months. Efficacy data by key subgroups is noted in Table: OS in ISS Staging and Cytogenetic Risk Pre-Specified Subgroups (ITT Population; POLLUX). Kaufman et al (2020)39 presented updated efficacy and safety data based cytogenetic risk status of DRd from POLLUX in patients with RRMM. Among patients with high cytogenetic risk, PFS was prolonged with D-Rd vs Rd in MRD-positive patients (median, 20.3 months vs 8.3 months; HR, 0.49; 95% CI, 0.23-1.03; P=0.0567); median PFS with D-Rd in MRD-negative patients was not estimable (NE). MRD-negativity rates were higher with D-Rd vs Rd, regardless of cytogenetic risk as noted in Table: Response and MRD-negativity Rates in Patients with Standard and High Cytogenetic Risk (POLLUX).
  • Mateos et al (2022)40 presented (at the 19th IMS Annual Meeting) the results of a post hoc subgroup analysis of the phase 3 CASTOR and POLLUX studies, evaluating the efficacy of D-Vd vs Vd alone (CASTOR) and D-Rd vs Rd alone (POLLUX) in patients with RRMM. In the pooled intent-to-treat (ITT) population of CASTOR and POLLUX, PFS benefit of the DARZALEX (median, 20.3 months) vs control (median, 7.3 months) arm was observed (HR, 0.45; 95% CI, 0.31-0.65) and OS benefit of the DARZALEX (median, 40 months) vs control (median, 23.6 months) arm was observed (HR, 0.68; 95% CI, 0.47-1) in patients with high cytogenetic risk.
  • CANDOR is a phase 3 study evaluating the safety and efficacy of D-Kd (n=312) vs carfilzomib and dexamethasone (Kd, n=154) in patients with RRMM.41,42 Usmani et al (2023)43 reported the final efficacy and safety results of the CANDOR study after a median follow-up of approximately 50 months. The HR for OS favored the D-Kd arm across prespecified subgroups including ISS stage and cytogenetic risk status and is presented in the Table: OS in Pre-specified ISS Staging and Cytogenetic Risk Subgroups (CANDOR).
  • APOLLO is a phase 2 study evaluating the safety of pomalidomide + lenalidomide (Pd) vs DARZALEX FASPRO + Pd (D-Pd) in patients with RRMM who received ≥1 prior treatment with both lenalidomide and a proteasome inhibitor (PI).44,45 Sonneveld et al (2021)46 presented (at the 63rd ASH Annual Meeting & Exposition) updated efficacy and safety data at a median follow-up of 30.7 months. PFS favored D-Pd vs Pd across subgroups, including ISS stage and cytogenetic risk status and is presented in the Table: PFS in the ISS Staging and Cytogenetic Profile Subgroups (APOLLO). Dimopoulos et al (2022)47 presented (at 64th ASH Annual Meeting and Exposition) final OS results and updated safety data at a median follow-up of 39.6 months. The updated analysis did not discuss cytogenetic risk status and the citation is listed in the reference below.
  • Giri et al (2020)48 conducted a systematic review and meta-analysis of 6 randomized clinical studies which evaluated the efficacy relative to DARZALEX in combination with backbone MM regimens for patients with newly diagnosed or relapsed/refractory high-risk multiple myeloma (HRMM). The percentage of patients with HRMM ranged from 15% to 33% in the included studies. The analysis reported an improved PFS associated with the addition of DARZALEX to various backbone MM regimens in patients with relapsed/refractory HRMM (pooled HR, 0.15; 95% CI, 0.30-0.67; P<0.001) and newly diagnosed HRMM (pooled HR, 0.67; 95% CI, 0.4-0.95; P=0.02).

PRODUCT LABELING

CLINICAL studies - nDMM

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 DVRd group or lenalidomide in VRd arm in patients with NDMM eligible for ASCT. Sonneveld et al (2023)1 reported efficacy and safety results from the PERSEUS study evaluating D-VRd vs VRd in patients with NDMM eligible for ASCT. Dimopoulos et al (2024)2 presented an expanded analysis of PERSEUS clinical outcomes (PFS, overall MRD negativity, and sustained MRD negativity) based on the presence of HRCAs, including gain(1q21) and amp(1q21). The results of the expanded analysis are presented below. Dimopoulos et al (2024)3 presented (at the 21st IMS Annual Meeting) an expanded subgroup analysis of PERSEUS clinical outcomes (PFS, overall MRD-negativity, and sustained MRD-negativity) based on R2-ISS and the presence of HRCAs, including gain(1q21) and amp(1q21).

Study Design/Methods

  • A total of 709 patients were randomized 1:1 to into D-VRd (n=355) vs VRd (n=354) arm.1
    • Stratification was done based on the 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]).1
  • Primary endpoint: PFS.1
  • Key secondary endpoints: Overall ≥CR, overall MRD-negativity, and OS.1
  • Other secondary endpoints: ORR, ≥VGPR, stringent complete response (sCR), duration of MRD-negativity.1

Results

Patient Characteristics

Demographics and Baseline Clinical Characteristics of the High-Risk ITT Populationa,2,3
D-VRd
(n=355)

VRd
(n=354)

Cytogenetic abnormalities, n (%)
   del(17p)
36 (10.1)
34 (9.6)
   t(4;14)
33 (9.3)
38 (10.7)
   t(14;16)
11 (3.1)
14 (4)
   Gain(1q21)b
59 (16.6)
71 (20.1)
   Amp(1q21)c
28 (7.9)
36 (10.2)
Cytogenetic risk profiled, n (%)
   Standard risk
264 (74.4)
266 (75.1)
   High risk
76 (21.4)
78 (22)
      del(17p)
36 (10.1)
34 (9.6)
      t(4;14)
33 (9.3)
38 (10.7)
      t(14;16)
11 (3.1)
14 (4)
   Indeterminate
15 (4.2)
10 (2.8)
Revised cytogenetic riske, n (%)
   Revised standard risk
174 (49)
167 (47.2)
   Revised high risk
130 (36.6)
148 (41.8)
   Indeterminate
51 (14.4)
39 (11)
ISS disease stage, n/N (%)
   I
186/355 (52.4)
178/353 (50.4)
   II
114/355 (32.1)
125/353 (35.4)
   III
55/355 (15.5)
50/353 (14.2)
R2-ISS disease stage, n (%)
   Low (I)
116 (32.7)
114 (32.2)
   Low-intermediate (II)
111 (31.3)
106 (29.9)
   Intermediate-high (III)
108 (30.4)
115 (32.5)
   High (IV)
20 (5.6)
19 (5.4)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; FISH; fluorescence in situ hybridization; ISS, International Staging System; ITT, intent-to-treat; R2-ISS, second revised International Staging System; VRd, bortezomib + lenalidomide + dexamethasone.
aThe ITT population was defined as all patients who underwent randomization.
bGain(1q21) was defined as the presence of 3 copies of chromosome 1q21.cAmp(1q21) was defined as the presence of 4 or more copies of chromosome 1q21. dCytogenetic risk was based on FISH; high risk was defined as the presence of del(17p), t(4;14), or t(14;16).eRevised cytogenetic risk was defined as the presence of del(17p), t(4;14), t(14;16), gain(1q21), or amp(1q21).

  • Across all cytogenetic risk subgroups, PFS was favored for D-VRd followed by D-R maintenance vs VRd followed by R maintenance and is summarized in Table: Cytogenetic Risk Subgroup Analysis of PFS in the ITT Population.2
    • HR point estimates for PFS favored D-VRd vs VRd for revised standard (HR, 0.29; 95% CI, 0.15-0.56; P=0.0001) and revised high cytogenetic risk (HR, 0.53; 95% CI, 0.35-0.81; P=0.0027).2,3
    • Irrespective of other HRCAs, HR point estimates for PFS also favored D-VRd vs VRd in patients with the presence of gain(1q21), amp(1q21), and gain(1q21) or amp(1q21).2
  • D-VRd increased PFS irrespective of the R2-ISS disease stage with a pronounced effect for R2-ISS stages II and III and is summarized in Table: Subgroup Analysis of PFS Based on R2-ISS Disease Stage.3
  • Regardless of high-risk cytogenetic markers, a subgroup analysis of overall and sustained (≥12 months) MRD negativity (at 10-5 and 10-6) ≥CR rates based on cytogenetic risk markers favored treatment with D-VRd followed by D-R maintenance over VRd followed by R maintenance and is summarized in Table: Subgroup Analysis of MRD Negativity.

Cytogenetic Risk Subgroup Analysis of PFS in the ITT Population2
Subgroup
D-VRd
D-VRd
VRd
VRd
HR (95% CI)
P Value
n/N
Median PFS,
Months

n/N
Median PFS,
Months

Standard riska
25/264
NE
62/266
NE
0.35 (0.22-0.56)
<0.0001
High riskb
24/76
NE
38/78
44.1
0.59 (0.36-0.99)
0.0439
Revised standard riskc
12/174
NE
35/167
NE
0.29 (0.15-0.56)
0.0001
Revised high riskd
33/130
NE
62/148
NE
0.53 (0.35-0.81)
0.0027
gain(1q21)e
15/59
NE
26/71
NE
0.62 (0.33-1.18)
0.1400
amp(1q21)f
6/28
NE
17/36
46.7
0.37 (0.15-0.94)
0.0306
gain(1q21)/ amp(1q21)g
21/87
NE
43/107
NE
0.52 (0.31-0.88)
0.0133
Isolated gain(1q21)h
8/37
NE
15/47
NE
0.57 (0.24-1.36)
0.2004
Isolated amp(1q21)i
1/17
NE
9/23
NE
0.11 (0.01-0.87)
0.0115
1 revised HRCA
21/97
NE
43/110
NE
0.47 (0.28-0.79)
0.0035
≥2 revised HRCAs
12/33
NE
19/38
44.1
0.73 (0.35-1.50)
0.3878
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; HR, hazard ratio; HRCA, high-risk cytogenetic abnormality; ITT, intent-to-treat; NE, not estimable; PFS, progression-free survival; VRd, bortezomib + lenalidomide + dexamethasone.
aStandard risk (per protocol), defined as none of the following HRCAs: del(17p), t(4;14), t(14;16).
bHigh risk (per protocol), defined as 1 or more of the following HRCAs: del(17p), t(4;14), t(14;16).
cDefined as none of the following HRCAs: del(17p), t(4;14), t(14;16), gain(1q21), amp(1q21).
dDefined as 1 or more of the following HRCAs: del(17p), t(4;14), t(14;16), gain(1q21), amp(1q21).
eDefined as the presence of 3 copies of chromosome 1q21, with or without other HRCAs.
fDefined as the presence of 4 or more copies of chromosome 1q21, with or without other HRCAs.
gDefined as the presence of gain(1q21) or amp(1q21), with or without other HRCAs.
hDefined as the presence of 3 copies of chromosome 1q21, without any other HRCAs.
iDefined as the presence of 4 or more copies of chromosome 1q21, without any other HRCAs.


Subgroup Analysis of PFS Based on R2-ISS Disease Stage3
Subgroup
D-VRd
D-VRd
VRd
VRd
HR (95% CI)
n/N
Median PFS,
Months

n/N
Median PFS,
Months

Low (I)
6/116
NE
13/114
NE
0.42 (0.16-1.11)
Low-intermediate (II)
11/111
NE
30/106
NE
0.29 (0.14-0.57)
Intermediate-high (III)
25/108
NE
50/115
NE
0.46 (0.29-0.75)
High (IV)
8/20
NE
10/19
39.8
0.63 (0.25-1.61)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; HR, hazard ratio; ITT, intent-to-treat; NE, not estimable; PFS, progression-free survival; VRd, bortezomib + lenalidomide + dexamethasone.

Subgroup Analysis of MRD Negativity2,3
Subgroup
D-VRd
VRd
OR (95% CI)
P Value
n/N (%)
n/N (%)
MRD negativity (10-5) with ≥CR
   Standard riska
204/264 (77.3)
128/266 (48.1)
3.67 (2.52-5.33)
<0.0001
   High riskb
52/76 (68.4)
37/78 (47.4)
2.40 (1.24-4.63)
0.0086
   Revised standard riskc
131/174 (75.3)
79/167 (47.3)
3.39 (2.14-5.37)
<0.0001
   Revised high riskd
95/130 (73.1)
73/148 (49.3)
2.79 (1.68-4.62)
<0.0001
   gain(1q21)e
41/59 (69.5)
33/71 (46.5)
2.62 (1.27-5.41)
0.0086
   amp(1q21)f
24/28 (85.7)
20/36 (55.6)
4.80 (1.38-16.69)
0.0104
   gain(1q21)/amp(1q21)g
65/87 (74.7)
53/107 (49.5)
3.01 (1.63-5.56)
0.0004
   Isolated gain(1q21)h
27/37 (73)
23/47 (48.9)
2.82 (1.12-7.10)
0.0268
   Isolated amp(1q21)i
16/17 (94.1)
13/23 (56.5)
12.31 (1.39-109.10)
0.0093
   1 revised HRCA
73/97 (75.3)
55/110 (50)
3.04 (1.68-5.51)
0.0002
   ≥2 revised HRCAs
22/33 (66.7)
18/38 (47.4)
2.22 (0.85-5.83)
0.1044
   Low (I)
92/116 (79.3)
55/114 (48.2)
4.11 (2.30-7.35)
-
   Low-intermediate (II)
84/111 (75.7)
49/106 (46.2)
3.62 (2.03-6.45)
-
   Intermediate-high (III)
79/108 (73.1)
55/115 (47.8)
2.97 (1.70-5.21)
-
   High (IV)
12/20 (60)
9/19 (47.4)
1.67 (0.47-5.93)
-
Sustained MRD negativity (10-5) lasting ≥12 months
   Standard riska
183/264 (69.3)
83/266 (31.2)
4.98 (3.45-7.20)
<0.0001
   High riskb
37/76 (48.7)
20/78 (25.6)
2.75 (1.40-5.42)
0.0032
   Revised standard riskc
115/174 (66.1)
53/167 (31.7)
4.19 (2.67-6.59)
<0.0001
   Revised high riskd
77/130 (59.2)
41/148 (27.7)
3.79 (2.30-6.26)
<0.0001
   gain(1q21)e
37/59 (62.7)
21/71 (29.6)
4 (1.92-8.34)
0.0002
   amp(1q21)f
20/28 (71.4)
10/36 (27.8)
6.50 (2.17-19.48)
0.0006
   gain(1q21)/amp(1q21)g
57/87 (65.5)
31/107 (29)
4.66 (2.54-8.56)
<0.0001
   Isolated gain(1q21)h
25/37 (67.6)
15/47 (31.9)
4.44 (1.77-11.17)
0.0012
   Isolated amp(1q21)i
15/17 (88.2)
6/23 (26.1)
21.25 (3.71-121.61)
0.0001
   1 revised HRCA
60/97 (61.9)
31/110 (28.2)
4.13 (2.31-7.41)
<0.0001
   ≥2 revised HRCAs
17/33 (51.5)
10/38 (26.3)
2.97 (1.10-8.04)
0.0303
   Low (I)
82/116 (70.7)
38/114 (33.3)
4.82 (2.76-8.43)
-
   Low-intermediate (II)
76/111 (68.5)
27/106 (25.5)
6.35 (3.51-11.49)
-
   Intermediate-high (III)
63/108 (58.3)
36/115 (31.3)
3.07 (1.77-5.32)
-
   High (IV)
9/20 (45)
4/19 (21.1)
3.07 (0.75-12.59)
-
MRD negativity (10-6) with ≥CR
   Standard riska
177/264 (67)
88/266 (33.1)
4.12 (2.87-5.91)
<0.0001
   High riskb
44/76 (57.9)
24/78 (30.8)
3.09 (1.60-6.00)
0.0007
   Revised standard riskc
115/174 (66.1)
56/167 (33.5)
3.86 (2.47-6.05)
<0.0001
   Revised high riskd
82/130 (63.1)
48/148 (32.4)
3.56 (2.17-5.84)
<0.0001
   gain(1q21)e
36/59 (61)
22/71 (31)
3.49 (1.69-7.20)
0.0006
   amp(1q21)f
21/28 (75)
15/36 (41.7)
4.20 (1.42-12.39)
0.0082
   gain(1q21)/amp(1q21)g
57/87 (65.5)
37/107 (34.6)
3.59 (1.98-6.52)
<0.0001
   Isolated gain(1q21)h
24/37 (64.9)
15/47 (31.9)
3.94 (1.58-9.80)
0.0028
   Isolated amp(1q21)i
14/17 (82.4)
9/23 (39.1)
7.26 (1.62-32.60)
0.0069
   1 revised HRCA
63/97 (64.9)
35/110 (31.8)
3.97 (2.23-7.08)
<0.0001
   ≥2 revised HRCAs
19/33 (57.6)
13/38 (34.2)
2.61 (1.00-6.83)
0.0500
   Low (I)
78/116 (67.2)
37/114 (32.5)
4.27 (2.46-7.41)
-
   Low-intermediate (II)
76/111 (68.5)
31/106 (29.2)
5.25 (2.94-9.38)
-
   Intermediate-high (III)
65/108 (60.2)
40/115 (34.8)
2.83 (1.65-4.88)
-
   High (IV)
12/20 (60)
6/19 (31.6)
3.25 (0.87-12.14)
-
Sustained MRD negativity (10-6) lasting ≥12 months
   Standard riska
137/264 (51.9)
54/266 (20.3)
4.24 (2.88-6.22)
<0.0001
   High riskb
23/76 (30.3)
11/78 (14.1)
2.64 (1.18-5.90)
0.0160
   Revised standard riskc
87/174 (50)
35/167 (21)
3.77 (2.34-6.07)
<0.0001
   Revised high riskd
55/130 (42.3)
23/148 (15.5)
3.99 (2.27-7.01)
<0.0001
   gain(1q21)e
25/59 (42.4)
11/71 (15.5)
4.01 (1.76-9.15)
0.0007
   amp(1q21)f
17/28 (60.7)
6/36 (16.7)
7.73 (2.42-24.63)
0.0003
   gain(1q21)/amp(1q21)g
42/87 (48.3)
17/107 (15.9)
4.94 (2.53-9.63)
<0.0001
   Isolated gain(1q21)h
19/37 (51.4)
9/47 (19.1)
4.46 (1.69-11.77)
0.0020
   Isolated amp(1q21)i
13/17 (76.5)
3/23 (13)
21.67 (4.15-113.02)
<0.0001
   1 revised HRCA
45/97 (46.4)
18/110 (16.4)
4.42 (2.32-8.42)
<0.0001
   ≥2 revised HRCAs
10/33 (30.3)
5/38 (13.2)
2.87 (0.87-9.51)
0.0797
   Low (I)
58/116 (50)
24/114 (21.1)
3.75 (2.10-6.69)
-
   Low-intermediate (II)
56/111 (50.5)
18/106 (17)
4.98 (2.65-9.34)
-
   Intermediate-high (III)
47/108 (43.5)
22/115 (19.1)
3.26 (1.79-5.94)
-
   High (IV)
7/20 (35)
2/19 (10.5)
4.58 (0.81-25.80)
-
Abbreviations: CI, confidence interval; ≥CR, complete response or better; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; HRCA, high-risk cytogenetic abnormality; MRD, minimal residual disease; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.
aStandard risk (per protocol), defined as none of the following HRCAs: del(17p), t(4;14), t(14;16).
bHigh risk (per protocol), defined as 1 or more of the following HRCAs: del(17p), t(4;14), t(14;16).
cRevised standard risk, defined as none of the following HRCAs: del(17p), t(4;14), t(14;16), gain(1q21), amp(1q21).
dRevised high risk, defined as 1 or more of the following HRCAs: del(17p), t(4;14), t(14;16), gain(1q21), amp(1q21).
eDefined as the presence of 3 copies of chromosome 1q21, with or without other HRCAs.
fDefined as the presence of 4 or more copies of chromosome 1q21, with or without other HRCAs.
gDefined as the presence of gain(1q21) or amp(1q21), with or without other HRCAs.
hDefined as the presence of 3 copies of chromosome 1q21, without any other HRCAs.
iDefined as the presence of 4 or more copies of chromosome 1q21, without any other HRCAs.

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

Bertamini et al (2024)4 presented (at the 66th ASH Annual Meeting) results from the PERSEUS study that highlighted the significance of CTC as a biomarker in transplant-eligible NDMM patients.

Results

Patient Characteristics

Baseline Characteristics in Patients From the PERSEUS Study - CTC Subgroup4
Characteristic
D-VRd
(n=231)
VRd
(n=220)
P Value
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 cell; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; 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 improved PFS in patients with high cytogenetic risk and low CTC levels. However, there was no improvement in the outcomes for patients with high cytogenetic risk combined with high CTC levels.4 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).4

Prognostic Impact of CTC and Other Risk Factors on PFS4
Risk Factor
HR (95% CI)
P Value
CTC (log10)
1.36 (1.15-1.6)
≤0.05
Cytogenetics high riska
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 cell; 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.

DARZALEX FASPRO in Combination with Bortezomib, Lenalidomide, and Dexamethasone

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 transplant ineligible (TIE) or for whom transplant is not planned as initial therapy (transplant deferred).5-7 Usmani et al (2025)7 reported results from the phase 3 CEPHEUS study.

Study Design/Methods

  • The trial has enrolled 395 patients from 13 different countries including the United States.5,6
  • Primary endpoint: Overall MRD-negativity (≥CR) rate at 10-5 sensitivity threshold.5,6
  • Key secondary endpoints: PFS, ORR, ≥VGPR, ≥CR, PFS2, OS, and sustained MRD-negativity (10-5) rate at ≥12 months.5,6

Results

Patient Characteristics

Baseline Demographics and Clinical Characteristics of the High-Risk ITT Population7
D-VRd (n=197)
VRd (n=198)
ISS disease stagea, n (%)
   I
68 (34.5)
68 (34.3)
   II
73 (37.1)
75 (37.9)
   III
56 (28.4)
55 (27.8)
Cytogenetic risk profileb, n (%)
   Standard risk
149 (75.6)
149 (75.3)
   High risk
25 (12.7)
27 (13.6)
   Indeterminatec
23 (11.7)
22 (11.1)
Abbreviations: D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intent-to-treat; VRd, bortezomib + lenalidomide + dexamethasone.
aBased on the combination of serum β2-microglobulin and albumin levels. Higher stages indicate more advanced disease. bAssessed by fluorescence in situ hybridization; high risk was defined as the presence of del(17p), t(4;14), and/or t(14;16). cIndeterminate includes patients with missing or unevaluable samples.

Efficacy

Prespecified Subgroup Analysis of Overall MRD-Negativity Rates (ISS Staging and Cytogenic Risk)7
Subgroup
D-VRd
VRd
OR (95% CI)
Number of Patients With MRD-Negativity/
Total Number of Patients (%)

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)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; ISS, International Staging System; MRD, minimal residual disease; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.

PFS in Prespecified Subgroups (ISS Staging and Cytogenic Risk)7
Subgroups
D-VRd
VRd
D-VRd
VRd
HR (95% CI)
Number of Disease Progression Events or Deaths/Total Number of Patients
Median PFS, months
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)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX FASPRO + bortezomib + lenalidomide + dexamethasone; HR, hazard ratio; ISS, International Staging System; NE, not estimated; PFS, progression-free survival; VRd, bortezomib + lenalidomide + dexamethasone.
Safety
  • The safety data were consistent with the established safety profile of each individual drug.7
    • Overall, deaths were reported in 51 vs 60 patients from the D-VRd vs VRd arm, respectively.
    • 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%).
    • 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).

DARZALEX FASPRO in Combination with Lenalidomide

AURIGA (MMY3021; clinicaltrials.gov identifier: NCT03901963) is an ongoing, open-label, active-controlled, multicenter, randomized, phase 3 study evaluating the conversion rate to MRDnegativity after maintenance treatment with DR vs lenalidomide alone in patients with NDMM who are anti-CD 38 naïve, have ≥VGPR, and are MRD positive after ASCT.8-10 Badros et al (2024)8,9 reported primary results from the phase 3 AURIGA study.

Study Design/Methods

  • The trial enrolled 200 patients from the United States and Canada.8
  • Patients underwent 1:1 randomization to receive D-R (n=99) or R alone (n=101) across 28-day cycles.8,10
    • D-R: DARZALEX FASPRO 1800 mg SC QW in cycles 1-2, Q2W in cycles 3-6, Q4W in cycles 7+.
    • R: Lenalidomide 10 mg PO once a dayon days 1-28.
  • The treatment regimen continued until unacceptable toxicity, disease progression, consent withdrawal, or for a maximum of 36 cycles.8
  • Primary endpoint: MRD-negativity conversion rate from baseline by 12 months.8
    • MRD was assessed at 12, 18, 24, and 36 months.
    • If lenalidomide is well tolerated, the dose may be increased to 15 mg daily after cycle 3, at the investigator’s discretion.
  • Key secondary endpoints: Safety, PFS, overall MRD-negativity conversion rate, sustained MRD-negativity rate (≥6 months), response rates including CR/sCR as assessed by International Myeloma Working Group 2016 criteria, duration of ≥CR, OS, HRQoL changes based on patient reported outcomes.8

Results

Patient Characteristics
  • A total of 200 patients were randomized into the D-R maintenance (n=99) and R alone maintenance (n=101) arms.8
  • The median duration of treatment was 30.7 (range, 0.7-37.5) months vs 20.6
    (range, 0-37.7) months in the D-R vs R arm, respectively.8
  • The baseline demographics and disease characteristics of the ITT population are presented in Table: Baseline Demographics and Disease Characteristics of the High-Risk ITT Population.8
  • The median follow-up was 32.3 months.8
  • Patients in the D-R vs R arm received a median of 33 (range, 1-36) vs 21.5
    (range, 1-36) maintenance cycles, respectively.8
  • Overall, 85 of 96 (88.5%) vs 77 of 98 (78.6%) patients in the D-R vs R arm completed ≥12 maintenance cycles, respectively.8

Baseline Demographics and Disease Characteristics of the High-Risk ITT Population8
D-R (n=99)
R (n=101)
ISS disease stagea, n (%)
   I
40 (44)
38 (38.8)
   II
28 (30.8)
37 (37.8)
   III
23 (25.3)
23 (23.5)
Median induction cycles (range)b, n
5 (4-8)
5 (4-8)
Cytogenetic risk at diagnosisc, n (%)
   Standard risk
63 (68.5)
66 (74.2)
   High riskd
22 (23.9)
15 (16.9)
      del(17p)
13 (14.1)
3 (3.4)
      t(4;14)
10 (10.9)
12 (13.5)
      t(14;16)
6 (6.5)
7 (7.9)
   Unknown
7 (7.6)
8 (9)
Revised cytogenetic risk at diagnosise, n (%)
   Standard risk
52 (55.9)
53 (59.6)
   High riskf
32 (34.4)
30 (33.7)
      del(17p)
13 (14)
3 (3.4)
      t(4;14)
10 (10.8)
12 (13.5)
      t(14;16)
6 (6.5)
7 (7.9)
      t(14;20)
1 (1.1)
2 (2.2)
      gain/amp(1q21)
16 (17.2)
22 (24.7)
   Unknown
9 (9.7)
6 (6.7)
Abbreviations: D-R, DARZALEX FASPRO+lenalidomide; ISS, International Staging System; ITT, intent-to-treat; NR, not reported; R, lenalidomide.
a
D-R vs R: n=91 vs n=98, respectively.
bD-R vs R: n=98 vs n=99, respectively.
cD-R vs R: n=92 vs n=89, respectively.
dHigh risk is defined as positive for any of del(17p), t(14;16), or t(4;14).
eD-R vs R: n=93 vs n=89, respectively.
fRevised high-risk cytogenetics is defined as ≥1 abnormality from del(17p), t(4;14), t(14;16), t(14;20), and gain/amp(1q21).

Efficacy

Subgroup Analysis of MRD-Negativity (10-5) Conversion Rate From Baseline to 12 Months of Maintenance Treatment in the High-Risk ITT Population8
Subgroup, n/N (%)
D-R (n=99)
R (n=101)
OR (95% CI)
ISS staging at diagnosis
   I
19/40 (47.5)
8/38 (21.1)
3.39 (1.25-9.19)
   II
13/28 (46.4)
7/37 (18.9)
3.71 (1.23-11.25)
   III
15/23 (65.2)
3/23 (13)
12.50 (2.83-55.25
Cytogenetic risk at diagnosis
   High riska
7/22 (31.8)
1/15 (6.7)
6.53 (0.71-60.05)
   Standard risk
35/63 (55.6)
14/66 (21.2)
4.64 (2.15-10.04)
Revised cytogenetic risk at diagnosis
   High riskb
14/32 (43.8)
4/30 (13.3)
5.06 (1.43-17.88)
   Standard risk
28/52 (53.8)
12/53 (22.6)
3.99 (1.72-9.26)
Abbreviations: CI, confidence interval; D-R, DARZALEX FASPRO+lenalidomide; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; OR, odds ratio; R, lenalidomide.
aHigh risk is defined as positive for any of the following abnormalities: del(17p), t(14;16), or t(4;14).
bRevised high-risk cytogenetics is defined as ≥1 abnormality from del(17p), t(4;14), t(14;16), t(14;20), and gain/amp(1q21).

Safety
  • Slightly higher occurrence rates of grade 3/4 cytopenias (54.2% vs 46.9%) and infections (18.8% vs 13.3%) were observed with D-R vs R.8
  • Serious AEs were reported in 30.2% vs 22.4% of patients in the D-R vs R arm, respectively.8
  • Any TEAEs occurred in 99% of patients in both the D-R and R arms.8
    • Grade 3/4 TEAEs occurred in 74% and 67.3% of patients in the D-R and R arms, respectively.
  • During the analysis, 32 of 96 (33.3%) vs 47 of 98 (48%) patients in the D-R vs R arm, respectively, discontinued ≥1 component of the study treatment.8

Clinically Relevant Subgroup Analysis - Phase 3 AURIGA Study

Foster et al (2024)12  presented (at the 66th ASH Annual Meeting) a post hoc analysis of the phase 3 AURIGA study that evaluated clinically relevant subgroups at a median follow-up of 32.3 months. The clinically relevant subgroups included elderly and Black patients; patients with high-risk disease per ISS disease staging; and patients with high cytogenetic risk per standard, revised, and IMS 2024 high-risk criteria.

Results

Demographics and Disease Characteristics

Demographics and Disease Characteristics of the ITT Population12
D-R (n=99)
R (n=101)
ISS disease stage, n (%)
   n
91
98
   I
40 (44)
38 (38.8)
   II
28 (30.8)
37 (37.8)
   III
23 (25.3)
23 (23.5)
Cytogenetic risk at diagnosis per standard definitiona,b, n (%)
   n
92
89
   Standard-risk
63 (68.5)
66 (74.2)
   High-risk
22 (23.9)
15 (16.9)
   Unknown
7 (7.6)
8 (9)
Cytogenetic risk at diagnosis per revised definitionc, n (%)
   n
93
89
   Revised standard risk (0 HRCAs)
52 (55.9)
53 (59.6)
   Revised high-risk (≥1 HRCA)
32 (34.4)
30 (33.7)
      1 HRCA
21 (22.6)
20 (22.5)
      ≥2 HRCAs
11 (11.8)
10 (11.2)
      Gain/amp(1q21)
16 (17.2)
22 (24.7)
      Isolated gain/amp(1q21)
10 (10.8)
15 (16.9)
   Unknown
9 (9.7)
6 (6.7)
Cytogenetic risk per modified IMS 2024 criteriad, n (%)
   n
93
90
   Modified IMS 2024 standard-risk
67 (72)
68 (75.6)
   Modified IMS 2024 high-risk
17 (18.3)
8 (8.9)
      ≥20% del(17p)
10 (10.8)
2 (2.2)
      t(4;14)/(14;16)/(14;20) + gain/amp(1q21) and/or del(1p32)
5 (5.4)
6 (6.7)
      Del(1p32) + gain/amp(1q21)
4 (4.3)
0 (0)
   Unknown
9 (9.7)
14 (15.6)
Abbreviations: D-R, DARZALEX FASPRO + lenalidomide; HRCA, high-risk cytogenetic abnormalities; IMS, International Myeloma Society; IMWG, International Myeloma Working Group; ISS, International Staging System; ITT, intention-to-treat; R, lenalidomide.
aHigh-risk cytogenetics per the standard definition are defined as ≥1 abnormality including del(17p), t(4;14), or t(14;16). bThe imbalance in cytogenetic risk between arms, especially a higher number of patients with del(17p) for patients randomized to the D-R arm, was since some assessments were made on cytogenetic data at screening and some on cytogenetic data at the time of diagnosis.cRevised high-risk cytogenetics per the revised definition are defined as ≥1 abnormality including del(17p), t(4;14), t(14;16), t(14;20), or gain/amp(1q21).dHigh risk per the modified IMS 2024 criteria is defined as the presence of ≥20% del(17p); or the association of ≥2 of the following: t(4;14) or t(14;16) or t(14;20); gain/amp(1q21); or del(1p32) [in the AURIGA study, data were not available on TP53 mutations, baseline ß2M, and creatinine levels and differentiation between monoallelic versus biallelic del(1p32)].

Efficacy
  • A subgroup analysis of MRD-negativity (10-5 sensitivity) conversion rates by 12 months of maintenance showed improvement for D-R vs R regardless of age, race, ISS disease stage, or response upon study entry.12 The results are summarized in Table: Subgroup Analysis of MRD-negativity (10-5 Sensitivity) Conversion Ratea from Baseline to 12 Months of Maintenance Treatment.
  • D-R vs R improved MRD-negativity (10-5 sensitivity) conversion rates by 12 months of maintenance in patients with ultra-high-risk disease and standard-risk disease.12 The results are summarized in Table: Subgroup Analysis of MRD-negativity (10-5 Sensitivity) Conversion Ratea from Baseline to 12 Months of Maintenance Treatment by Risk Status.
  • D-R vs R improved MRD-negative (10-5 sensitivity) conversion rate by 12 months of maintenance, among patients with modified IMS 2024 high-risk disease.12 The results are summarized in Table: Subgroup Analysis of MRD-negativity (10-5 Sensitivity) Conversion Rate from Baseline to 12 Months of Maintenance Treatment by Modified IMS 2024 Criteria.
  • D-R vs R maintenance showed a PFS benefit across various definitions of high cytogenetic risk, in addition to patients with standard-risk disease. The HR and 95% CI values presented below are derived from a Cox proportional hazards model with treatment as the sole explanatory variable. A HR <1 indicates an advantage for D-R maintenance.12 
    • Standard criteria: High-risk cytogenetics was defined as ≥1 abnormality including del(17p), t(4;14), or t(14;16).
      • Standard-risk: HR, 0.59; 95% CI, 0.23-1.49.
      • High-risk: HR, 0.60; 95% CI, 0.21-1.70.
    • Revised criteria: Revised high-risk cytogenetics was defined as ≥1 abnormality including del(17p), t(4;14), t(14;16), t(14;20), or gain/amp(1q21).
      • Revised standard-risk: HR, 0.69; 95% CI, 0.24-1.95.
      • Revised high-risk: HR, 0.53; 95% CI, 0.21-1.31.
    • Modified IMS 2024 criteria: High risk per the modified IMS 2024 criteria was defined as the presence of ≥20% del(17p); or the association of ≥2 of the following: t(4;14) or t(14;16) or t(14;20); gain/amp(1q21); or del(1p32). In the AURIGA study, data were not available on TP53 mutations, baseline ß2M, and creatinine levels and differentiation between monoallelic versus biallelic del(1p32).
      • Modified IMS 2024 standard-risk: HR, 0.42; 95% CI, 0.16-1.07.
      • Modified IMS 2024 high-risk: HR, 0.45; 95% CI, 0.13-1.53.
  • D-R vs R maintenance demonstrated a PFS benefit regardless of the number of HRCAs. The HRCA numbers presented below were defined as number of abnormalities from del(17p), t(4;14), t(14;16), t(14;20), or gain/amp(1q21). Cytogenetic results at diagnosis (based on case report forms collected data from local laboratories) were used in the analysis. The HR and 95% CI were derived from a Cox proportional hazards model with treatment as the sole explanatory variable. A HR <1 indicates an advantage for D-R maintenance.12 
    • 0 HRCAs: HR, 0.69; 95% CI, 0.24-1.95.
    • 1 HRCAs: HR, 0.36; 95% CI, 0.09-1.45.
    • ≥2 HRCAs: HR, 0.61; 95% CI, 0.17-2.25.
  • D-R vs R maintenance improved PFS among patients with gain/amp(1q21), irrespective of other HRCAs (HR, 0.46; 95% CI, 0.13-1.59).12 

Subgroup Analysis of MRD-negativity (10-5 Sensitivity) Conversion Ratea from Baseline to 12 Months of Maintenance Treatment12
Subgroup, n/N (%)
D-R
R
ORb (95% CI)
ITTc
50/99 (50.5)
19/101 (18.8)
4.51 (2.37-8.57)
Age
   <65 years
30/61 (49.2)
12/61 (19.7)
3.95 (1.76-8.85)
   ≥65 years
20/38 (52.6)
7/40 (17.5)
5.24 (1.86-14.74)
Race
   White
31/67 (46.3)
14/68 (20.6)
3.32 (1.55-7.10)
   Black
12/20 (60)
4/24 (16.7)
7.50 (1.85-30.34)
ISS staging at diagnosis
   I
19/40 (47.5)
8/38 (21.1)
3.39 (1.25-9.19)
   II
13/28 (46.4)
7/37 (18.9)
3.71 (1.23-11.25)
   III
15/23 (65.2)
3/23 (13)
12.50 (2.83-55.25)
Baseline response statusd
   <CR
27/71 (38)
11/71 (15.5)
3.35 (1.50-7.46)
   ≥CR
23/28 (82.1)
8/30 (26.7)
12.65 (3.58-44.64)
Abbreviations: CI, confidence interval; CR, complete response; D-R, DARZALEX FASPRO + lenalidomide; IMWG, International Myeloma Working Group; ISS, International Staging System; ITT, intention-to-treat; MRD, minimal residual disease; NGS, nextgeneration sequencing; OR, odds ratio; R, lenalidomide.
aDefined as the proportion of patients who achieved MRD-negative status (at 10–5) by NGS by 12 months after maintenance treatment and prior to progressive disease or subsequent antimyeloma therapy.
b
Mantel-Haenszel estimate of the common OR for stratified tables is used for ITT; Mantel-Haenszel estimate of the common OR for unstratified tables is used for subgroups. An OR >1 indicates an advantage for D-R.cITT analysis set is defined as all patients who were randomized to treatment.dResponse status upon entering the study as assessed by IMWG 2016 criteria.


Subgroup Analysis of MRD-negativity (10-5 Sensitivity) Conversion Ratea from Baseline to 12 Months of Maintenance Treatment by Risk Status12
Subgroup, n/N (%)
D-R
R
ORb (95% CI)
Cytogenetic risk at diagnosis
   Standard risk
35/63 (55.6)
14/66 (21.2)
4.64 (2.15-10.04)
   High riskc
7/22 (31.8)
1/15 (6.7)
6.53 (0.71-60.05)
Revised cytogenetic risk at diagnosis
   Revised standard risk (0 HRCAs)
28/52 (53.8)
12/53 (22.6)
3.99 (1.72-9.26)
   Revised high-risk (≥1 HRCA)d
14/32 (43.8)
4/30 (13.3)
5.06 (1.43-17.88)
   1 HRCA
8/21 (38.1)
4/20 (20)
2.46 (0.60-10.04)
   ≥2 HRCAs
6/11 (54.5)
0/10 (0)
NE (NE-NE)e
   Gain/amp(1q21)
10/16 (62.5)
3/22 (13.6)
10.56 (2.17-51.42)
   Isolated gain/amp(1q21)
7/10 (70)
3/15 (20)
9.33 (1.46-59.48)
Abbreviations: CI, confidence interval; D-R, DARZALEX FASPRO + lenalidomide; HRCA, high-risk cytogenetic abnormalities; MRD, minimal residual disease; NE, not estimable; NGS, nextgeneration sequencing; OR, odds ratio; R, lenalidomide.
aDefined as the proportion of patients who achieved MRD-negative status (at 10–5) by NGS by 12 months after maintenance treatment and prior to progressive disease or subsequent antimyeloma therapy.bMantel-Haenszel estimate of the common OR for unstratified tables is used. An OR >1 indicates an advantage for D-R.cHigh-risk cytogenetics per the standard definition are defined as ≥1 abnormality including del(17p), t(4;14), or t(14;16). dRevised high-risk cytogenetics per the revised definition are defined as ≥1 abnormality including del(17p), t(4;14), t(14;16), t(14;20), or gain/amp(1q21). eNot evaluable because no patient in the R group had MRD-negative conversion.


Subgroup Analysis of MRD-negativity (10-5 Sensitivity) Conversion Rate from Baseline to 12 Months of Maintenance Treatment by Modified IMS 2024 Criteriaa,b,c,12
Subgroup, n/N (%)
D-R
R
Modified IMS 2024 standard-risk
34/67 (50.7)
16/68 (23.5)
Modified IMS 2024 high-risk
7/17 (41.2)
0/8 (0)
≥20% del(17p)
2/10 (20)
0/2 (0)
t(4;14)/(14;16)/(14;20) + gain/amp(1q21) and/or del(1p32)
2/5 (40)
0/6 (0)
Del(1p21) + gain/amp(1q21)
3/4 (75)
0/0 (0)
Abbreviations: D-R, DARZALEX FASPRO + lenalidomide; IMS, International Myeloma Society; MRD, minimal residual disease; R, lenalidomide.
aHigh risk per the modified IMS 2024 criteria is defined as the presence of ≥20% del(17p), or the association of ≥2 of the following: t(4;14) or t(14;16) or t(14;20); gain/amp(1q21); or del(1p32) [in the AURIGA study, data were not available on TP53 mutations, baseline ß2M, and creatinine levels and differentiation between monoallelic versus biallelic del(1p32)]. bDefined as the proportion of patients who achieved MRD-negative status (at 10–5) by NGS by 12 months after maintenance treatment and prior to progressive disease or subsequent antimyeloma therapy. cDefined as the proportion of patients who achieved MRD-negative status any time after the date of randomization.

DARZALEX in Combination with Lenalidomide and Dexamethasone

MAIA (MMY3008) is an international, phase 3, randomized, open-label, active-controlled, multicenter study that evaluated the efficacy and safety of combination of D-Rd vs Rd in patients with NDMM not eligible for high-dose chemotherapy and ASCT (N=737).13 Kumar et al (2022)14 presented the results of the updated efficacy and safety analysis (median follow-up, 64.5 months), including updated OS results (median follow-up, 73.6 months), of the MAIA study in patients with NDMM who were ineligible for high-dose chemotherapy and ASCT. Results in the high-risk cytogenetic subgroup are summarized below.

Study Design/Methods

  • Primary endpoint: PFS
  • Secondary endpoints: ≥CR rate, ≥VGPR rate, duration of response (DOR), MRD-negativity rate (10-5) via NGS, ORR, OS, PFS on subsequent line of therapy (PFS2), sCR, time to next (2nd-line) treatment, time to response (TTR), time to progression (TTP), and safety
  • Cytogenetic analysis: efficacy in the subgroup of patients with a high-risk cytogenic profile (defined by del17p, t[14;16], or t[4;14] abnormality [or a combination of these] on FISH or karyotype analysis)

Results

Efficacy
  • At a median follow-up of 73.6 months, the median OS in the D-Rd vs Rd arm was NR vs 64.1 months (HR, 0.65; 95% CI, 0.52-0.80; P<0.0001).
    • The 60-month OS rate in the D-Rd vs Rd arm was 66.7% vs 53.7%. Among patients with high-risk cytogenetic profile, median OS was 55.6 months vs 42.5 months in the D-Rd vs Rd arm, respectively (HR, 0.65; 95% CI, 0.39-1.06). See Table: OS in ISS Staging and Cytogenetic Risk Subgroups (MAIA).

OS in ISS Staging and Cytogenetic Risk Subgroups (MAIA)14
Subgroup
D-Rda
Rda
HR (95% CI)a,b
n/N
Median OS, Months
n/N
Median OS, Months
ISS disease stage
   I
28/98
NE
36/103
NE
0.78 (0.48-1.28)
   II
64/163
NE
88/156
61.7
0.59 (0.43-0.81)
   III
58/107
65.2
78/110
47.3
0.66 (0.47-0.93)
Cytogenetic risk at study entry
   High riskc
28/48
55.6
36/44
42.5
0.65 (0.39-1.06)
   Standard risk
105/271
NE
147/279
65.5
0.64 (0.50-0.82)
Abbreviations: CI, confidence interval; D-Rd, daratumumab + lenalidomide + dexamethasone; HR, hazard ratio; IgG, immunoglobulin G; ISS, International Staging System; NE, not estimable; OS, overall survival; Rd, lenalidomide + dexamethasone.
aData are based on a median follow-up of 73.6 months.
bHR and 95% CI from a Cox proportional hazards model with treatment as the sole explanatory variable. An HR <1 indicates an advantage for D-Rd.
cPatients with high cytogenetic risk were positive by fluorescence in situ hybridization or karyotype testing for ≥1 of the following cytogenetic abnormalities: t(4;14), t(14;16), or del(17p).

Safety
  • No new safety concerns were reported with the longer follow-up.
  • Grade 3/4 TEAEs occurring in ≥20% of patients in the D-Rd vs Rd arm were neutropenia (54.1% vs 37%) and anemia (17% vs 21.6%).
  • The most common serious TEAE in both arms was pneumonia (D-Rd, 18.7%; Rd, 10.7%).
  • The rate of treatment discontinuation due TEAEs in the D-Rd vs Rd arm was 14.6% vs 23.8%.

Analysis of Clinically Important Subgroups from the MAIA Study

Moreau et al (2022)15 presented the efficacy and safety results in clinically important subgroups of patients from the MAIA study.

Study Design/Methods

  • Efficacy outcomes (PFS, ORR, and MRD-negativity) were analyzed in ISS stage III and cytogenetic risk based on the following patient characteristics:
    • ISS stage III disease
    • Cytogenetic risk
      • Standard cytogenetic risk (0 of the following HRCAs: t[4;14], t[14;16], and del[17p])
      • High cytogenetic risk (≥1 of the following HRCAs: t[4;14], t[14;16], and del[17p])
      • Revised standard cytogenetic risk (0 of the following HRCAs: t[4;14], t[14;16], del[17p], t[14;20], gain[1q21], and amp[1q21])
      • Revised high cytogenetic risk (≥1 of the following HRCAs: t[4;14], t[14;16], del[17p], t[14;20], gain[1q21], and amp[1q21])
      • Gain(1q21) (3 copies of chromosome 1q21, with or without other HRCAs)
      • Amp(1q21) (≥4 copies of chromosome 1q21, with or without other HRCAs)
      • Gain(1q21) or amp(1q21) (3 or ≥4 copies of chromosome 1q21, with or without other HRCAs)
      • 1 HRCA (only 1 of the following HRCAs: t[4;14], t[14;16], del[17p], t[14;20], gain[1q21], or amp[1q21])
      • Two or more HRCAs (≥2 of the following HRCAs: t[4;14], t[14;16], del[17p], t[14;20], gain[1q21], and amp[1q21])
      • Isolated gain(1q21) (3 copies of chromosome 1q21, without any other HRCAs)
      • Isolated amp(1q21) (≥4 copies of chromosome 1q21, without any other HRCAs)
      • Isolated gain(1q21) or amp(1q21) (3 or ≥4 copies of chromosome 1q21, without any other HRCAs)
      • Gain(1q21) or amp(1q21) plus ≥1 HRCA (3 or ≥4 copies of chromosome 1q21, plus ≥1 of the following HRCAs: t[4;14], t[14;16], del[17p], and t[14;20])

Results

Patient Characteristics
  • Overall, 737 patients (D-Rd, 368; Rd, 369) were included in the ITT population.
  • The median duration of follow-up was 64.5 months.
Efficacy

PFS in the ISS Stage III and Cytogenetic Risk Subgroups (ITT Population; MAIA)15
Subgroup
D-Rd
Rd
HR (95% CI)a
n/N
Median PFS, Month
n/N
Median PFS, Month
ITT (overall)
176/368
61.9
228/369
34.4
0.55 (0.45-0.67)
Baseline characteristic
   ISS stage III
61/107
42.4
73/110
24.2
0.61 (0.43-0.86)
Cytogenetic risk
      Standard cytogenetic risk
126/271
63.8
174/279
34.4
0.51 (0.41-0.64)
      High cytogenetic risk
28/48
45.3
31/44
29.6
0.57 (0.34-0.96)
      Revised standard cytogenetic risk
78/176
NR
115/187
35.1
0.50 (0.37-0.66)
      Revised high cytogenetic risk
82/156
56
96/152
30.7
0.59 (0.44-0.80)
      Gain(1q21)
20/53
NR
28/44
37.8
0.43 (0.24-0.76)
      Amp(1q21)
48/74
40
45/76
26.1
0.81 (0.54-1.21)
      Gain(1q21) or amp(1q21)
68/127
53.2
73/120
32.3
0.63 (0.46-0.88)
      1 HRCA
68/137
61.4
86/137
31.2
0.55 (0.40-0.76)
      ≥2 HRCAs
14/19
24.9
10/15
24
0.92 (0.40-2.10)
      Isolated gain(1q21)
16/47
NR
27/42
37.8
0.36 (0.19-0.67)
      Isolated amp(1q21)
38/61
42.8
38/65
28.9
0.78 (0.50-1.22)
      Isolated gain(1q21) or amp(1q21)
54/108
61.4
65/107
37.1
0.58 (0.40-0.83)
      Gain(1q21) or amp(1q21) plus
      ≥1 HRCA
14/19
24.9
8/13
24
1.03 (0.42-2.48)
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; HR, hazard ratio; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; NR, not reached; PFS, progression-free survival; Rd, lenalidomide + dexamethasone.
aHR <1 indicates an advantage for D-Rd.


ORR in the ISS Stage III and Cytogenetic Risk Subgroups (ITT Population; MAIA)15
Subgroup
D-Rd
n/N (%)

Rd
n/N (%)

OR (95% CI)a
ITT (overall)
342/368 (92.9)
301/369 (81.6)
2.97 (1.84-4.79)
Baseline characteristic
   ISS stage III
93/107 (86.9)
86/110 (78.2)
1.85 (0.90-3.81)
Cytogenetic risk
      Standard cytogenetic risk
253/271 (93.4)
228/279 (81.7)
3.14 (1.78-5.54)
      High cytogenetic risk
44/48 (91.7)
33/44 (75)
3.67 (1.07-12.55)
      Revised standard cytogenetic risk
162/176 (92)
149/187 (79.7)
2.95 (1.54-5.66)
      Revised high cytogenetic risk
147/156 (94.2)
126/152 (82.9)
3.37 (1.52-7.46)
      Gain(1q21)
51/53 (96.2)
39/44 (88.6)
3.27 (0.60-17.75)
      Amp(1q21)
70/74 (94.6)
63/76 (82.9)
3.61 (1.12-11.65)
      Gain(1q21) or amp(1q21)
121/127 (95.3)
102/120 (85)
3.56 (1.36-9.30)
      1 HRCA
129/137 (94.2)
114/137 (83.2)
3.25 (1.40-7.56)
      ≥2 HRCAs
18/19 (94.7)
12/15 (80)
4.50 (0.42-48.53)
      Isolated gain(1q21)
46/47 (97.9)
37/42 (88.1)
6.22 (0.70-55.56)
      Isolated amp(1q21)
57/61 (93.4)
55/65 (84.6)
2.59 (0.77-8.75)
      Isolated gain(1q21) or amp(1q21)
103/108 (95.4)
92/107 (86)
3.36 (1.17-9.60)
      Gain(1q21) or amp(1q21) plus ≥1 HRCA
18/19 (94.7)
10/13 (76.9)
5.40 (0.49-59.02)
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; OR, odds ratio; ORR, overall response rate; Rd, lenalidomide + dexamethasone.
aOR >1 indicates an advantage for D-Rd.


MRD-Negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups (ITT Population; MAIA)15
Subgroup
D-Rd
n/N (%)

Rd
n/N (%)

OR (95% CI)a
ITT (overall)
118/368 (32.1)
41/369 (11.1)
3.78 (2.55-5.59)
Baseline characteristic
   ISS stage III
29/107 (27.1)
12/110 (10.9)
3.04 (1.46-6.34)
Cytogenetic risk
      Standard cytogenetic risk
93/271 (34.3)
33/279 (11.8)
3.89 (2.50-6.06)
      High cytogenetic risk
12/48 (25)
1/44 (2.3)
14.33 (1.78-115.59)
      Revised standard cytogenetic risk
60/176 (34.1)
21/187 (11.2)
4.09 (2.36-7.09)
      Revised high cytogenetic risk
49/156 (31.4)
15/152 (9.9)
4.18 (2.22-7.86)
      Gain(1q21)
19/53 (35.8)
6/44 (13.6)
3.54 (1.27-9.89)
      Amp(1q21)
23/74 (31.1)
8/76 (10.5)
3.83 (1.59-9.27)
      Gain(1q21) or amp(1q21)
42/127 (33.1)
14/120 (11.7)
3.74 (1.92-7.30)
      1 HRCA
44/137 (32.1)
15/137 (10.9)
3.85 (2.02-7.34)
      ≥2 HRCAs
5/19 (26.3)
0/15 (0)
NE (NE-NE)
      Isolated gain(1q21)
17/47 (36.2)
6/42 (14.3)
3.40 (1.19-9.71)
      Isolated amp(1q21)
20/61 (32.8)
8/65 (12.3)
3.48 (1.39-8.66)
      Isolated gain(1q21) or amp(1q21)
37/108 (34.3)
14/107 (13.1)
3.46 (1.74-6.89)
      Gain(1q21) or amp(1q21) plus ≥1
      HRCA

5/19 (26.3)
0/13 (0)
NE (NE-NE)
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; NE, not evaluable; OR, odds ratio; Rd, lenalidomide + dexamethasone.
aOR >1 indicates an advantage for D-Rd.


Sustained MRD-Negativity Rates in the ISS Stage and Cytogenetic Risk Subgroups Lasting ≥12 months (ITT Population; MAIA)15
Subgroup
D-Rd
n/N (%)

Rd
n/N (%)

OR (95% CI)a
ITT (overall)
69/368 (18.8)
15/369 (4.1)
5.45 (3.05-9.72)
Baseline characteristic
   ISS stage III
17/107 (15.9)
3/110 (2.7)
6.74 (1.91-23.73)
Cytogenetic risk
      Standard cytogenetic risk
55/271 (20.3)
11/279 (3.9)
6.20 (3.17-12.14)
      High cytogenetic risk
6/48 (12.5)
0/44 (0)
NE (NE-NE)
      Revised standard cytogenetic risk
31/176 (17.6)
5/187 (2.7)
7.78 (2.95-20.52)
      Revised high cytogenetic risk
32/156 (20.5)
7/152 (4.6)
5.35 (2.28-12.53)
      Gain(1q21)
14/53 (26.4)
3/44 (6.8)
4.91 (1.31-18.40)
      Amp(1q21)
13/74 (17.6)
4/76 (5.3)
3.84 (1.19-12.38)
      Gain(1q21) or amp(1q21)
27/127 (21.3)
7/120 (5.8)
4.36 (1.82-10.44)
      1 HRCA
31/137 (22.6)
7/137 (5.1)
5.43 (2.30-12.83)
      ≥2 HRCAs
1/19 (5.3)
0/15 (0)
NE (NE-NE)
      Isolated gain(1q21)
14/47 (29.8)
3/42 (7.1)
5.52 (1.46-20.86)
      Isolated amp(1q21)
12/61 (19.7)
4/65 (6.2)
3.73 (1.13-12.31)
      Isolated gain(1q21) or amp(1q21)
26/108 (24.1)
7/107 (6.5)
4.53 (1.87-10.97)
      Gain(1q21) or amp(1q21) plus ≥1 HRCA
1/19 (5.3)
0/13 (0)
NE (NE-NE)
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; NE, not evaluable; OR, odds ratio; Rd, lenalidomide + dexamethasone.
aOR >1 indicates an advantage for D-Rd.

Safety (Patients Aged ≥75 Years)
  • Among patients aged ≥75 years, grade 3/4 TEAEs were reported in 95.5% vs 95% patients in the D-Rd vs Rd arm.
    • The most common (≥20%) grade 3/4 TEAEs were neutropenia (D-Rd, 62.4%; Rd, 41.5%), lymphopenia (D-Rd, 21%; Rd, 12.6%), anemia (DRd, 20.4%; Rd, 25.2%), and pneumonia (D-Rd, 20.4%; Rd, 14.5%).
  • Serious TEAEs were reported in 80.9% vs 79.2% of patients in the D-Rd vs Rd arm, the most common of which was pneumonia (D-Rd, 19.7%; Rd, 12.6%).
  • TEAEs led to treatment discontinuation in 15.3% vs 27.7% of patients in the D-Rd vs Rd arm.
  • TEAEs resulting in death were reported in 11.5% vs 13.2% of patients in the D-Rd vs Rd arm.

DARZALEX in Combination with Bortezomib, Melphalan, and Prednisone

ALCYONE (MMY3007) was a phase 3, multicenter, randomized, open-label, active-controlled study that assessed the safety and efficacy of VMP alone and D-VMP in patients with NDMM who were ineligible for HDT with ASCT.16 Mateos et al (2022)17 presented updated analysis of the ALCYONE study that evaluated the efficacy and safety of D-VMP vs VMP in patients with NDMM who were ineligible for high-dose chemotherapy and ASCT. Results in the high-risk cytogenetic subgroup are summarized below.

Study Design/Methods

  • Primary endpoint: PFS
  • Key secondary endpoints: CR rate, ORR, OS, safety
  • Cytogenetic analysis: efficacy in the subgroup of patients with a high-risk cytogenic profile (defined by del17p, t[14;16], or t[4;14] abnormality [or a combination of these] on FISH or karyotype analysis)

Results

Patient Characteristics
  • A total of 98 patients (D-VMP, n=53; VMP, n=45) were classified as high cytogenetic risk and 518 patients (D-VMP, n=261; VMP, n=257) were classified as standard cytogenetic risk.
Efficacy
  • Median duration of follow-up was 78.8 months.
  • In the D-VMP vs VMP arm, the median OS was 82.7 vs 53.6 months (HR, 0.64; 95% CI, 0.52-0.79; P<0.0001) and the 72-month OS rate was 55.7% vs 39.7%.

OS in ISS Staging and Cytogenetic Risk Subgroups (ALCYONE)17
Subgroup
D-VMP
VMP
HR (95% CI)a
n/N
Median OS (months)
n/N
Median OS (months)
ISS disease stage
   I
18/69
NE
26/67
NE
0.52 (0.29-0.96)
   II
63/139
83
88/160
61.3
0.72 (0.52-1)
   III
79/142
63
93/129
42.3
0.57 (0.42-0.78)
Cytogenetic risk at study entry
   High riskb
33/53
46.2
31/45
39.5
0.85 (0.52-1.38)
   Standard risk
113/261
83
149/257
55.1
0.58 (0.45-0.74)
Abbreviations: CI, confidence interval; D-VMP, daratumumab + bortezomib + melphalan + prednisone; HR, hazard ratio; ISS, International Staging System; NE, not estimable; OS, overall survival; VMP, bortezomib + melphalan + prednisone.
aHR and 95% CI from a Cox proportional hazards model with treatment as the sole explanatory variable. An HR <1 indicates an advantage for D-VMP.
bPatients with high cytogenetic risk were positive by fluorescence in situ hybridization or karyotype testing for ≥1 of the following cytogenetic abnormalities: t(4;14), t(14;16), or del(17p).

Safety
  • In the D-VMP vs VMP arm, grade 3/4 TEAEs were reported in 82.9% vs 77.4% of patients, and grade 3/4 infections were reported in 30.3% vs 15% of patients.
  • Grade 3/4 TEAEs occurring in ≥20% of patients in the D-VMP vs VMP arm were neutropenia (40.5% vs 39%) and thrombocytopenia (34.7% vs 37.9%).
  • The most common serious TEAE in both treatment arms was pneumonia (D-VMP, 14.7%; VMP, 3.7%).
  • The rate of treatment discontinuation due to TEAEs in the D-VMP vs VMP arm was 9% vs 9.3%.

Pooled Analysis of MAIA and ALCYONE Studies

Jakubowiak et al (2022)18 reported the results of a pooled analysis of patient-level data of transplant-ineligible patients with NDMM and high cytogenetic risk from the MAIA and ALCYONE studies.

Study Design/Methods

  • Patients with baseline high-risk cytogenetics [del(17p), t(4;14), or t(14;16)] were included.
  • Primary endpoint: PFS
  • Secondary endpoint: ORR, best response, ≥VGPR, MRD-negative CR

Results

Patient Characteristics
  • Overall, 101 patients (D-Rd [MAIA], n=48; DARZALEX + bortezomib + melphalan + prednisone arm [D-VMP; ALCYONE], n=53) were included in the pooled DARZALEX cohort and 89 patients (Rd [MAIA], n=44; VMP [ALCYONE], n=45) were included in the pooled control cohort.
  • The combined median follow-up duration of both the trials was 43.7 months (MAIA, 47.9 months; ALCYONE, 40.1 months).
  • In the pooled DARZALEX and control cohorts, the median duration of treatment was 19.9 and 12 months, respectively.
  • Baseline characteristics of patients within the MAIA and ALCYONE studies are summarized in Table: Key Baseline Characteristics in MAIA and ALCYONE.

Key Baseline Characteristics in MAIA and ALCYONE18
Characteristic, n (%)
MAIA
ALCYONE
D-Rd (n=48)
Rd (n=44)
Standardized Differencea
D-VMP (n=53)
VMP (n=45)
Standardized Differencea
ISS stageb
   I
6 (12.5)
8 (18.2)
15.8%
6 (11.3)
4 (8.9)
8.1%
   II
21 (43.8)
15 (34.1)
19.9%
23 (43.4)
18 (40)
6.9%
   III
21 (43.8)
21 (47.7)
8%
24 (45.3)
23 (51.1)
11.7%
Cytogenetic riskc
   del(17p)
25 (52.1)
29 (65.9)
28.4%
29 (54.7)
27 (60)
10.7%
   t(4;14)
21 (43.8)
12 (27.3)
35%
25 (47.2)
17 (37.8)
19.1%
   t(14;16)
4 (8.3)
5 (11.4)
10.2%
6 (11.3)
6 (13.3)
6.1%
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; D-VMP, DARZALEX + bortezomib + melphalan + prednisone; ECOG PS, Eastern Cooperative Oncology Group performance status; FISH, fluorescence in situ hybridization; FLC, free light chain; IgG, immunoglobulin G; ISS, International Staging System; MM, multiple myeloma; Rd, lenalidomide + dexamethasone; VMP, bortezomib + melphalan + prednisone.
aStandardized difference is a measure of effect size independent of sample size, where characteristics with a standardized difference <10% were considered balanced.
bISS staging was derived based on the combination of serum β2-microglobulin and albumin.
cCytogenetic risk was based on FISH or karyotype testing.

Efficacy
  • The median PFS was 21.2 months in the pooled DARZALEX cohort vs 19.3 months in the pooled control cohort (adjusted HR, 0.59; 95% CI, 0.41-0.85; P=0.0046), representing a 41% reduction in risk of disease progression or death.
    • After adjusting for age differences, the pooled HR for PFS was 0.59 (95% CI, 0.410.85; P=0.0044).
    • In the subgroup of patients with del(17p) at baseline (pooled DARZALEX, n=54; pooled control, n=56) the adjusted HR for PFS was 0.63 (95% CI, 0.39-1.03; P=0.0659).
    • In the individual studies, the adjusted HRs for PFS were 0.73 (95% CI, 0.46-1.14) in the ALCYONE study and 0.57 (95% CI, 0.33-1) in the MAIA study. See Table: HRs for PFS in Patient with High-Risk Cytogenetics (MAIA and ALYCONE).
  • The 36-month PFS rate was 41.3% in the pooled DARZALEX cohort vs 19.9% in the pooled control cohort.

HRs for PFS in Patients with High-Risk Cytogenetics (MAIA and ALCYONE)18
Study Name
Progression Events
Adjusted HR (95% CI)
DARZALEX + Control
n/N

Control
n/N

ALCYONE
41/53
36/45
0.73 (0.46-1.14)
MAIA
23/48
28/44
0.57 (0.33-1)
Pooleda
64/101
64/89
0.59 (0.41-0.85)
Abbreviations: ECOG, Eastern Cooperative Oncology Group; CI, confidence interval; HR, hazard ratio; IgG, immunoglobulin G; ISS, International Staging System; PFS, progression-free survival.
aFor the pooled analysis, a multivariate stratified Cox regression analysis was used to calculate adjusted HR, with the study identifier as the stratification factor. HR was adjusted for cytogenetic abnormalities [ie, del(17p), t(4, 14), 4(14, 16)], baseline ECOG performance status, ISS stage, type of multiple myeloma (ie, IgG vs. non-IgG), and renal impairment (defined as creatinine clearance <60 mL/min).

  • The median time to ≥CR was 9.3 months (range, 3.5-34.5) in the pooled DARZALEX cohort vs 7.1 months (range, 2.3-43.8) in the pooled control cohort. See Table: Response Rates Among ASCT-Ineligible High-Risk NDMM Patients from MAIA and ALYCONE.
  • In the pooled DARZALEX vs control cohorts, the median PFS was NR vs 31 months in patients achieving ≥CR and 16.4 months vs 15.6 months in patients not achieving CR.

Response Rates Among ASCT-Ineligible High-Risk NDMM Patients from MAIA and ALYCONE18
Parameter
DARZALEX + Control (n=101)
Control (n=89)
Relative Response Ratioa
(95% CI)

Adjusted ORb
(95% CI)

P value
Sensitivity Analysis Adjusting for Age
Adjusted ORc (95% CI)
P value
Best response
   ≥CR (sCR + CR)
42
(41.6%)

20 (22.5%)
1.85
(1.18-2.90)

2.63
(1.34-5.16)

0.0051
2.57
(1.30-5.06)

0.0064
      sCR
27 (26.7%)
5 (5.6%)
-
-
-
-
-
      CR
15
(14.9%)

15 (16.9%)
-
-
-
-
-
      MRD-negative CR
25
(24.8%)

5
(5.6%)

4.35
(1.75-10.82)

5.5
(1.97-15.34)

0.0011
5.31
(1.89-14.88)

0.0015
   VGPR
34
(33.7%)

21 (23.6%)
-
-
-
-
-
   PR
17
(16.8%)

25 (28.1%)
-
-
-
-
-
   SD
3
(3%)

19 (21.3%)
-
-
-
-
-
   PD
0 (0%)
0 (0%)
-
-
-
-
-
   NE
5 (5%)
4 (4.5%)
-
-
-
-
-
≥VGPR (sCR + CR + VGPR)
76
(75.2%)

41 (46.1%)
1.64
(1.27-2.10)

4.03
(2.09-7.78)

<0.0001
4.08
(2.10-7.91)

<0.0001
Overall response (sCR + CR + VGPR + PR)
93
(92.1%)

66 (74.2%)
1.24
(1.08-1.42)

4.88
(1.94-12.27)

0.0008
4.71
(1.87-11.88)

0.0010
Abbreviations: ASCT, autologous stem cell transplant; CI, confidence interval; CR, complete response; ECOG,  Eastern Cooperative Oncology Group; IgG, immunoglobulin G; ISS, International Staging System; MM, multiple myeloma; MRD, minimal residual disease; NE, not evaluable; OR, odds ratio; PD, progressive disease; PR, partial response; sCR, stringent complete response; SD, stable disease; VGPR, very good partial response.
a
Relative response ratio was calculated using the Mantel-Haenszel method, with the study identifier as the stratification factor.
bAdjusted OR was calculated using stratified logistic regression analysis, with the study identifier as the stratification factor. OR was adjusted for cytogenetic abnormalities [ie, del(17p), t(4;14), 4(14;16)], baseline ECOG performance status, ISS stage, type of MM (ie, IgG vs non-IgG), and renal impairment (defined as creatinine clearance <60 mL/minute).
cOR was additionally adjusted for age (<75 vs ≥75 years).

DARZALEX in Combination with Bortezomib, Melphalan, and Prednisone in Asian Patients

OCTANS (MMY3011; ClinicalTrials.gov identifier: NCT03217812) is a phase 3, multicenter, randomized, active-controlled, open-label study evaluating the efficacy and safety of D-VMP vs VMP alone in Asian patients (from China, Hong Kong SAR, Taiwan, Korea, and Malaysia) with NDMM ineligible for high-dose chemotherapy with ASCT.19,20

Pooled Analysis of the OCTANS and ALCYONE Studies

Hou et al (2021)21 presented clinical benefit results in patient subgroups within the pooled population of Asian patients from the OCTANS study and global patients from the ALCYONE study with NDMM ineligible for ASCT.


Methods

  • The patient populations from the OCTANS and ALCYONE studies were pooled for the efficacy analyses presented below. The pooled ITT population included all randomized patients.

Results

Baseline Characteristics

Key Demographic and Baseline Characteristics in the ITT Populations from OCTANS and ALCYONE and in the Pooled ITT Population from OCTANS and ALCYONE21
Characteristic
OCTANS
ALCYONE
OCTANS and ALCYONE Pooled
D-VMP
(n=146)

VMP
(n=74)

D-VMP
(n=350)

VMP
(n=356)

D-VMP
(n=496)

VMP
(n=430)

ISS disease stagea, n (%)
   I
37 (25.3)
19 (25.7)
69 (19.7)
67 (18.8)
106 (21.4)
86 (20)
   II
68 (46.6)
32 (43.2)
139 (39.7)
160 (44.9)
207 (41.7)
192 (44.7)
   III
41 (28.1)
23 (31.1)
142 (40.6)
129 (36.2)
183 (36.9)
152 (35.3)
Cytogenetic profileb, n
145
74
314
302
459
376
   Standard risk, n (%)
117 (80.7)
54 (73)
261 (83.1)
257 (85.1)
378 (82.4)
311 (82.7)
   High risk, n (%)
28 (19.3)
20 (27)
53 (16.9)
45 (14.9)
81 (17.6)
65 (17.3)
      del17p
15 (10.3)
9 (12.2)
29 (9.2)
27 (8.9)
44 (9.6)
36 (9.6)
      t(4;14)
15 (10.3)
12 (16.2)
25 (8)
17 (5.6)
40 (8.7)
29 (7.7)
      t(14;16)
7 (4.8)
2 (2.7)
6 (1.9)
6 (2)
13 (2.8)
8 (2.1)
Abbreviations: D-VMP, DARZALEX + bortezomib + melphalan + prednisone; ISS, International Staging System; ITT, intent-to-treat; VMP, bortezomib + melphalan + prednisone.
aBased on the combination of serum β2-microglobulin and albumin.
bCytogenetic risk was based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality. Patients with standard cytogenetic risk had an absence of high-risk cytogenetic abnormalities.

Efficacy
  • Among patients with standard cytogenetic risk, a PFS benefit was observed with D-VMP vs VMP.
    • Median PFS: D-VMP, NR; VMP, 17.4 months (HR, 0.41; 95% CI, 0.31-0.54; P<0.0001)
    • 12-month PFS: D-VMP, 86.7%; VMP, 74.3%
    • 18-month PFS: D-VMP, 76.4%; VMP, 46.6%
  • Among patients with high cytogenetic risk, although PFS improved with D-VMP vs VMP, PFS benefit was less pronounced than that observed for patients with standard cytogenetic risk.
    • Median PFS: D-VMP, 19.5 months; VMP, 18.1 months (HR, 0.58; 95% CI, 0.32-1.04; P=0.0638)
    • 12-month PFS: D-VMP, 82.9%; VMP, 69.8%
    • 18-month PFS: D-VMP, 60.6%; VMP, 52.7%
  • D-VMP was associated with PFS, ORR, and ≥VGPR benefits across clinically relevant subgroups, including patients with ISS stage III disease. See Tables: Subgroup Analysis of PFS in the Pooled ITT Population, Subgroup Analysis of ORR in the Pooled ITT Population, and Subgroup Analysis of ≥VGPR in the Pooled ITT Population.

Subgroup Analysis of PFS in the Pooled ITT Population21
Subgroup
Events/Patients
Median PFS, Months
HR (95% CI)a
D-VMP
VMP
D-VMP
VMP
ISS disease stageb
   I
19/106
21/86
NE
NE
0.60 (0.32-1.12)
   II
46/207
82/192
NE
17.5
0.43 (0.30-0.62)
   III
49/183
63/152
NE
16.7
0.52 (0.36-0.75)
Cytogenetic risk at study entryc
   High risk
28/81
24/65
19.5
18.1
0.65 (0.37-1.13)
   Standard risk
76/378
126/311
NE
17.4
0.41 (0.31-0.54)
Abbreviations: CI, confidence interval; D-VMP, DARZALEX + bortezomib + melphalan + prednisone; HR, hazard ratio; ISS, International Staging System; ITT, intent-to-treat; NE, not estimable; PFS, progression-free survival; VMP, bortezomib + melphalan + prednisone.
aHRs and 95% CIs were calculated from a Cox proportional hazards model, with treatment as the sole explanatory variable. An HR <1 indicates an advantage for D-VMP.
bBased on the combination of serum β2-microglobulin and albumin.
cCytogenetic risk was based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality. Patients with standard cytogenetic risk had an absence of high-risk cytogenetic abnormalities.


Subgroup Analysis of ORR in the Pooled ITT Population21
Subgroup
D-VMP, n (%)
VMP, n (%)
OR (95% CI)a
ISS disease stageb
   I
98 (92.5)
66 (76.7)
3.71 (1.54-8.93)
   II
187 (90.3)
145 (75.5)
3.03 (1.72-5.34)
   III
162 (88.5)
110 (72.4)
2.95 (1.65-5.25)
Cytogenetic risk at study entryc
   High risk
73 (90.1)
51 (78.5)
2.50 (0.98-6.41)
   Standard risk
342 (90.5)
224 (72)
3.69 (2.42-5.63)
Abbreviations: CI, confidence interval; D-VMP, DARZALEX + bortezomib + melphalan + prednisone; ISS, International Staging System; ITT, intent-to-treat; OR, odds ratio; ORR, overall response rate; VMP, bortezomib + melphalan + prednisone.
aORs and 95% CIs were calculated from a stratified Cochran-Mantel-Haenszel test. An OR >1 indicates an advantage for D-VMP.
bBased on the combination of serum β2-microglobulin and albumin.
cCytogenetic risk was based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality. Patients with standard cytogenetic risk had an absence of high-risk cytogenetic abnormalities.


Subgroup Analysis of ≥VGPR in the Pooled ITT Population21
Subgroup
D-VMP, n (%)
VMP, n (%)
OR (95% CI)a
ISS disease stageb
   I
75 (70.8)
46 (53.5)
2.10 (1.16-3.82)
   II
152 (73.4)
93 (48.4)
2.94 (1.94-4.47)
   III
130 (71)
70 (46.1)
2.87 (1.83-4.51)
Cytogenetic risk at study entryc
   High risk
60 (74.1)
30 (46.2)
3.33 (1.66-6.69)
   Standard risk
272 (72)
147 (47.3)
2.86 (2.09-3.93)
Abbreviations: CI, confidence interval; D-VMP, DARZALEX + bortezomib + melphalan + prednisone; ISS, International Staging System; ITT, intent-to-treat; OR, odds ratio; VGPR, very good partial response; VMP, bortezomib + melphalan + prednisone.
aORs and 95% CIs were calculated from a stratified Cochran-Mantel-Haenszel test. An OR >1 indicates an advantage for D-VMP.
bBased on the combination of serum β2-microglobulin and albumin.
cCytogenetic risk was based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality. Patients with standard cytogenetic risk had an absence of high-risk cytogenetic abnormalities.

Pooled Analysis of OCTANS and ALCYONE: PFS Outcomes by MRD and Cytogenetic Risk Status

Fu et al (2023)22 reported the subgroup analysis results of PFS by MRD status using data from the OCTANS and ALCYONE studies.

Study Design/Methods

  • The data cutoff date for the OCTANS analysis was December 23, 2022, and the data cutoff date for the ALCYONE analysis was June 24, 2019.

Results

Patient Characteristics

Demographic and Baseline Characteristics in the ITT Population22
OCTANS
ALCYONE
D-VMP
(n=146)

VMP
(n=74)

D-VMP
(n=350)

VMP
(n=356)

ISS disease stagea, n (%)
   I
37 (25.3)
19 (25.7)
69 (19.7)
67 (18.8)
   II
68 (46.6)
32 (43.2)
139 (39.7)
160 (44.9)
   III
41 (28.1)
23 (31.1)
142 (40.6)
129 (36.2)
Cytogenetic profileb
   n
145
74
314
302
   Standard risk, n (%)
117 (80.7)
54 (73)
261 (83.1)
257 (85.1)
   High risk, n (%)
28 (19.3)
20 (27)
53 (16.9)
45 (14.9)
Abbreviations: D-VMP, DARZALEX + bortezomib + melphalan + prednisone; ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; ITT, intent-to-treat; MM, multiple myeloma; VMP, bortezomib + melphalan + prednisone.
aBased on the combination of serum β2-microglobulin and albumin.
bCytogenetic risk was based on local fluorescence in situ hybridization or karyotype analysis. Patients with high-risk cytogenetics had a del17p, t(4;14), and/or t(14;16) abnormality. Patients with standard-risk cytogenetics had an absence of high-risk cytogenetic abnormalities.

Efficacy
  • In a pooled analysis of OCTANS and ALCYONE (D-VMP, n=496; VMP, n=430), D-VMP vs VMP alone was associated with prolonged PFS across most of the high-risk subgroups evaluated. See Table: PFS by High-Risk Subgroups in Pooled Data from the OCTANS and ALCYONE Studies.
    • D-VMP vs VMP alone prolonged PFS both in patients with revised standard cytogenetic risk (NE vs 18.9 months; HR, 0.33; 95% CI, 0.26-0.42) and in those with revised high cytogenetic risk (25.6 vs 18.9 months; HR, 0.54; 95% CI, 0.41-0.71).
    • Prolonged PFS was also observed for D-VMP vs VMP alone in patients with gain(1q21) and/or amp(1q21), 1 HRCA, and ≥2 HRCAs.

PFS by High-Risk Subgroups in Pooled Data from the OCTANS and ALCYONE Studies22
Subgroups
OCTANS
ALCYONE
HR
(95% CI)

n/N
Median PFS, Months
n/N
Median PFS, Months
Baseline characteristic
   Age ≥75 years
61/127
37.3
77/117
20.4
0.50 (0.35-0.70)
   ISS stage lll disease
95/183
33
113/152
18.2
0.42 (0.32-0.56)
   Renal insufficiency
102/213
40.5
130/178
19.3
0.40 (0.30-0.52)
   Extramedullary plasmacytomas
25/36
20.7
25/29
14.6
0.50 (0.28-0.88)
Cytogenetic risk
      Standard cytogenetic risk
172/378
43
222/311
19.1
0.37 (0.30-0.45)
      High cytogenetic risk
57/81
21.1
49/65
18.1
0.61 (0.41-0.89)
      Revised standard cytogenetic risk
120/291
NE
177/251
18.9
0.33 (0.26-0.42)
      Revised high cytogenetic risk
109/168
25.6
94/125
18.9
0.54 (0.41-0.71)
      Gain(1q21)
4/8
38.9
9/11
17.5
0.31 (0.08-1.18)
      Amp(1q21)
69/114
31.3
64/84
18.9
0.46 (0.32-0.65)
      Gain(1q21) or amp(1q21)
70/118
31.9
69/90
18.9
0.45 (0.32-0.63)
      1 HRCA
87/131
24.4
64/88
19.3
0.62 (0.44-0.86)
      ≥2 HRCAs
22/37
28.2
30/37
17.5
0.36 (0.20-0.63)
      Isolated gain(1q21)
1/3
NE
4/4
18.4
0.27 (0.03-2.52)
      Isolated amp(1q21)
50/83
31.6
38/53
19.4
0.53 (0.34-0.82)
      Isolated gain(1q21) or amp(1q21)
52/87
32
45/60
19.4
0.49 (0.32-0.73)
      Gain(1q21) or amp(1q21) plus
      ≥1 HRCA

18/31
28.2
24/30
15.6
0.37 (0.20-0.71)
Abbreviations: CI, confidence interval; HR, hazard ratio; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; NE, not estimable; PFS, progression-free survival.

DARZALEX in Combination with Bortezomib, Thalidomide and Dexamethasone

CASSIOPEIA (MMY3006) is an ongoing, open-label, 2-arm, multicenter, phase 3 study evaluating the safety and efficacy of D-VTd in patients with NDMM who are eligible for high dose chemotherapy and ASCT.23 Moreau et al (2021)24 reported the results from Part 2 of the CASSIOPEIA study (maintenance treatment). Results in the high-risk cytogenetic subgroup are summarized below.

Study Design/Methods

  • Part 2: Responders (patients who completed consolidation and achieved partial response or better [≥PR]) were rerandomized to 1 of 2 treatment groups:
    • Arm A: DARZALEX 16 mg/kg maintenance therapy every 8 weeks for 2 years
    • Arm B: Observation
  • Primary endpoint: Part 2: PFS after second randomization
  • Secondary endpoints: Part 2: TTP from second randomization, rates of ≥CR, MRD-negativity rates (in patients with ≥CR at a threshold of 10-5 by NGS), PFS2, ORR, and OS from second randomization

Results

Baseline Characteristics

Key Baseline Demographics and Disease Characteristics after Second Randomization (CASSIOPEIA Part 2)24
Characteristics, n (%)
DARZALEX monotherapy
(n=442)

Observation
(n=444)

ISS disease stagea
   I
189 (43)  
171 (39)  
   II
181 (41)
214 (48)
   III
72 (16)
59 (13)
Cytogenetic profilea, n/total
   Standard-risk
383/440 (87)
374/444 (84)
   High-risk
57/440 (13)
70/444 (16)
Abbreviation: ISS, International Staging System.
aPre-induction.

Efficacy
  • After a median follow-up of 35.4 months (interquartile range [IQR], 30.2-39.9), median PFS from second randomization was NR (95% CI, not evaluable) in the DARZALEX monotherapy group vs 46.7 months (95% CI, 40-NE) in the observation group (HR, 0.53; 95% CI, 0.42-0.68; P<0.0001).
    • The DARZALEX monotherapy group experienced 108 PFS events, while the observation group experienced 173 events.
    • The PFS benefit of the DARZALEX monotherapy group vs the observation group was consistent across most prespecified subgroups, except for patients with ISS stage III disease, patients with creatinine clearance ≤90 mL/min, and those who received VTd as a part of induction, ASCT, and consolidation treatment group, as presented in Table: PFS in the Prespecified ISS and Cytogenetic Risk Subgroups (CASSIOPEIA Part 2).
  • After a median follow-up of 44.5 months (IQR, 38.9-49.1), an updated analysis was performed from first randomization (Part 1 CASSIOPEIA study) of patients in the D-VTd group vs VTd group.
    • The median PFS was NR (95% CI, NE-NE) in the D-VTd group vs 51.5 months (95% CI, 46.3-NE) in the VTd group (HR, 0.58; 95% CI, 0.47-0.72; P<0.0001).
    • The median OS was NR for patients in the D-VTd group vs VTd group (41 [8%] deaths of patients in D-VTd group vs 73 [13%] deaths in VTd group; HR, 0.54; 95% CI, 0.37-0.79).

PFS in the Prespecified ISS and Cytogenetic Risk Subgroups (CASSIOPEIA Part 2)24
Hazard Ratio (95% CI)
ISS disease staging
   I
0.50 (0.32-0.78)
   II
0.56 (0.40-0.79)
   III
0.75 (0.44-1.29)
Cytogenetic risk
   High-risk
0.43 (0.25-0.73)
   Standard-risk
0.62 (0.48-0.82)
Abbreviations: CI, confidence interval; ISS, International Staging System; PFS, progression-free survival.
Safety
  • Any TEAEs occurred in 95% (n=420) of patients in the DARZALEX monotherapy group vs 89% (n=394) in the observation group.
  • Grade ≥3 TEAEs occurred in 28% (n=122) of patients in the DARZALEX monotherapy group (who received at least 1 dose) vs 24% (n=108) in the observation group.
  • Serious AEs that occurred in >1% of patients in the DARZALEX monotherapy vs observation groups were pneumonia (n=11 [3%] vs n=7 [2%]) and lung infection (n=6 [1%] vs n=7 [2%], respectively.
  • Two AEs led to death in the DARZALEX monotherapy group (septic shock and natural killer-cell lymphoblastic lymphoma, n=1 each); both were treatment-related.

Subgroup Analysis of Part 1 Of CASSIOPEIA

Sonneveld et al (2019)25 presented a subgroup analysis of high-risk patients (based on cytogenetic risk status and ISS stage) in Part 1 of the CASSIOPEIA study.

Study Design/Methods

  • Patients were stratified by ISS stage (I, II, or III), and cytogenetic risk status.
  • Cytogenetic risk was assessed by FISH or karyotyping.
  • Patients with high-risk cytogenetics had at least one high-risk abnormality centrally confirmed during screening: del17p (≥50% abnormal cells) or t(4;14) (≥30% abnormal cells).

Results

  • High-risk (del17p or t[4;14]): 15% (n=82) D-VTd vs 16% (n=86) VTd
  • ISS Stage III: 15.5% (n=84) D-VTd vs 14.9% (n=81) VTd
Efficacy
  • Median follow-up was 18.8 (range, 0-32.2) months.
  • Efficacy data from the subgroup analyses of the study are presented in Table: Post-consolidation Response and MRD Status for High-Risk Patients (CASSIOPEIA Part 1).
  • Treatment effect of D-VTd over VTd with respect to rates of sCR was not demonstrated in patients with high-risk cytogenetics:(odds ratio [OR], 0.83; 95% CI, 0.42-1.66) or patients with ISS stage III disease (OR, 1.07; 95% CI, 0.54-2.12).
  • The rate of ≥CR was 36.6% with D-VTd vs. 32.6% with VTd (OR, 1.11; 95% CI, 0.58-2.10) in the high-risk cytogenetic subgroup and 44% with D-VTd vs 33.3% with VTd (OR, 1.54; 95% CI, 0.83-2.88) in the ISS stage III subgroup.
  • MRD-negative rate was 59.8% with D-VTd vs 44.2% with VTd (OR, 1.88; 95% CI, 1.02-3.46) and 64.3% with D-VTd vs 45.7% with VTd (OR, 2.14; 95% CI, 1.15-4) in the high-risk cytogenetics and ISS stage III subgroups, respectively.
  • PFS: D-VTd reduced the risk of disease progression or death vs VTd in cytogenetic high-risk (HR, 0.67; 95% CI, 0.35-1.30) and ISS stage III (HR, 0.66; 95% CI, 0.32-1.39) subgroups.

Post-consolidation Response and MRD Status for High-Risk Patients (CASSIOPEIA Part 1)25
Parameter
ISS Disease Stage III
High-risk Cytogenetics
D-VTd (n=84)
VTd (n=81)
P value
D-VTd (n=82)
VTd (n=86)
P value
Responsea, %
   ORR
86
89
-
84
85
-
   sCR
29
27
0.8506
24
28
0.4839
   ≥CR
44
33
-
37
33
-
   CR
16
6
-
12
5
-
   VGPR
35
51
-
42
48
-
   PR
7
5
-
6
5
-
MRD-negative Status (10-5)b, %
   MRD-negative
64
46
0.0190
60
44
0.0679
   MRD-negative and ≥CR
39
25
0.0516
29
23
0.5103
Abbreviations: CR, complete response; D, DARZALEX; MRD, minimal residual disease; ORR, overall response rate; PR, partial response; sCR, stringent complete response; VGPR, very good partial response; VTd, bortezomib+ thalidomide + dexamethasone.
aP values are given only for sCR.
bEuroFlow-based multiparametric flow cytometry.

DARZALEX with Bortezomib, Lenalidomide, and Dexamethasone

Final Subgroup Analysis From GRIFFIN

GRIFFIN (MMY2004) is a 2-part, phase 2, randomized, active-controlled US study evaluating the safety and efficacy of DARZALEX in combination with VRd in patients with NDMM eligible for HDT and ASCT.26-28 Chari et al (2024)29,30 reported the final efficacy and safety analysis results of clinically relevant subgroups from the GRIFFIN study, including ISS stage III disease and cytogenetic risk (median follow-up, 49.6 months).

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.
  • This final analysis was conducted after all patients completed ≥1 year of follow-up after concluding study treatment, died, or withdrew.
  • Primary endpoint: sCR (by end of post-ASCT consolidation)
  • Secondary endpoints: MRD (10-5 via NGS), CR, ORR, ≥VGPR
  • Cytogenetic risk was assessed by FISH; high-risk was defined as the presence of del17p, t(4;14), or t(14;16)

Results

Efficacy
  • MRD-negativity (10-5) rates favored the D-VRd vs VRd arm in the patient subgroup of ISS stage III and cytogenetic risk. See Table: Final Analysis of MRD-Negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN).
  • A final analysis of sCR rates were higher for D-VRd vs VRd for most subgroups, including the patient subgroup of ISS stage III and cytogenetic risk. See Table: Final Analysis of sCR in the ISS Stage III and Cytogenetic Subgroups (GRIFFIN).
  • MRD-negativity (10-5) rates favored the D-VRd vs VRd arm in the patient subgroup of ISS stage III and cytogenetic risk among patients who achieved a best response ≥CR by the end of the study. See Table: Final Analysis of MRD-negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups with a Best Response of ≥CR (GRIFFIN).  
  • D-VRd was associated with higher rates of sustained MRD-negativity (10-5) lasting ≥12 in the patient subgroup of ISS stage III and cytogenetic risk. See Table: Final Analysis of Rates of Sustained MRD-negativity (10-5) lasting ≥12 months in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN).
    • No patients who achieved sustained MRD-negativity (10-5) lasting ≥12 months developed PD.
  • Among MRD-evaluable patients who achieved MRD-negativity (10-5) at any time, 2 (both with ≥2 HRCAs) patients in the D-VRd arm and 5 (0 HRCA, n=2; 1 HRCA, n=3) patients in the VRd arm developed PD.
    • Two patients from the D-VRd arm and 3 patients from the VRd arm, who initially achieved MRD-negativity, developed PD after they became MRD positive again. The remaining 2 patients from the VRd arm developed PD while they continued to be MRD negative; however, MRD was not evaluated around the time of PD.
  • At a median follow-up of 49.6 months, the HR point estimates for PFS among ISS stage III disease and cytogenetic risk subgroups favored the D-VRd vs VRd arm, except for patients with ≥2 HRCAs. See Table: Final Analysis of PFS in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN).
    • Median PFS was not reached for either treatment group among patients with 0 HRCA, and the PFS HR was 0.39 (95% CI, 0.10-1.51) for D-VRd vs VRd.
    • Median PFS was not reached for D-VRd and was 47.9 months for VRd among patients with 1 HRCA, and the PFS HR was 0.19 (95% CI, 0.05-0.75) for D-VRd vs VRd.
    • Median PFS was 33.9 months for D-VRd and was not reached for VRd among patients with ≥2 HRCAs, (HR, 1.65; 95% CI, 0.30-9.18).
    • PFS was not reached for D-VRd and was 47.9 months for VRd among patients with gain/amp(1q21), with or without HRCAs, and the PFS HR was 0.42 (95% CI, 0.14-1.27) for D-VRd vs VRd.
  • Within the functionally high-risk subgroup of patients with a best response of <VGPR by the end of induction, more D-VRd vs VRd patients had revised high cytogenetic risk and 1 HRCA at baseline, and similar proportions had ≥2 HRCAs.
    • Revised high risk: 48.3% (n=14/29) vs 32.6% (n=14/43), D-VRd vs VRd respectively.
    • 1 HRCA: 37.9% (n=11/29) vs 23.3% (n=10/43), D-VRd vs VRd respectively.
    • ≥2 HRCAs: 10.3% (n=3/29) vs 9.3% (n=4/43), D-VRD vs VRd respectively.
  • Among patients who did not achieve MRD-negativity (10-5)by the end of consolidation, the proportion of patients with revised high cytogenetic risk, 1 HRCA, and ≥2 HRCAs was higher for D-VRd vs VRd at baseline.
    • Revised high risk: 52.1% (n=25/48) vs 34.2% (n=26/76), D-VRd vs VRd respectively.
    • 1 HRCA: 39.6% (n=19/48) vs 25% (n=19/76), D-VRd vs VRd respectively.
    • ≥2 HRCAs: 12.5% (n=6/48) vs 9.2% (n=7/76), D-VRd vs VRd respectively.

Final Analysis of MRD-Negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN)29
Subgroup
D-VRd
n/N (%)

VRd
n/N (%)

OR (95% CI)a
ITT (overall)
67/104 (64.4)
31/103 (30.1)
4.23 (2.35-7.62)
Baseline characteristic
   ISS stage III disease
10/14 (71.4)
5/14 (35.7)
4.50 (0.91-22.15)
   Cytogenetic risk
      High cytogenetic riskb
7/16 (43.8)
4/14 (28.6)
1.94 (0.42-8.92)
      Revised high cytogenetic riskc
23/42 (54.8)
12/37 (32.4)
2.52 (1.01-6.32)
      0 HRCAc
42/56 (75)
19/60 (31.7)
6.47 (2.87-14.60)
      1 HRCAc
17/32 (53.1)
11/29 (37.9)
1.85 (0.67-5.15)
      ≥2 HRCAsc
6/10 (60)
1/8 (12.5)
10.50 (0.91-121.39)
      Gain/amp(1q21)d
21/34 (61.8)
8/28 (28.6)
4.04 (1.38-11.81)
      Gain/amp(1q21) + 1 HRCAc
6/9 (66.7)
0/6
NE (NE-NE)
      Gain/amp(1q21) isolatede
15/25 (60)
8/22 (36.4)
2.62 (0.81-8.55)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; FISH, fluorescence in situ hybridization; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; NE, not evaluable; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.
aMantel-Haenszel estimate of the common OR for unstratified tables is used. An OR >1 indicates an advantage for D-VRd.
bHigh-risk cytogenetics are defined based on FISH testing as ≥1 of the following: del17p, t(4;14), or t(14;16).
cRevised high-risk cytogenetics are defined based on FISH testing as ≥1 HRCA: del17p, t(4;14), t(14;16), t(14;20), or gain/amp(1q21) (≥3 copies of chromosome 1q21).
dPatients in this group had gain/amp(1q21) with or without other HRCAs (del17p, t[4;14], t[14;16], or t[14;20]).
ePatients with isolated gain/amp(1q21) did not have any other HRCAs.


Final Analysis of sCR in the ISS Stage III and Cytogenetic Subgroups (GRIFFIN)30
Subgroup
D-VRd, n/N (%)
VRd, n/N (%)
OR (95% CI)a
Response evaluable (overall)b
67/100 (67%)
47/98 (48%)
2.18 (1.22-3.89)
Baseline characteristic
   ISS stage III disease
9/14 (64.3)
8/13 (61.5)
1.13 (0.24-5.37)
   Cytogenetic risk
      High cytogenetic riskc
8/16 (50)
5/13 (38.5)
1.60 (0.36-7.07)
      Revised high cytogenetic riskd
23/41 (56.1)
20/36 (55.6)
1.02 (0.42-2.52)
      0 HRCAd
43/55 (78.2)
26/58 (44.8)
4.41 (1.94-10.04)
      1 HRCAd
18/31 (58.1)
17/28 (60.7)
0.90 (0.32-2.54)
      ≥2 HRCAsd
5/10 (50)
3/8 (37.5)
1.67 (0.25-11.07)
      Gain/amp(1q21)e
19/33 (57.6)
16/28 (57.1)
1.02 (0.37-2.82)
      Gain/amp(1q21) + 1 HRCAd
5/9 (55.6)
2/6 (33.3)
2.50 (0.29-21.40)
      Gain/amp(1q21) isolatedf
14/24 (58.3)
14/22 (63.6)
0.80 (0.24-2.63)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; FISH, fluorescence in situ hybridization; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; MM, multiple myeloma; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.
aMantel-Haenszel estimate of the common OR for unstratified tables is used. An OR >1 indicates an advantage for D-VRd.
bThis analysis included patients from the response evaluable population, which included all randomized patients who had measurable disease (confirmed MM diagnosis), received ≥1 dose of study treatment, and had ≥ postbaseline disease assessmentcHigh-risk cytogenetics are defined based on FISH testing as ≥1 of the following: del17p, t(4;14), or t(14;16).
dRevised high-risk cytogenetics are defined based on FISH testing as ≥1 HRCA: del17p, t(4;14), t(14;16), t(14;20), or gain/amp(1q21) (≥3 copies of chromosome 1q21).
ePatients in this group had gain/amp(1q21) with or without other HRCAs (del17p, t[4;14], t[14;16], or t[14;20]).
fPatients with isolated gain/amp(1q21) did not have any other HRCAs.


Final Analysis of MRD-negativity (10-5) Rates in the ISS Stage III and Cytogenetic Risk Subgroups with a Best Response of ≥CR (GRIFFIN)29
Subgroup
D-VRd, n/N (%)
VRd, n/N (%)
OR (95% CI)a
Patients with best response of ≥CRb
64/83 (77.1)
28/59 (47.5)
3.70 (1.77-7.72)
Baseline characteristic
   ISS stage III disease
10/13 (76.9)
4/8 (50)
3.33 (0.50-22.14)
   Cytogenetic risk
      High cytogenetic riskc
6/10 (60)
4/7 (57.1)
1.13 (0.16-7.99)
      Revised high cytogenetic riskd
21/30 (70)
12/23 (52.2)
2.14 (0.69-6.63)
      0 HRCAd
41/49 (83.7)
16/35 (45.7)
6.09 (2.22-16.68)
      1 HRCAd
16/24 (66.7)
11/20 (55)
1.64 (0.48-5.56)
      ≥2 HRCAsd
5/6 (83.3)
1/3 (33.3)
10 (0.40-250.42)
      Gain/amp(1q21)e
19/25 (76)
8/17 (47.1)
3.56 (0.95-13.37)
      Gain/amp(1q21) + 1 HRCAd
5/6 (83.3)
0/2
NE (NE-NE)
      Gain/amp(1q21) isolatedf
14/19 (73.7)
8/15 (53.3)
2.45 (0.58-10.33)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; FISH, fluorescence in situ hybridization; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; MM, multiple myeloma; MRD, minimal residual disease; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.
aMantel-Haenszel estimate of the common OR for unstratified tables is used. An OR >1 indicates an advantage for D-VRd.
bThis analysis included patients from the response evaluable population, which included all randomized patients who had measurable disease (confirmed MM diagnosis), received ≥1 dose of study treatment, and had ≥ postbaseline disease assessmentcHigh-risk cytogenetics are defined based on FISH testing as ≥1 of the following: del17p, t(4;14), or t(14;16).
dRevised high-risk cytogenetics are defined based on FISH testing as ≥1 HRCA: del17p, t(4;14), t(14;16), t(14;20), or gain/amp(1q21) (≥3 copies of chromosome 1q21).
ePatients in this group had gain/amp(1q21) with or without other HRCAs (del17p, t[4;14], t[14;16], or t[14;20]).
fPatients with isolated gain/amp(1q21) did not have any other HRCAs.


Final Analysis of Rates of Sustained MRD-negativity (10-5) lasting ≥12 months in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN)29
Subgroup
D-VRd, n/N (%)
VRd, n/N (%)
OR (95% CI)a
ITT (overall)
46/104 (44.2)
14/103(13.6)
5 (2.50-9.99)
Baseline characteristic
   Age ≥65 years
14/28 (50)
3/28 (10.7)
8.33 (2.04 (34.07)
   ISS stage III disease
6/14 (42.9)
2/14 (14.3)
4.50 (0.72-28.15)
   Cytogenetic risk
      High cytogenetic riskb
3/16 (18.8)
2/14 (14.3)
1.38 (0.20-9.77)
      Revised high cytogenetic riskc
14/42 (33.3)
6/37 (16.2)
2.58 (0.87-7.64)
      0 HRCAc
31/56 (55.4)
8/60 (13.3)
8.06 ((3.24-20.06)
      1 HRCAc
12/32 (37.5)
5/29 (17.2)
2.88 (0.87-9.56)
      ≥2 HRCAsc
2/10 (20)
1/8 (12.5)
1.75 (0.13-23.70)
      Gain/amp(1q21)d
13/34 (38.2)
4/28 (14.3)
3.71 (1.05-13.15)
      Gain/amp(1q21) + 1 HRCAc
2/9 (22.2)
0/6
NE (NE-NE)
      Gain/amp(1q21) isolatede
11/25 (44)
4/22 (18.2)
3.54 (0.93-13.51)
Abbreviations: CI, confidence interval; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; FISH, fluorescence in situ hybridization; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; NE, not evaluable; OR, odds ratio; VRd, bortezomib + lenalidomide + dexamethasone.
aMantel-Haenszel estimate of the common OR for unstratified tables is used. An OR >1 indicates an advantage for D-VRd.
bHigh-risk cytogenetics are defined based on FISH testing as ≥1 of the following: del17p, t(4;14), or t(14;16).
cRevised high-risk cytogenetics are defined based on FISH testing as ≥1 HRCA: del17p, t(4;14), t(14;16), t(14;20), or gain/amp(1q21) (≥3 copies of chromosome 1q21).
dPatients in this group had gain/amp(1q21) with or without other HRCAs (del17p, t[4;14], t[14;16], or t[14;20]).
ePatients with isolated gain/amp(1q21) did not have any other HRCAs.


Final Analysis of PFS in the ISS Stage III and Cytogenetic Risk Subgroups (GRIFFIN)29
Subgroup
D-VRd
VRd
HR (95% CI)a
n/N
Median PFS, Months
n/N
Median PFS, Months
ITT (overall)
11/104
NR
18/103
NR
0.45 (0.21-0.95)
Baseline characteristic
   ISS stage III disease
2/14
NR
6/14
33.1
0.23 (0.05-1.13)
   Cytogenetic risk
      High cytogenetic riskb
5/16
NR
5/14
36.1
0.54 (0.15-1.88)
      Revised high cytogenetic riskc
7/42
NR
10/37
47.9
0.38 (0.14-1.01)
      0 HRCAc
3/56
NR
7/60
NR
0.39 (0.10-1.51)
      1 HRCAc
3/32
NR
8/29
47.9
0.19 (0.05-0.75)
      ≥2 HRCAsc
4/10
33.9
2/8
NR
1.65 (0.30-9.18)
      Gain/amp(1q21)d
6/34
NR
7/28
47.9
0.42 (0.14-1.27)
      Gain/amp(1q21) + 1 HRCAc
4/9
33.9
2/6
38.7
0.81 (0.15-4.47)
      Gain/amp(1q21) isolatede
2/25
NR
5/22
47.9
0.21 (0.04-1.09)
Abbreviations: CI, confidence interval; CrCl, creatinine clearance; D-VRd, DARZALEX + bortezomib + lenalidomide + dexamethasone; FISH, fluorescence in situ hybridization; HR, hazard ratio; HRCA, high-risk cytogenetic abnormality; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; NR, not reached; PFS, progression-free survival; VRd, bortezomib + lenalidomide + dexamethasone.
aHR and 95% CI are from a Cox proportional hazards model with treatment as the sole explanatory variable. A HR <1 indicates an advantage for D-VRd.
bHigh-risk cytogenetics are defined based on FISH testing as ≥1 of the following: del17p, t(4;14), or t(14;16).
cRevised high-risk cytogenetics are defined based on FISH testing as ≥1 HRCA: del17p, t(4;14), t(14;16), t(14;20), or gain/amp(1q21) (≥3 copies of chromosome 1q21).
dPatients in this group had gain/amp(1q21) with or without other HRCAs (del17p, t[4;14], t[14;16], or t[14;20]).
ePatients with isolated gain/amp(1q21) did not have any other HRCAs.

Safety
  • A summary of the most common (>30%) TEAEs in the safety analysis population separated by age <65 years and ≥65 years is provided in Table: Most Common (>30%) and grade TEAEs by Age (<60 years and ≥65 years).
  • Among patients aged <65 years (84.7% vs 80%) and ≥65 years (88.9% vs 77.8%), the rates of grade 3/4 TEAEs were slightly higher for D-VRd vs VRd.
  • The incidence of serious TEAEs was lower in D-VRd vs VRd among patients <65 years, 41.7% vs 56% respectively.
  • The incidence of serious TEAEs was higher in the D-VRd vs VRd among patients ≥65 years, 59.3% vs 40.7% respectively.
  • The discontinuation of ≥1 therapeutic agent due to TEAEs were comparable between patients aged <65 years (D-VRd, 31.9% vs VRd, 33.3%) and higher for D-VRd vs VRd in patients ≥65 years (D-VRd, 37% vs 25.9%).
    • The most common TEAE leading to discontinuation of ≥1 drug was peripheral neuropathy for patients <65 years (D-VRd, 11.1% vs VRd, 13.3%) and ≥65 years (D-VRd, 18.5% vs VRd, 11.1%).
  • The incidence of TEAEs leading to lenalidomide dose reduction was higher for D-VRd vs VRd (<65 years: D-VRd, 33.3% vs VRd, 28%; ≥65 years: D-VRd, 59.3% vs VRd, 33.3%) among patients aged <65 and ≥65 years.
    • The most common TEAE leading to lenalidomide dose reduction was neutropenia for patients <65 years (D-VRd, 15.3% vs VRd, 5.3%) and ≥65 years (D-VRD, 22.2% vs VRd, 14.8%).
  • Two deaths due to a TEAE occurred and were considered unrelated to study treatment.
    • <65 years (VRd, n=1; cause unknown)
    • ≥65 years (D-VRd, n=1; pneumonia)

Most Common (>30%)a any grade TEAEs by Age (<65 years and ≥65 years)29
Most common TEAEs, n (%)
<65 years
≥65 years
D-VRd
(n=72)

VRd
(n=75)

D-VRd
(n=27)

VRd
(n=27)

Hematologic
   Neutropenia
47 (65.3)
29 (38.7)
16 (59.3)
12 (44.4)
   Thrombocytopenia
30 (41.7)
24 (32)
14 (51.9)
12 (44.4)
   Leukopenia
29 (40.3)
21 (28)
10 (37)
9 (33.3)
   Anemia
25 (34.7)
25 (33.3)
12 (44.4)
8 (29.6)
   Lymphopenia
23 (31.9)
23 (30.7)
8 (29.6)
6 (22.2)
Nonhematologic
   Upper respiratory tract
   infection

51 (70.8)
37 (49.3)
16 (59.3)
14 (51.9)
Diarrhea
48 (66.7)
39 (52)
18 (66.7)
17 (63)
Fatigue
48 (66.7)
45 (60)
23 (85.2)
18 (66.7)
Peripheral neuropathyb
41 (56.9)
56 (74.7)
21 (77.8)
22 (81.5)
Nausea
38 (52.8)
37 (49.3)
14 (51.9)
14 (51.9)
Constipation
37 (51.4)
29 (38.7)
14 (51.9)
13 (48.1)
Insomnia
36 (50)
25 (33.3)
9 (33.3)
6 (22.2)
Cough
35 (48.6)
26 (34.7)
18 (66.7
5 (18.5)
Pyrexia
34 (47.2)
27 (36)
14 (51.9)
6 (22.2)
Back pain
30 (41.7)
29 (38.7)
11 (40.7)
7 (25.9)
Arthralgia
27 (37.5)
26 (34.7)
12 (44.4)
12 (44.4)
Headache
27 (37.5)
18 (24)
6 (22.2)
6 (22.2)
Muscle spasms
26 (36.1)
11 (14.7)
4 (14.8)
9 (33.3)
Vomiting
25 (34.7)
21 (28)
7 (25.9)
8 (29.6)
Peripheral edema
24 (33.3)
25 (33.3)
12 (44.4)
12 (44.4)
Hypokalemia
19 (26.4)
20 (26.7)
9 (33.3)
7 (25.9)
Pain in extremity
19 (26.4)
13 (17.3)
3 (11.1)
9 (33.3)
Dyspnea
14 (19.4)
24 (32)
10 (37)
7 (25.9)
Dizziness
15 (20.8)
16 (21.3)
8 (29.6)
9 (33.3)
Pneumonia
14 (19.4)
16 (21.3)
10 (37)
2 (7.4)
Dysgeusia
14 (19.4)
14 (18.7)
9 (33.3)
5 (18.5)
Abbreviations: D-VRd, daratumumab + lenalidomide + bortezomib + dexamethasone; TEAE, treatment-emergent adverse event; VRd, lenalidomide + bortezomib + dexamethasone.aIncludes TEAEs occurring in ≥30% of patients aged <65 years or ≥65 years in either treatment group from the safety analysis population (all randomized patients who received ≥1 dose of study treatment).bIncludes preferred terms neuropathy peripheral and peripheral sensory neuropathy.

DARZALEX in Combination with KRd

MASTER (ClinicalTrials.gov identifier: NCT03224507) evaluated the efficacy and safety of D-KRd induction followed by AHCT and MRD-adapted consolidation therapy in patients with NDMM.31 Costa et al (2023)31 reported the results from the final analysis of the MASTER study at a median follow-up of 42.2 months. Results from the final analysis are reported below.

Study Design/Methods

  • Key eligibility criteria: Patients of any age group with NDMM with measurable disease; with Eastern Cooperative Oncology Group performance status (ECOG PS) of ≤2; who had measurable renal function (creatinine clearance ≥40 mL/min); and who were either untreated or had received up to 1 cycle of bortezomib, cyclophosphamide, and dexamethasone (VCd) were included. Patients with a concomitant or recent malignancy, or significant cardiopulmonary disease were excluded.
  • Primary endpoint: Rate of MRD-negative responses (<10-5) by NGS (clonoSEQ®)
  • Secondary endpoints: Toxicity of D-KRd, rates and kinetics of MRD resurgence upon treatment discontinuation, PFS, and OS
  • Exploratory endpoints: PFS and OS for patients who transitioned to MRD-surveillance (MRD-SURE) and were monitored off therapy
  • Enrichment for patients with HRCA was planned during recruitment.

Results

Patient Characteristics
  • A total of 123 patients were enrolled, of whom 118 (96%) patients with MRD were evaluable. See Table: Patient Demographics and Baseline Characteristics (MASTER).
  • The median duration of follow-up was 42.2 months (IQR, 34.5-46) overall (0 HRCA, 43.7 months [IQR, 36.3-47.4]; 1 HRCA, 42.1 months [IQR, 32.9-45.8]; ≥2 HRCAs, 35.4 months [IQR, 26.1-43.4]).
  • The median duration of therapy was 11.6 months (IQR, 8-14.8) overall and 11.2 months (IQR, 8.412.4) for those who entered MRD-SURE.

Patient Demographics and Baseline Characteristics (MASTER)31
Characteristic
0 HRCA
(n=53)

1 HRCA
(n=46)

≥2 HRCAs
(n=24)

Total
(N=123)

Sex, n (%)
   Men
33 (62)
24 (52)
13 (54)
70 (57)
   Women
20 (38)
22 (48)
11 (46)
53 (43)
Age, years
   Median (IQR)
60 (50-69)
61 (57-68)
60 (56-66)
61 (55-68)
   ≥70 years, n (%)
12 (23)
10 (22)
2 (8)
24 (20)
Racial/ethnicity, n (%)
   Non-Hispanic White
42 (79)
33 (72)
19 (79)
94 (76)
   Non-Hispanic Black
10 (19)
11 (24)
4 (17)
25 (20)
   Other
1 (2)
2 (4)
1 (4)
4 (3)
ECOG PS, n (%)
   0-1
42 (79)
40 (87)
17 (71)
99 (80)
   2
11 (21)
6 (13)
7 (29)
24 (20)
LDH concentration, n (%)
   <240 U/L
45 (85)
34 (74)
18 (75)
97 (79)
   ≥240 U/L
8 (15)
12 (26)
6 (25)
26 (21)
β₂ microglobulin concentration, n (%)
   <3.5 g/L
36 (67)
22 (48)
7 (29)
65 (53)
   ≥3.5 to <5.5 g/L
12 (23)
12 (26)
6 (25)
29 (24)
   ≥5.5 g/L
5 (9)
12 (26)
11 (46)
29 (24)
Albumin concentration, n (%)
   <3.5 g/dL
15 (28)
20 (44)
12 (50)
47 (38)
   ≥3.5 g/dL
38 (72)
26 (57)
12 (50)
66 (54)
Cytogenetic abnormality, n (%)
   Hyperdiploidy
27 (51)
20 (44)
4 (17)
51 (41)
   del(13q)
19 (36)
20 (44)
18 (75)
57 (46)
   Gain/amp 1q
0 (0)
24 (52)
20 (83)
44 (36)
   del(1p)
3 (6)
4 (9)
5 (21)
12 (10)
   t(11;14)
14 (26)
7 (15)
0 (0)
21 (17)
   t(4;14)
0 (0)
8 (17)
13 (54)
21 (17)
   t(14;16)
0 (0)
2 (4)
4 (17)
6 (5)
   del(17p)
0 (0)
12 (26)
14 (58)
26 (21)
ISS, n (%)
   I
28 (53)
15 (33)
5 (21)
48 (39)
   II
20 (38)
19 (41)
8 (33)
46 (37)
   III
5 (9.4)
12 (26)
11 (46)
29 (24)
R-ISS, n (%)
   I
25 (47)
11 (24)
0 (0)
35 (28)
   II
27 (51)
23 (50)
13 (54)
63 (51)
   III
1 (2)
12 (26)
11 (46)
25 (20)
Multiple myeloma clinical manifestation, n (%)
   Bone disease
43 (81)
29 (63)
18 (75)
90 (73)
   Renal failure
2 (4)
8 (17)
6 (25)
16 (13)
   Anemia
22 (42)
30 (65)
20 (83)
72 (59)
   Hypercalcemia
8 (15)
7 (15)
7 (29)
22 (18)
Ig isotype, n (%)
   IgG
27 (51)
26 (57)
14 (58)
67 (54)
   IgA
12 (23)
15 (33)
8 (33)
35 (28)
   Light chain
14 (26)
5 (11)
2 (8)
21 (17)
Therapy before enrollment,a n (%)
   Yes
26 (49)
25 (54)
10 (42)
61 (50)
   No
27 (51)
21 (46)
14 (58)
62 (50)
MRD trackable by NGS (clonoSEQ®), n (%)
   Yes
50 (94)
44 (96)
24 (100)
118 (96)
   No
3 (6)
2 (4)
0 (0)
5 (4)
Abbreviations: 0 HRCA, standard risk cytogenetic abnormalities; 1 HRCA, high-risk cytogenetic abnormalities; ≥2 HRCAs, ultra high-risk cytogenetic abnormalities; ECOG PS, Eastern Cooperative Oncology Group performance status; Ig, immunoglobulin; IQR, interquartile range; ISS, International Staging System; LDH, lactate dehydrogenase; MRD, minimal residual disease; NGS, next-generation sequencing; R-ISS, Revised International Staging System. aOne cycle of bortezomib with or without cyclophosphamide and with or without dexamethasone was allowed according to the protocol.
Efficacy
  • A total of 96 patients (81%; 95% CI, 73-88) reached MRD <10-5 at any point during treatment. See Table: MRD-Negative Response Rates (MASTER).
  • Of the 118 MRD-evaluable patients, 84 (71%) entered protocol-directed observation and MRD-SURE, 24 (20%) proceeded to lenalidomide maintenance, and 10 (8%) discontinued treatment (died, n=3; discontinued due to disease progression, n=5; patient’s choice, n=2).

MRD-Negative Response Rates (MASTER)a,31
0 HRCA
(n=50)

1 HRCA
(n=44)

≥2 HRCAs
(n=24)

Total
(N=118)

Rate of NGS MRD <10-5 (primary endpoint)
   At any point in treatment, n (%)
39 (78)
38 (86)
19 (79)
96 (81)
      95% CI
64-88
73-95
58-93
73-88
Rate of NGS MRD <10-6 (post hoc exploratory endpoint)
   At any point in treatment, n (%)
34 (68)
35 (80)
15 (63)
84 (71)
      95% CI
53-80
65-90
41-81
62-79
CR + MRD <10-5, n (%)
38 (76)
33 (75)
14 (58)
85 (72)
   95% CI
62-87
60-87
37-78
63-80
MRD <10-5 at 2 consecutive assessments and transitioned to MRD-SURE, n (%)
33 (66)
36 (82)
15 (63)
84 (71)
   95% CI
51-66
67-92
41-81
62-79
Sustained MRD <105, n (%)
32 (64)
32 (73)
11 (46)
-
   95% CI
49-77
57-85
26-67
-
Abbreviations: 0 HRCA, standard risk cytogenetic abnormalities; 1 HRCA, high-risk cytogenetic abnormalities; ≥2 HRCAs, ultra high-risk cytogenetic abnormalities; CI, confidence interval; CR, complete response; MRD, minimal residual disease; MRD-SURE, MRD-surveillance; NGS, next-generation sequencing.
aHRCA included gain/amp 1q, t(4;14), t(14;16), t(14;20), or del(17p).

  • A total of 33 PFS events were recorded among 123 included patients and 32 PFS events were recorded among 118 MRD-evaluable patients. See Table: Final Analysis Survival Outcomes (MASTER).
  • A total of 6 patients had disease progression while receiving protocol-directed therapy (induction phase, n=1; after AHCT and before consolidation, n=1; consolidation, n=4).
    • All 6 patients had gain/amp 1q, 5 had del(17p), and 5 had ≥2 HRCAs; all patients died from disease progression/complications of subsequent therapy within 1.3-10.8 months after initial progression.
  • Of the 118 MRD-evaluable patients, the 3-year PFS rate for those who reached sustained MRD-negativity (n=75 [64%]) vs those who did not reach sustained MRD-negativity (n=43 [36%]) was 89% (95% CI, 82-94) vs 55% (95% CI, 39-69), respectively. See Table: PFS and OS for Different Study Populations (MASTERS).
  • Of the 106 patients without disease progression at 18 months from enrollment, the 3year PFS rate for those who reached sustained MRD-negativity vs those who did not reach sustained MRD-negativity was 89% (n=75; 95% CI, 82-94) vs 70% (n=31; 95% CI, 5483), respectively.
  • At the last MRD-SURE observation timepoint, 61 patients (MRD-evaluable patients, 52%; transitioned to MRDSURE, 73%) remained free of therapy with sustained MRDnegativity, including 3 patients who died of unrelated causes without previous disease progression. See Table: Landmark PFS and OS According to MRD Status at Different Timepoints (MASTER).
  • Of the 84 patients who transitioned to MRD-SURE, 23 (27%) resumed therapy, of whom 16 resumed due to disease progression without MRD resurgence and 7 due to MRD resurgence without progression.

Final Analysis Survival Outcomes (MASTER)31
Characteristic
0 HRCA
(n=53)

1 HRCA
(n=46)

≥2 HRCAs
(n=24)

3-year PFS rate (N=123), %
88
79
50
   95% CI
78-95
67-88
30-70
3-year OS rate (N=123), %
94
92
75
   95% CI
88-98
86-96
63-85
3-year PFS rate for MRD-evaluable patients (n=118), %
88
80
50
   95% CI
78-94
68-90
30-70
3-year OS rate for MRD-evaluable patients (n=118), %
94
94
75
   95% CI
88-98
87-99
63-85
3-year PFS rate for patients reaching MRD-SURE (n=84), %
88
85
60
   95% CI
77-96
73-96
35-82
3-year OS rate for patients reaching MRD-SURE (n=84), %
97
93
100
   95% CI
91-100
84-100
NC-100
Cumulative incidence of progression rates for patients reaching MRD-SURE (n=84), %
9
9
47
   95% CI
1-19
1-18
23-72
2-year cumulative incidences of disease progression or MRD resurgence rates for patients reaching MRD-SURE (n=84), %
9
14
60
   95% CI
1-19
4-26
35-81
2-year PFS rate after cessation of therapy, %
88
85
53
   95% CI
77-95
73-94
28-78
2-year OS rate after cessation of therapy, %
97
93
100
   95% CI
91-100
85-99
NC-100
Abbreviations: 0 HRCA, standard risk cytogenetic abnormalities; 1 HRCA, high-risk cytogenetic abnormalities; ≥2 HRCAs, ultra high-risk cytogenetic abnormalities; CI, confidence interval; MRD, minimal residual disease; MRD-SURE, MRD-surveillance; NC, not calculated; OS, overall survival; PFS, progression-free survival.

PFS and OS for Different Study Populations (MASTER)31,32
Characteristic
1 HRCA vs 0 HRCA
≥2 HRCAs vs 0 HRCA
PFS for entire population
   HR (95% CI)
2.27 (0.91-5.68)
6.29 (2.49-15.89)
P value
0.81
<0.0001
OS for entire population
   HR (95% CI)
1.22 (0.30-4.88)
5.36 (1.53-18.75)
P value
0.78
0.0085
PFS for MRD-evaluable patients
   HR (95% CI)
2.03 (0.80-5.16)
5.98 (2.37-15.09)
P value
0.14
<0.0001
OS for MRD-evaluable patients
   HR (95% CI)
0.91 (0.20-4.08)
5.12 (1.46-17.97)
P value
0.90
0.011
PFS for MRD-SURE
   HR (95% CI)
1.84 (0.62-5.51)
4.37 (1.38-13.82)
P value
0.27
0.012
OS for MRD-SURE
   HR (95% CI)
1.24 (0.17-8.87)
1.74 (0.15-20.16)
P value
0.83
0.66
Abbreviations: 0 HRCA, standard risk cytogenetic abnormalities; 1 HRCA, high-risk cytogenetic abnormalities; ≥2 HRCAs, ultra high-risk cytogenetic abnormalities; CI, confidence interval; HR, hazard ratio; MRD, minimal residual disease; MRD-SURE, MRD-surveillance; OS, overall survival; PFS, progression-free survival.

Landmark PFS and OS According to MRD Status at Different Timepoints (MASTER)32
Characteristic
Landmark PFS according to MRD status (<10-5) at end of induction
HR (95% CI)
1.06 (0.50-2.12)
   Log-rank P value
0.94
Landmark OS according to MRD status (<10-5) at end of induction
   HR (95% CI)
0.33 (0.07-1.49)
   Log-rank P value
0.11
Landmark PFS according to MRD status (<10-5) post-AHCT
   HR (95% CI)
0.95 (0.43-2.08)
   Log-rank P value
0.90
Landmark OS according to MRD status (<10-5) post-AHCT
   HR (95% CI)
0.63 (0.28-2.27)
   Log-rank P value
0.48
Exploratory landmark PFS according to MRD status (<10-6) at end of induction
   HR (95% CI)
0.85 (0.37-2)
   Log-rank P value
0.72
Exploratory landmark OS according to MRD status (<10-6) at end of induction
   HR (95% CI)
0.28 (0.04-2.17)
   Log-rank P value
0.15
Exploratory landmark PFS according to MRD status (<10-6) post-AHCT
   HR (95% CI)
1 (0.48-2.10)
   Log-rank P value
0.99
Exploratory landmark OS according to MRD status (<10-6) post-AHCT
   HR (95% CI)
0.26 (0.05-1.22)
   Log-rank P value
0.065
Abbreviations: AHCT, autologous hematopoietic cell transplantation; CI, confidence interval; HR, hazard ratio; MRD, minimal residual disease; OS, overall survival; PFS, progression free survival.
Safety
  • All patients had ≥1 TEAE.
  • DARZALEX doses were neither reduced nor discontinued in any patient due to toxicity.
    • Carfilzomib and lenalidomide was discontinued in 2 patients each due to toxicity.
  • The most common TEAEs reported in the MASTER study are presented in Table: Most Common TEAEs (MASTER).
  • The most common serious TEAEs were pneumonia (n=8) and thromboembolic events (n=3).
  • No patient developed a secondary malignancy due to protocol-directed therapy.
  • A total of 15 patients died among 123 included patients.
    • Three patients died during protocol-directed therapy (not judged to be treatment related), of whom 2 had sudden death and 1 died from metapneumovirus pneumonia.
    • Three patients died after therapy and while on MRD-SURE without disease progression due to sudden death, COVID-19 pneumonia, and an accidental fall (n=1 each).
    • Nine patients died from progression of MM while receiving protocoldirected therapy (n=6) and after the completion of therapy (n=3).
  • A total of 4 patients died among 118 MRD-evaluable patients.

Most Common TEAEs (MASTER)31
Event, n (%)
Grade 1/2
Grade 3
Grade 4
Grade 5
All events
123 (100)
69 (56)
22 (18)
3 (2)
Hematologic
   Neutropenia
8 (7)
36 (29)
7 (6)
0 (0)
   Lymphopenia
6 (5)
18 (15)
10 (8)
0 (0)
   Anemia
13 (11)
11 (9)
2 (2)
0 (0)
   Thrombocytopenia
11 (9)
9 (7)
3 (2)
0 (0)
   Leukopenia
10 (8)
6 (5)
6 (5)
0 (0)
Non-hematologic
   Fatigue
58 (47)
11 (9)
0 (0)
0 (0)
   Bone pain
61 (50)
7 (6)
0 (0)
0 (0)
   Maculopapular rash
45 (37)
5 (4)
0 (0)
0 (0)
   Nausea
49 (40)
0 (0)
0 (0)
0 (0)
   Constipation
48 (39)
0 (0)
0 (0)
0 (0)
   Upper respiratory tract infection
44 (36)
1 (1)
0 (0)
0 (0)
   Diarrhea
38 (31)
5 (4)
0 (0)
0 (0)
   Insomnia
32 (26)
3 (2)
0 (0)
0 (0)
   Dyspnea
32 (26)
2 (2)
0 (0)
0 (0)
   Cough
33 (27)
0 (0)
0 (0)
0 (0)
   Hypertension
19 (15)
13 (11)
0 (0)
0 (0)
   Dizziness
29 (24)
1 (1)
0 (0)
0 (0)
   Peripheral sensory neuropathy
24 (20)
2 (2)
0 (0)
0 (0)
   Dysgeusia
25 (20)
0 (0)
0 (0)
0 (0)
   Hyperglycemia
18 (15)
5 (4)
1 (1)
0 (0)
   Headache
22 (18)
2 (2)
0 (0)
0 (0)
   Fever
23 (19)
0 (0)
0 (0)
0 (0)
   Edema in limbs
21 (17)
1 (1)
0 (0)
0 (0)
   Increased ALT concentration
19 (15)
2 (2)
0 (0)
0 (0)
   Weight loss
17 (14)
1 (1)
0 (0)
0 (0)
   Hypophosphatemia
7 (6)
9 (7)
0 (0)
0 (0)
   Weight gain
13 (11)
1 (1)
0 (0)
0 (0)
   Increased ASP concentration
12 (10)
1 (1)
0 (0)
0 (0)
   Hypocalcemia
11 (9)
1 (1)
1 (1)
0 (0)
   Thromboembolic event
8 (7)
3 (2)
2 (2)
0 (0)
   Lung infection
4 (3)
3 (2)
2 (2)
1 (1)
   Acute kidney injury
8 (7)
1 (1)
0 (0)
0 (0)
   Sudden death
0 (0)
0 (0)
0 (0)
2 (2)
   Hemolytic uremic syndrome
0 (0)
0 (0)
1 (1)
0 (0)
   Heart failure
1 (1)
0 (0)
0 (0)
0 (0)
IRR
32 (26)
2 (2)
0 (0)
0 (0)
Abbreviations: AE, adverse event; ALT, alanine transaminase; ASP, aspartate aminotransferase; IRR, infusion-related reaction; TEAE, treatment-emergent adverse event.

Analysis of Transplant-Eligible Patients with HRCAs from MASTER and GRIFFIN Studies

Callander et al (2024)33 reported the results of a post hoc analysis evaluating the clinical efficacy of the DARZALEX-based quadruplet therapies D-KRd and D-VRd in transplant-eligible patients with NDMM and HRCAs from the MASTER (median follow-up, 31.1 months) and GRIFFIN (median follow-up, 49.6 months) studies, respectively.

Study Design/Methods

  • Patients with HRCAs included ≥1 genetic abnormality: del17p, t(4;14), t(14;16), t(14;20), and/or gain/amp(1q21) (≥3 copies of chromosome 1q21).

Results

Patient Characteristics
  • Among patients receiving DARZALEX-based treatment in the MASTER (n=123) study, a total of 53 (431.%), 46 (37.4%) and 24 (19.5%) patients, respectively, had 0 HRCA, 1 HRCA and 2+ HRCA cytogenetic risk.
  • Among patients receiving DARZALEX-based treatment in the GRIFFIN (N=120 [randomized phase, n=104; safety run-in, n=16]) study, a total of 67 (55.8%), 34 (28.3%) and 13 (10.8%) patients, respectively, had 0 HRCA, 1 HRCA and 2+ HRCA cytogenetic risk.
  • Baseline patient and disease characteristics from the GRIFFIN and MASTER studies are summarized in Table: Baseline Patient and Disease Characteristics (MASTER and GRIFFIN).

Baseline Patient and Disease Characteristics (MASTER and GRIFFIN)33
Characteristic
MASTER
GRIFFINa
0 HRCA
(n=53)

1 HRCA
(n=46)

≥2 HRCAs
(n=24)

Total
(n=123)

0 HRCA
(n=67)

1 HRCA
(n=34)

≥2 HRCAs
(n=13)

Total
(n=123)

Median age (range), years
60
(36-79)

61
(35-77)

60
(41-72)

60
(35-79)

59
(34-70)

59.5
(29-70)

62
(49-70)

60
(29-70)

Sex, n (%)
   Male
33 (62.3)
24 (52.2)
13 (54.2)
70 (56.9)
37 (55.2)
18 (52.9)
9 (69.2)
64 (56.1)
   Female
20 (37.7)
22 (47.8)
11 (45.8)
53 (43.1)
30 (44.8)
16 (47.1)
4 (30.8)
50 (43.9)
ISS stageb, n (%)
   I
28 (52.8)
15 (32.6)
5 (20.8)
48 (39)
42 (62.7)
13 (38.2)
5 (38.5)
60 (52.6)
   II
20 (37.7)
19 (41.3)
8 (33.3)
46 (37.4)
20 (29.9)
17 (50)
4 (30.8)
41 (36)
   III
5 (9.4)
12 (26.1)
11 (45.8)
29 (23.6)
5 (7.5)
4 (11.8)
4 (30.8)
13 (11.4)
Cytogenetic abnormalityc, n (%)
   del(17p)
0 (0)
12 (26.1)
14 (58.3)
26 (21.1)
0 (0)
4 (11.8)
8 (61.5)
12 (10.5)
   t(4;14)
0 (0)
8 (17.4)
13 (54.2)
21 (17.1)
0 (0)
3 (8.8)
5 (38.5)
8 (7)
   t(14;16)
0 (0)
2 (4.3)
4 (16.7)
6 (4.9)
0 (0)
0 (0)
1 (7.7)
1 (0.9)
   Gain/amp(1q21)
0 (0)
24 (52.2)
20 (83.3)
44 (35.8)
0 (0)
26 (76.5)
12 (92.3)
38 (33.3)
   t(14;20)
0 (0)
0 (0)
0 (0)
0 (0)
0 (0)
1 (2.9)
0 (0)
1 (0.9)
Median duration of study treatmentd, months
        Induction/consolidatione
11.5
11.5
11.7
11.5
8.1
8.1
7.4
8.1
   Maintenance
-
-
-
-
24.4
24.2
23.9
24.2
Abbreviation: ASCT, allogeneic stem cell transplant; FISH, fluorescence in situ hybridization; HRCA, high-risk cytogenetic abnormalities; ISS, International Staging System.
aFor GRIFFIN, the D-VRd group included patients from the randomized phase (n=104) and the safety run-in phase (n=16). Patients were grouped by HRCA: 0 HRCA (n=67), 1 HRCA (n=34), or ≥2 HRCAs (n=13). Six patients were not evaluable for cytogenetic abnormalities.
b
ISS 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 (local testing).
d Study duration is reported for evaluable patients for induction/consolidation (0 HRCA, n=66; 1 HRCA, n=32; ≥2 HRCAs, n=13; total, n=111) and maintenance (0 HRCA, n=62; 1 HRCA, n=29; ≥2 HRCAs, n=10; total, n=101).
eDuration of study treatment is from initiation of therapy to completion of consolidation therapy, including ASCT.

Efficacy

Efficacy Outcomes by Cytogenetic Risk Status (MASTER)a,33
Parameter
Standard-Risk
0 HRCA
(n=53)

High-Risk
1 HRCA
(n=46)

Ultra High-Risk
≥2 HRCAs
(n=24)

≥CRb, %
90.6
89.1
70.8
24-month PFS rate, %
92.4
95.7
65.5
36-month PFS rate, %
89.9
86.2
52.4
MRD negativity
   Evaluable population, n
50c
44c
24c
      10-5 sensitivity, %
80
86.4
83.3
      10-6 sensitivity, %
68
79.5
66.7
   In patients achieving ≥CR, n
45
39
17
      10-5 sensitivity, %
84.4
89.7
94.1
Durable MRD-negativity lasting ≥12 months
   Evaluable population, n
50c
44c
24c
      10-5 sensitivity, %
64
72.7
50
MRD (10-5) conversion rate
   Evaluable population, n
-
-
-
      MRD-positive by the end of induction and then
      became MRD-negative, %
NA
NA
NA
      MRD-positive by the end of consolidation and
      then became MRD-negative, %
NA
NA
NA
   Median time to MRD-negativity (10-5)c, months
7.5
7.1
7.6
Abbreviations: ≥CR, complete response or better; HRCA, high-risk cytogenetic abnormalities; MRD, minimal residual disease; NA, not available; PFS, progression-free survival.
aHRCAs include any of the following genetic abnormalities: del(17p), t(4;14), t(14;16), t(14;20), and gain/amp(1q21) (≥3 copies of chromosome 1q21). Patients were grouped into categories: 0 HRCA, 1 HRCA, or ≥2 HRCAs.
bEvaluable patients in MASTER included all enrolled patients (0 HRCA, n=53; 1 HRCA, n=46; ≥2 HRCAs, n=24).
c
For MASTER, data are for all enrolled patients with available MRD data.


Efficacy Outcomes by Cytogenetic Risk Status(GRIFFIN)a,33
Parameter
Standard-Risk
0 HRCA
(n=67)

High-Risk
1 HRCA
(n=34)

Ultra High-Risk
≥2 HRCAs
(n=13)

≥CRb, %
90.9
78.8
61.5
24-month PFS rate, %
96.7
93.8
64.2
36-month PFS rate, %
96.7
90.5
53.5
48-month PFS rate, %
93.7
90.5
53.5
MRD negativity
   Evaluable population, n
67c
34c
13c
      10-5 sensitivity, %
76.1
55.9
61.5
      10-6 sensitivity, %
44.8
26.5
15.4
   In patients achieving ≥CR, n
60
26
8
      10-5 sensitivity, %
83.3
69.2
87.5
Durable MRD-negativity lasting ≥12 months
   Evaluable population, n
67c
34c
13c
      10-5 sensitivity, %
53.7
38.2
30.8
MRD (10-5) conversion rate
   Evaluable population, n
67c
34c
13c
      MRD-positive by the end of induction and
      then became MRD-negative, %

49.3
41.2
38.5
      MRD-positive by the end of consolidation
      and then became MRD-negative, %

19.4
11.8
23.1
   Median time to MRD-negativity (10-5)c,months
8.5
8.6
19.6
Abbreviations: ≥CR, complete response or better; HRCA, high-risk cytogenetic abnormalities; MRD, minimal residual disease; PFS, progression-free survival.
aHRCAs include any of the following genetic abnormalities: del(17p), t(4;14), t(14;16), t(14;20), and gain/amp(1q21) (≥3 copies of chromosome 1q21). Patients were grouped into categories: 0 HRCA, 1 HRCA, or ≥2 HRCAs.
bEvaluable patients in GRIFFIN were the response-evaluable population (0 HRCA, n=66; 1 HRCA, n=33; ≥2 HRCAs, n=13).
cFor GRIFFIN, the D-VRd group included patients from the randomized phase (n=104) and safety run-in phase (n=16). Patients were grouped by HRCA: 0 HRCA (n=67), 1 HRCA (n=34), or ≥2 HRCAs (n=13). Six patients were not evaluable for cytogenetic abnormalities.

Safety
  • In MASTER, 3 patients died while on D-KRd therapy (0 HRCA, n=2 [both sudden death]; ≥2 HRCAs, n=1 [metapneumovirus infection during transplant]).
    • Two patients died after discontinuation of therapy and during follow-up, respectively (1 HRCA, n=2 [fall and coronavirus disease 2019 (COVID-19) pneumonia]). Neither of the deaths were preceded by PD.
  • In GRIFFIN, 8 patients died while on D-VRd therapy (0 HRCA, n=2 [due to adverse event (bronchopneumonia) and PD]; 1 HRCA, n=1 [due to PD]; ≥2 HRCAs, n=4 [all due to PD]; not evaluable for cytogenetics, n=1 [due to respiratory failure]).

CLINICAL STUDIES - RELAPSED OR RELAPSED REFRACTORY MULTIPLE MYELOMA

DARZALEX in Combination with Bortezomib and Dexamethasone

CASTOR (MMY3004) is a phase 3, open-label, randomized, multicenter, active-controlled study that assessed the safety and efficacy of Vd alone and D-Vd in patients with RRMM.34 Sonneveld et al (2022)35 reported updated results of the CASTOR study, including OS, at a median follow-up of 72.6 months. Results in the high-risk cytogenetic subgroup are summarized below.

Study Design/Methods

  • Primary endpoint: PFS
  • Secondary endpoints: time to disease progression; percentage of patients with overall response; DOR; TTR, percentage of patients with a ≥VGPR; percentage of patients with OS; MRD

Results


Key Baseline Characteristics (ITT Population; CASTOR)35
Characteristic
D-Vd
(N=251)
Vd
(N=247)
ISS staginga, n (%)
   I
98 (39)
96 (39)
   II
94 (37)
100 (40)
   III
59 (24)
51 (21)
Cytogenetic profileb, n/N (%)
   Standard risk
140/181 (77)
137/174 (79)
   High risk
41/181 (23)
37/174 (21)
Abbreviations: D-Vd, DARZALEX + bortezomib + dexamethasone; ISS, International Staging System; Vd, bortezomib + dexamethasone.
aISS staging is derived based on the combination of serum β2-microglobulin and albumin.
bCytogenetic risk was assessed locally by fluorescence in situ hybridization or karyotype testing; high risk was defined as the presence of t(4;14), t(14;16), or del17p.

Efficacy

OS in ISS Staging and Cytogenetic Risk Pre-Specified Subgroups (ITT Population; CASTOR)35
Subgroup
D-Vd
Vd
Hazard Ratio (95% CI)
n/N
Median
n/N
Median
ISS disease stagea
   I
49/98
72.7
59/96
49.8
0.70 (0.47-1.03)
   II
57/94
48.5
70/100
40.3
0.73 (0.50-1.05)
   III
42/59
21.6
42/51
12.9
0.77 (0.48-1.24)
Cytogenetic risk at study entryb
   High risk
30/40
38.4
27/35
28.8
0.77 (0.41-1.46)
   Low risk
78/141
55.8
93/140
41.8
0.71 (0.52-0.98)
Abbreviations: CI, confidence interval; D-Vd, DARZALEX + bortezomib + dexamethasone; ISS, International Staging System; ITT, intent-to-treat; OS, overall survival; Vd, bortezomib + dexamethasone.
aISS disease stage was derived based on the combination of serum β2-microglobulin and albumin levels. Higher stages indicate more severe disease.bCytogenetic risk was assessed locally by fluorescence in situ hybridization or karyotype testing. High risk was defined as the presence of t(4;14), t(14;16), or del17p.

Safety
  • Grade 3/4 TEAEs occurring in ≥10% of patients in the D-Vd vs Vd arm were thrombocytopenia (46.1% vs 32.9%), anemia (16% in both arms), neutropenia (13.6% vs 4.6%), lymphopenia (10.3% vs 2.5%), and pneumonia (10.7% vs 10.1%).
  • Serious TEAEs were reported in 134 (55.1%) patients vs 81 (34.2%) patients in the D-Vd vs Vd arms, respectively.
    • The most common serious TEAE was pneumonia (D-Vd, 10.7%; Vd, 10.1%).
  • Treatment discontinuations due to TEAEs was 10.7% vs 9.3% in the D-Vd vs Vd arms, respectively.
  • Deaths due to TEAEs were reported in 17 (7%) patients vs 14 (5.9%) patients in the D-Vd vs Vd arms, respectively.
    • The most frequent TEAEs leading to death were pneumonia (0.8% in both arms) and general physical health (D-Vd, 0.4%; Vd, 1.3%).

Subgroup Analysis from CASTOR

Weisel et al (2020)36 reported updated efficacy and safety data based on cytogenetic risk status of D-Vd from CASTOR in patients with RRMM.

Study Design/Methods

  • Cytogenetic risk was evaluated using local FISH or karyotyping.
  • Patients in the ITT population who had ≥1 FISH or karyotyping assessment were included in the analysis.
  • Patients with high-risk cytogenetics included those who had ≥1 of the following abnormalities: t(4;14), t(14;16), or del17p.

Results


Key Patient Baseline Disease and Clinical Characteristics (CASTOR)36
Characteristic
Standard Cytogenetic Riska
High Cytogenetic Riska,b
D-Vd
(n=141)

Vd
(n=140)

D-Vd
(n=40)

Vd
(n=35)

ISS stagingc, n (%)
   I
48 (34)
55 (39)
22 (55)
14 (40)
   II
57 (40)
56 (40)
11 (28)
16 (46)
   III
36 (26)
29 (21)
7 (18)
5 (14)
Cytogenetic profilea, n (%)
   t(4;14)
-
-
13 (33)
15 (43)
   t(14;16)
-
-
4 (10)
5 (14)
   del17p
-
-
27 (68)
20 (57)
   ≥2 risk factorsd
-
-
4 (10)
4 (11)
Abbreviations: D-Vd, DARZALEX + bortezomib + dexamethasone; FISH, fluorescence in situ hybridization;
ISS, International Staging System; Vd, bortezomib + dexamethasone.
Note: percentages may not equal 100% due to rounding.
aBased on FISH/karyotype testing.
bPatients with high cytogenetic risk had a t(4;14), t(14;16), or del17p abnormality.
cISS staging is derived based on the combination of serum β2-microglobulin and albumin.
dPatients with ≥2 of the t(4;14), t(14;16), or del17p risk factors.

Efficacy
  • After a median follow-up of 40 months (ITT population), treatment with D-Vd significantly prolonged PFS compared with Vd in patients with standard cytogenetic risk (median: 16.6 months vs 6.6 months; HR, 0.26; 95% CI, 0.19-0.37; P<0.0001) and high cytogenetic risk (median: 12.6 vs 6.2 months; HR, 0.41; 95% CI, 0.21-0.83; P=0.0106).
    • Among patients who had received 1 prior line of therapy, treatment with D-Vd significantly prolonged PFS compared with Vd in patients with standard cytogenetic risk (median: 29.8 vs 7.5 months; HR, 0.25; 95% CI, 0.15-0.42; P<0.0001) and high cytogenetic risk (median: 20.1 months vs 8.4 months; HR, 0.20; 95% CI, 0.06-0.62; P=0.0026).
  • ORR and rates of ≥VGPR and ≥CR were higher with D-Vd vs Vd, regardless of cytogenetic risk status; Please see Table: Response and MRD-negative Rates in Patients with Standard and High Cytogenetic Risk (CASTOR).
    • Time to ≥VGPR was decreased with D-Vd vs Vd in patients with standard cytogenetic risk (median: 3.5 months vs NE; HR, 2.16; 95% CI, 1.46-3.20; P<0.0001) and high cytogenetic risk (median: 3.5 months vs 6.2 months; HR, 1.96; 95% CI, 0.86-4.45; P=0.1004).
  • Rates of MRD-negativity were higher with D-Vd vs Vd, regardless of cytogenetic risk status as shown in Table: Response and MRD-negative Rates in Patients with Standard and High Cytogenetic Risk (CASTOR).
  • PFS was prolonged with D-Vd vs Vd in MRD-positive (median: 13.5 months vs 6.5 months; HR, 0.28; 95% CI, 0.20-0.40; P<0.0001) and MRD-negative (median, NE vs 37.6 months; HR, NE; P=0.4142) patients with standard cytogenetic risk.
  • Among patients with high cytogenetic risk, PFS was prolonged with D-Vd vs Vd in MRD-positive patients (median: 10.5 months vs 6.2 months; HR, 0.49; 95% CI, 0.25-0.97; P=0.0383); median PFS with D-Vd in MRD-negative patients was NE.
  • PFS2 was prolonged with D-Vd vs Vd in the standard cytogenetic risk (median: 34.2 months vs 18.5 months; HR, 0.41; 95% CI, 0.30-0.58; P<0.0001) and high cytogenetic risk (median: 28.1 months vs 19.7 months; HR, 0.58; 95% CI, 0.30-1.10; P=0.0915) subgroups.
  • At the time of analysis, OS data were immature and follow-up for OS was ongoing.

Response and MRD-negative Rates in Patients with Standard and High Cytogenetic Risk (CASTOR)36
Responsea, n (%)
Standard-Risk
High-Riskb
D-Vd
(n=135)

Vd
(n=134)

P value
D-Vd
(n=39)

Vd
(n=34)

P value
ORR
113 (84)
83 (62)
<0.0001
33 (85)
19 (56)
0.0512
≥CR
38 (28)
13 (10)
2 (6)
2 (6)
sCR
12 (9)
3 (2)
4 (10)
0
CR
26 (19)
10 (8)
7 (18)
2 (6)
≥VGPR
83 (62)
38 (28)
<0.0001
23 (59)
11 (32)
0.1259
VGPR
45 (33)
25 (19)
12 (31)
9 (27)
PR
30 (22)
45 (34)
10 (26)
8 (24)
MRD-negative (10-5)c
n=141
n=140
n=40
n=35
n (%)
16 (11)
4 (3)
0.0091
6 (15)
0
0.0271
Sustained MRD negativity (≥6 months), n (%)
9 (6)
3 (2)
0.1374
5 (13)
0
0.0569
Sustained MRD negativity (≥12 months), n (%)
2 (1)
0
0.4982
3 (8)
0
0.2432
Abbreviations: CR, complete response; D, DARZALEX; ITT, intent-to-treat; MRD, minimal residual disease; ORR, overall response rate; PR, partial response; sCR, stringent complete response; Vd, bortezomib + dexamethasone; VGPR, very good partial response.
aResponse-evaluable population.bPatients with high cytogenetic risk had a t(4;14), t(14;16), or del17p abnormality.
cITT population.

Safety
  • The most frequent any grade (≥25% of patients) TEAEs and most frequent grade 3/4 (≥5% of patients) TEAEs are summarized in Table: Most Common Any-grade (≥25% of Patients) and Grade 3/4 (≥5% of Patients) TEAEs (CASTOR).
  • Treatment discontinuation rates due to TEAEs were similar between treatment groups for patients with standard cytogenetic risk (D-Vd, n=11 [8%]; Vd, n=14 [10%]).
  • Among patients with high cytogenetic risk, a similar proportion discontinued treatment due to TEAEs (D-Vd, n=4 [10%]; Vd, n=3 [9%]).

Most Common Any-grade (≥25% of Patients) and Grade 3/4 (≥5% of Patients) TEAEs (CASTOR)36
TEAE, n (%)
Any-Grade
Grade 3/4
Standard-Risk
High-Riska
Standard-Risk
High-Riska
D-Vd
(n=137)

Vd
(n=136)

D-Vd
(n=40)

Vd
(n=34)

D-Vd
(n=137)

Vd
(n=136)

D-Vd
(n=40)

Vd
(n=34)

Hematologic
   Thrombocytopenia
85 (62)
58 (43)
24 (60)
16 (47)
65 (47)
45 (33)
19 (48)
12 (35)
   Anemia
45 (33)
41 (30)
7 (18)
14 (41)
25 (18)
23 (17)
4 (10)
6 (18)
   Neutropenia
29 (21)
16 (12)
9 (23)
3 (9)
21 (15)
6 (4)
6 (15)
2 (6)
   Lymphopenia
18 (13)
5 (4)
4 (10)
4 (12)
14 (10)
3 (2)
3 (8)
3 (9)
   Leukopenia
15 (11)
5 (4)
3 (8)
3 (9)
5 (4)
1 (1)
1 (3)
2 (6)
Nonhematologic
   Peripheral sensory
   neuropathy

67 (49)
50 (37)
22 (55)
13 (38)
4 (3)
8 (6)
2 (5)
4 (12)
   Upper respiratory tract
   infection
43 (31)
20 (15)
15 (38)
8 (24)
1 (1)
0 (0)
3 (8)
1 (3)
   Diarrhea
42 (31)
35 (26)
11 (28)
6 (18)
6 (4)
2 (2)
1 (3)
0 (0)
   Cough
40 (29)
19 (14)
9 (23)
4 (12)
0 (0)
0 (0)
0 (0)
0 (0)
   Fatigue
25 (18)
40 (29)
17 (43)
8 (24)
6 (4)
5 (4)
2 (5)
1 (3)
   Back pain
24 (18)
15 (11)
13 (33)
1 (3)
3 (2)
0 (0)
1 (3)
0 (0)
   Insomnia
22 (16)
20 (15)
11 (28)
5 (15)
2 (2)
0 (0)
0 (0)
1 (3)
   Pneumonia
22 (16)
20 (15)
5 (13)
4 (12)
15 (11)
14 (10)
2 (5)
3 (9)
   Asthenia
15 (11)
19 (14)
4 (10)
9 (27)
1 (1)
3 (2)
0 (0)
1 (3)
   Hypertension
15 (11)
5 (4)
4 (10)
1 (3)
9 (7)
1 (1)
2 (5)
0 (0)
   Decreased appetite
14 (10)
8 (6)
10 (25)
1 (3)
0 (0)
1 (1)
1 (3)
0 (0)
   Spinal pain
4 (3)
3 (2)
2 (5)
0 (0)
1 (1)
0 (0)
2 (5)
0 (0)
   Gastroenteritis
2 (2)
3 (2)
2 (5)
1 (3)
0 (0)
2 (2)
2 (5)
1 (3)
   Squamous cell carcinoma
   of skin
0 (0)
0 (0)
2 (5)
0 (0)
0 (0)
0 (0)
2 (5)
0 (0)
Abbreviations: D-Vd, DARZALEX + bortezomib + dexamethasone; TEAE, treatment-emergent adverse event; Vd, bortezomib + dexamethasone.
aPatients with high cytogenetic risk had a t(4;14), t(14;16), or del17p abnormality.

DARZALEX in Combination with Lenalidomide and Dexamethasone

POLLUX (MMY3003) was a phase 3, randomized, open-label, multicenter study assessed the safety and efficacy of Rd vs D-Rd in patients with RRMM.37 Dimopoulos et al (2022)38 presented updated results of the POLLUX study, including OS, at a median follow-up of 79.7 months. Results in the high-risk cytogenetic subgroup are summarized below.

Study Design/Methods

  • Primary endpoint: PFS
  • Secondary endpoints: TTP, ORR, rate of ≥VGPR, rate of CR or better, OS, DOR, safety, and MRD

Results


Key Baseline Characteristics (ITT Population; POLLUX)38
Characteristic
D-Rd
(N=286)

Rd
(N=283)

ISS staginga, n (%)
   I
137 (47.9)
140 (49.5)
   II
93 (32.5)
86 (30.4)
   III
56 (19.6)
57 (20.1)
Cytogenetic profileb, n/N (%)
   Standard risk
193/228 (84.6)
176/211 (83.4)
   High risk
35/228 (15.4)
35/211 (16.6)
Abbreviations: aISS staging was based on the combination of serum β2-microglobulin and albumin.bCytogenetic risk was assessed locally by fluorescence in situ hybridization or karyotype testing; high risk was defined as the presence of t(4;14), t(14;16), or del17p.
Efficacy

OS in ISS Staging and Cytogenetic Risk Pre-Specified Subgroups (ITT Population; POLLUX)38
Subgroup
D-Rd
Rd
Hazard Ratio (95% CI)
n/N
Median
n/N
Median
ISS disease stagea
   I
54/137
NE
70/140
71.9
0.76 (0.53-1.08)
   II
61/93
50.4
65/86
38.5
0.71 (0.50-1.01)
   III
38/56
39
40/57
20.3
0.74 (0.47-1.15)
Cytogenetic risk at study entryb
   High risk
25/35
40
28/35
23.6
0.70 (0.41-1.20)
   Low risk
104/193
67.6
107/176
51.8
0.80 (0.61-1.05)
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intent-to-treat; OS, overall survival; Rd, lenalidomide + dexamethasone.
aISS disease stage was derived based on the combination of serum β2-microglobulin and albumin levels. Higher stages indicate more severe disease.
bCytogenetic risk was assessed locally by fluorescence in situ hybridization or karyotype testing. High risk was defined as the presence of t(4;14), t(14;16), or del17p.

Safety
  • Grade 3/4 TEAEs occurring in ≥10% of patients in the D-Rd vs Rd arm were neutropenia (57.6% vs 41.6%), anemia (19.8% vs 22.4%), pneumonia (17.3% vs 11%), thrombocytopenia (15.5% vs 15.7%), and diarrhea (10.2% vs 3.9%).
  • Treatment discontinuations due to TEAEs was 19.1% vs 16% in the DRd vs Rd arms, respectively.
  • Deaths due to TEAEs were reported in 35 (12.4%) patients vs 24 (8.5%) patients in the DRd vs Rd arms, respectively.

Subgroup Analysis from POLLUX

Kaufman et al (2020)39 presented updated efficacy and safety data based cytogenetic risk status of D-Rd from POLLUX in patients with RRMM.

Study Design/Methods

  • Cytogenetic abnormalities were detected by local FISH or karyotyping on bone marrow aspirates collected at screening visits.
  • Patients in the ITT population who had ≥1 assessment from FISH or karyotyping were included in the analysis.
  • Patients with high-risk cytogenetic status included those who had ≥1 of the following abnormalities: t(4;14), t(14;16), or del17p.


Results


Patient Demographic, Baseline Disease, and Clinical Characteristics (POLLUX)39
Characteristic
Standard Cytogenetic
Riska

High Cytogenetic
Riska,b

D-Rd
(n=193)

Rd
(n=176)

D-Rd
(n=35)

Rd
(n=35)

Age, years
   Median (range)
66 (36-89)
64 (42-85)
67(50-80)
67 (50-81)
   ≥75 years, n (%)
21 (11)
17 (10)
4 (11)
6 (17)
ISS stagec, n (%)
   I
96 (50)
92 (52)
13 (37)
14 (40)
   II
62 (32)
50 (28)
15 (43)
13 (37)
   III
35 (18)
34 (19)
7 (20)
8 (23)
ECOG performance status score, n (%)
   0
94 (49)
90 (51)
15 (43)
22 (63)
   1
91 (47)
78 (44)
18 (51)
12 (34)
   2
8 (4)
8 (5)
2 (6)
1 (3)
Cytogenetic profilea,b, n (%)
   t(4;14)
-
-
10 (29)
15 (43)
   t(14;16)
-
-
2 (6)
6 (17)
   del17p
-
-
25 (71)
20 (57)
   ≥2 risk factorsd
-
-
2 (6)
5 (14)
Median (range) time from diagnosis, years
3.7
(0.6-22.5)

4
(0.4-18.3)

3.2
(0.4-27)

2.3
(0.4-14.6)

Prior lines of therapy, n (%)
   1
96 (50)
90 (51)
22 (63)
20 (57)
   2
62 (32)
47 (27)
6 (17)
9 (26)
   3
24 (12)
24 (14)
5 (14)
4 (11)
   >3
11 (6)
15 (9)
2 (6)
2 (6)
   Median (range)
2 (1-11)
1 (1-8)
1 (1-6)
1 (1-6)
Prior ASCT, n (%)
115 (60)
115 (65)
22 (63)
19 (54)
Prior PI, n (%)
162 (84)
148 (84)
31 (89)
31 (89)
   Bortezomib
160 (83)
145 (82)
30 (86)
31 (89)
Prior IMiD, n (%)
103 (53)
102 (58)
22 (63)
15 (43)
   Lenalidomide
27 (14)
33 (19)
10 (29)
4 (11)
Prior PI + IMiD, n (%)
79 (41)
80 (46)
18 (51)
12 (34)
Refractory to PI only, n (%)
42 (22)
29 (17)
8 (23)
9 (26)
Refractory to IMiD only, n (%)
7 (4)
10 (6)
3 (9)
1 (3)
Refractory to PI and IMiD, n (%)
6 (3)
7 (4)
1 (3)
3 (9)
Refractory to last line of therapy, n (%)
62 (32)
50 (28)
11 (31)
13 (37)
Abbreviations: ASCT, autologous stem cell transplant; D-Rd, DARZALEX + lenalidomide + dexamethasone; ECOG, Eastern Cooperative Oncology Group; FISH, fluorescence in situ hybridization; IMiD, immunomodulatory drug; ISS, International Staging System; PI, proteasome inhibitor; Rd, lenalidomide + dexamethasone.
Note: percentages may not equal 100% due to rounding.
aBased on FISH/karyotyping.
bPatients with high cytogenetic risk had a t(4;14), t(14;16), or del17p abnormality.
cISS stage is derived based on the combination of serum β2-microglobulin and albumin.
dPatients with ≥2 of the t(4;14), t(14;16), or del17p risk factors.

Efficacy
  • After a median follow-up of 44.3 months (ITT population), D-Rd significantly prolonged PFS vs Rd in patients with standard cytogenetic risk (median: NE vs 18.6 months; HR, 0.43; 95% CI, 0.32-0.57; P<0.0001; 42-month PFS rate: 53% vs 25%) and high cytogenetic risk (median: 26.8 months vs 8.3 months; HR, 0.34; 95% CI, 0.16-0.72; P=0.0035; 42-month PFS rate: 35% vs 13%).
    • Among patients with 1 prior line of therapy, D-Rd significantly prolonged PFS vs Rd in patients with standard cytogenetic risk (median: NE vs 20.2 months; HR, 0.41; 95% CI, 0.27-0.63; P<0.0001; 42-month PFS rate: 58% vs 27%) and high cytogenetic risk (median: 29.6 months vs 6.6 months; HR, 0.26; 95% CI, 0.09-0.75; P=0.0083; 42-month PFS rate: 43% vs 12%).
  • The ORR and rates of ≥VGPR and ≥CR were higher with D-Rd vs Rd, regardless of cytogenetic risk status (Table: Response and MRD-negativity Rates in Patients with Standard and High Cytogenetic Risk [POLLUX]).
    • Among patients with standard cytogenetic risk, time to ≥VGPR (median: 3.7 months vs 8.3 months; HR, 1.98; 95% CI, 1.52-2.59; P<0.0001) and ≥CR (median: 13.9 months vs NE; HR, 2.38; 95% CI, 1.66-3.43; P<0.0001) were decreased with D-Rd vs Rd.
    • Among patients with high cytogenetic risk, time to ≥VGPR (median: 3.7 months vs 18.7 months; HR, 3.45; 95% CI, 1.41-8.39; P=0.0040) and ≥CR (median: 14.4 months vs NE; HR, 2.07; 95% CI, 0.51-8.48; P=0.3011) were also decreased with D-Rd vs Rd.
  • MRD-negativity rates were higher with D-Rd vs Rd, regardless of cytogenetic risk (Table: Response and MRD-negativity Rates in Patients with Standard and High Cytogenetic Risk [POLLUX]).
  • PFS was prolonged with D-Rd vs Rd in MRD-positive (median: 36.4 months vs 17.1 months; HR, 0.51; 95% CI, 0.37-0.69; P<0.0001) and MRD-negative (median: NE vs 42 months; HR, 0.56; 95% CI, 0.22-1.47; P=0.2367) patients with standard cytogenetic risk.
  • Among patients with high cytogenetic risk, PFS was prolonged with D-Rd vs Rd in MRD-positive patients (median: 20.3 months vs 8.3 months; HR, 0.49; 95% CI, 0.23-1.03; P=0.0567); median PFS with D-Rd in MRD-negative patients was NE.
  • D-Rd significantly prolonged PFS2 vs Rd in the standard cytogenetic risk (median: NE vs 33.3 months; HR, 0.54, 95% CI, 0.40-0.74; P<0.0001) and high cytogenetic risk (median: 37.7 months vs 20.8 months; HR, 0.38; 95% CI, 0.18-0.83; P=0.0121) subgroups.
  • At the time of analysis, OS data were immature and follow-up for OS was ongoing.

Response and MRD-negativity Rates in Patients with Standard and High Cytogenetic Risk (POLLUX)39
Responsea, n (%)
Standard-Risk
High-Riskb
D-Rd
(n=190)

Rd
(n=172)

P value
D-Rd
(n=35)

Rd
(n=34)

P value
ORR
178 (94)
135 (79)
<0.0001
31 (89)
23 (68)
0.0145
≥CR
111 (58)
43 (25)
15 (43)
3 (9)
   sCR
61 (32)
23 (13)
10 (29)
1 (3)
   CR
50 (26)
20 (12)
5 (14)
2 (6)
≥VGPR
156 (82)
92 (54)
<0.0001
25 (71)
10 (29)
0.0004
VGPR
45 (24)
49 (29)
10 (29)
7 (21)
PR
22 (12)
43 (25)
6 (17)
13 (38)
MRD-negative (10-5)c
n=193
n=176
n=35
n=35
n (%)
63 (33)
15 (9)
<0.0001
9 (26)
0 (0)
0.0022
Sustained MRD-negativity (≥6 months), n (%)
35 (18)
2 (1)
<0.0001
1 (3)
0 (0)
Sustained MRD-negativity (≥12 months), n (%)
27 (14)
1 (1)
<0.0001
1 (3)
0 (0)
Abbreviations: CR, complete response; D-Rd, DARZALEX + lenalidomide + dexamethasone; ITT, intent-to-treat; MRD, minimal residual disease; ORR, overall response rate; PR, partial response; Rd, lenalidomide + dexamethasone; sCR, stringent complete response; VGPR, very good partial response.
aResponse-evaluable population.bPatients with high cytogenetic risk had a t(4;14), t(14;16), or del17p abnormality.
cITT population.

Safety

  • The most common (≥25% of patients) any-grade TEAEs and the most common (≥5% of patients) grade 3/4 TEAEs are presented in Table: Most Common Any-grade (≥25% of Patients) and Grade 3/4 (≥5% of Patients) TEAEs (POLLUX).
  • The rates of treatment discontinuation due to TEAEs were similar among patients with standard cytogenetic risk (D-Rd, n=28 [15%]; Rd, n=22 [13%]).
    • Among patients with high cytogenetic risk, a higher proportion of patients discontinued treatment due to a TEAE in the D-Rd arm (D-Rd, n=7 [20%]; Rd, n=3 [9%]).

Most Common Any-grade (≥25% of Patients) and Grade 3/4 (≥5% of Patients) TEAEs (POLLUX)39
TEAE, n (%)
Any-Grade
Grade 3/4
Standard-Risk
High-Riska
Standard-Risk
High-Riska
D-Rd
(n=192)

Rd
(n=176)

D-Rd
(n=35)

Rd
(n=34)

D-Rd
(n=192)

Rd
(n=176)

D-Rd
(n=35)

Rd
(n=34)

Hematologic
   Neutropenia
116 (60)
80 (46)
22 (63)
18 (53)
103 (54)
69 (39)
17 (49)
16 (47)
      Febrile neutropenia
10 (5)
4 (2)
2 (6)
0 (0)
10 (5)
4 (2)
2 (6)
0 (0)
   Anemia
71 (37)
62 (35)
13 (37)
15 (44)
31 (16)
32 (18)
7 (20)
10 (29)
   Thrombocytopenia
55 (29)
46 (26)
13 (37)
14 (41)
25 (13)
24 (14)
8 (23)
10 (29)
   Leukopenia
18 (9)
15 (9)
3 (9)
4 (12)
6 (3)
4 (2)
1 (3)
3 (9)
   Lymphopenia
10 (5)
10 (6)
5 (14)
3 (9)
8 (4)
7 (4)
5 (14)
3 (9)
Nonhematologic
   Diarrhea
104 (54)
53 (30)
18 (51)
16 (47)
20 (10)
5 (3)
3 (9)
4 (12)
   Upper respiratory tract
   infection

82 (43)
56 (32)
10 (29)
8 (24)
4 (2)
3 (2)
0 (0)
0 (0)
   Fatigue
74 (39)
52 (30)
12 (34)
12 (35)
13 (7)
9 (5)
4 (11)
2 (6)
   Cough
65 (34)
23 (13)
11 (31)
6 (18)
0 (0)
0 (0)
0 (0)
0 (0)
   Nasopharyngitis
64 (33)
37 (21)
11 (31)
9 (27)
0 (0)
0 (0)
0 (0)
0 (0)
   Constipation
57 (30)
46 (26)
12 (34)
10 (29)
0 (0)
1 (1)
2 (6)
1 (3)
   Insomnia
54 (28)
41 (23)
9 (26)
4 (12)
5 (3)
3 (2)
0 (0)
1 (3)
   Muscle spasms
52 (27)
36 (21)
12 (34)
7 (21)
2 (1)
3 (2)
1 (3)
0 (0)
   Pneumonia
51 (27)
30 (17)
10 (29)
6 (18)
32 (17)
20 (11)
4 (11)
4 (12)
   Nausea
48 (25)
37 (21)
14 (40)
8 (24)
5 (3)
2 (1)
1 (3)
0 (0)
   Peripheral edema
47 (25)
24 (14)
8 (23)
3 (9)
1 (1)
1 (1)
0 (0)
0 (0)
   Pyrexia
48 (25)
18 (10)
10 (29)
5 (15)
5 (3)
5 (3)
1 (3)
1 (3)
   Dyspnea
37 (19)
23 (13)
7 (20)
4 (12)
2 (1)
1 (1)
4 (11)
0 (0)
   Hypokalemia
37 (19)
19 (11)
7 (20)
4 (12)
14 (7)
5 (3)
2 (6)
1 (3)
   Cataract
35 (18)
24 (14)
4 (11)
2 (6)
13 (7)
8 (5)
1 (3)
1 (3)
   Bronchitis
32 (17)
24 (14)
10 (29)
4 (12)
6 (3)
5 (3)
0 (0)
0 (0)
   Arthralgia
27 (14)
22 (13)
10 (29)
5 (15)
2 (1)
1 (1)
1 (3)
0 (0)
   Influenza
20 (10)
9 (5)
5 (14)
2 (6)
2 (1)
0 (0)
2 (6)
0 (0)
   Hyperglycemia
18 (9)
14 (8)
6 (17)
3 (9)
5 (3)
7 (4)
4 (11)
2 (6)
   Hypophosphatemia
12 (6)
7 (4)
4 (11)
2 (6)
9 (5)
5 (3)
3 (9)
0 (0)
   Hypertension
12 (6)
8 (5)
9 (26)
2 (6)
5 (3)
2 (1)
4 (11)
0 (0)
   Increased alanine
   aminotransferase

12 (6)
7 (4)
5 (14)
3 (9)
6 (3)
2 (1)
2 (6)
1 (3)
   Syncope
9 (5)
2 (1)
2 (6)
1 (3)
9 (5)
2 (1)
2 (6)
1 (3)
   Pulmonary embolism
4 (2)
8 (5)
2 (6)
2 (6)
4 (2)
7 (4)
2 (6)
2 (6)
   Hypercalcemia
2 (1)
6 (3)
3 (9)
2 (6)
1 (1)
2 (1)
2 (6)
2 (6)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; Rd, lenalidomide + dexamethasone; TEAE, treatment-emergent adverse event.
aPatients with high cytogenetic risk had a t(4;14), t(14;16), or del17p abnormality.

Post Hoc Analyses of CASTOR and POLLUX Studies in Clinically Relevant Subgroups

Mateos et al (2022)40 presented a post hoc analysis of the phase 3 CASTOR and POLLUX studies to evaluate the efficacy of D-Vd vs Vd and DRd vs Rd in subgroup of patients with RRMM. Results of a subgroups of patients based on ISS stage III disease and High-risk cytogenetic risk abnormalities (t[4;14], t[14;16], and/or del17p) have been summarized below.

Study Design/Methods

  • Patients with RRMM who had received ≥1PL were included.
  • Patients refractory or intolerant to bortezomib or refractory to another PI were excluded from CASTOR, and patients refractory or intolerant to lenalidomide were excluded from POLLUX.
  • Patients were randomly allocated to receive D-Vd or Vd in CASTOR and D-Rd or Rd in POLLUX.
  • Primary endpoint: PFS

Results


PFS and OS in the ISS Stage III and High Cytogenetic Risk Subgroups of CASTOR and POLLUX (Pooled Analysis; ITT Population)40
Subgroup
PFS
OS
DARZALEX Arm
Control Arm
HR
(95%
CI)

DARZALEX Arm
Control Arm
HR
(95%
CI)

n/N
Median, months
n/N
Median, Mos
n/N
Median, months
n/N
Median, months
ISS stage IIIa
89/115
17.9
86/108
6.1
0.53
(0.39-0.72)

80/115
27.6
82/108
17.1
0.70
(0.52-0.96)

High cytogenetic riskb
64/75
20.3
61/70
7.3
0.45
(0.31-0.65)

55/75
40
55/70
23.6
0.68
(0.47-1)

Abbreviations: CI, confidence interval; HR, hazard ratio; ITT, intent-to-treat; Mos, months; OS, overall survival; PFS, progression-free survival.
aISS staging was based on the combination of serum β2-microglobulin and albumin levels.
bCytogenetic risk was determined based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality.


PFS in the ISS Stage III and High Cytogenetic Risk Subgroups of CASTOR and POLLUX (ITT Population)40
Subgroup
CASTOR
POLLUX
D-Vd
Vd
HR
(95%
CI)

D-Rd
Rd
HR
(95%
CI)

n/N
Median, months
n/N
Median, months
n/N
Median, months
n/N
Median, months
ISS stage IIIa
48/59
8.3
41/51
5.3
0.43 (0.27-0.69)
41/56
26.7
45/57
8.8
0.49 (0.32-0.77)
High cytogenetic riskb
35/40
12.6
31/35
6.2
0.37 (0.21-0.64)
29/35
26.8
30/35
8.3
0.47 (0.28-0.79)
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; D-Vd, DARZALEX + bortezomib + dexamethasone; HR, hazard ratio; ISS, International Staging System; ITT, intent-to-treat; PFS, progression-free survival; Rd, lenalidomide + dexamethasone; Vd, bortezomib + dexamethasone.
aISS staging was based on the combination of serum β2-microglobulin and albumin levels.
bCytogenetic risk was determined based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality.


ORR and MRD-Negativity (10-5) Rates in ISS Stage III and High Cytogenetic Risk Subgroups (CASTOR)40
Subgroup
ORRa
MRD-negativity Rateb
D-Rd
(n/N [%])

Rd
(n/N [%])

OR
(95% CI)

P value
D-Rd
(n/N [%])

Rd
(n/N [%])

OR
(95% CI)

P value
ISS stage IIIc
36/51 (70.6)
21/47 (44.7)
2.97
(1.29-6.83)
0.0098
6/59 (10.2)
0/51
NE
(NE-NE)

0.0294
High cytogenetic riskd
33/39 (84.6)
19/34 (55.9)
4.34
(1.44-13.07)
0.0072
6/40 (15)
0/35
NE
(NE-NE)

0.0271
Abbreviations: CI, confidence interval; CR, complete response; D-Rd, DARZALEX + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; OR, odds ratio; ORR, overall response rate; PR, partial response; Rd, lenalidomide + dexamethasone; sCR, stringent complete response; VGPR, very good partial response.
aAnalyses were performed for the response-evaluable populations, where the overall response was defined as sCR+CR+VGPR+PR.
bAnalyses were performed for the ITT population.
cISS staging was based on the combination of serum β2-microglobulin and albumin levels.
d
Cytogenetic risk was determined based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality.


ORR and MRD-Negativity (10-5) Rates in ISS Stage III and High Cytogenetic Risk Subgroups (POLLUX)40
Subgroup
ORRa
MRD-negativity Rateb
D-Rd
(n/N [%])

Rd
(n/N [%])

OR
(95% CI)

P value
D-Rd
(n/N [%])

Rd
(n/N [%])

OR
(95% CI)

P value
ISS stage IIIc
47/53 (88.7)
38/52 (73.1)
2.89
(1.01-8.23)

0.0428
18/56 (32.1)
6/57
(10.5)

4.03
(1.46-11.11)

0.0059
High cytogenetic riskd
31/35 (88.6)
23/34 (67.6)
3.71
(1.05-13.13)

0.0365
10/35 (28.6)
1/35
(2.9)

13.60
(1.63-113.25)

0.0063
Abbreviations: CI, confidence interval; CR, complete response; D-Rd, DARZALEX + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intent-to-treat; MRD, minimal residual disease; OR, odds ratio; ORR, overall response rate; PR, partial response; Rd, lenalidomide + dexamethasone; sCR, stringent complete response; VGPR, very good partial response.
aAnalyses were performed for the response-evaluable populations, where the overall response was defined as sCR+CR+VGPR+PR.
bAnalyses were performed for the ITT population.
cISS staging was based on the combination of serum β2-microglobulin and albumin levels.
d
Cytogenetic risk was determined based on local fluorescence in situ hybridization or karyotype analysis. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality.

DARZALEX in Combination with Carfilzomib and Dexamethasone

CANDOR is a randomized, open-label, multicenter, phase 3 study evaluating the efficacy and safety of D-Kd vs Kd in patients with RRMM.41,42 Usmani et al (2023)43 reported the final efficacy and safety results of the CANDOR study after a median follow-up of approximately 50 months. Results in high-risk patients are summarized below.

Study Design/Methods

  • High-risk cytogenetic status was assessed by FISH and included those who had ≥1 of the following abnormalities: t(4;14), t(14;16), or del17p.
  • Primary endpoint: PFS
  • Key secondary endpoints: ORR, MRD (10-5), and OS

Results

Patient Characteristics

Key Baseline Patient and Disease Characteristics (CANDOR)43
Characteristic 
D-Kd (n=312)
Kd (n=154)
ISS stage, n (%)
   I
147 (47)
79 (51)
   II
103 (33)
48 (31)
   III
61 (20)
27 (18)
   Unknown
1 (<1)
0 (0)
Cytogenetic risk group by FISH, n (%)
   High-riska
48 (15)
26 (17)
   Standard-risk
107 (34)
56 (36)
   Unknownb
157 (50)
72 (47)
Abbreviations: D-Kd, DARZALEX + carfilzomib + dexamethasone; FISH, fluorescence in situ hybridization; ISS, International Staging System; Kd, carfilzomib + dexamethasone.
aConsists of genetic subtypes t(4;14), t(14;16), or del(17p).
bIncludes samples that failed or were cancelled.

Efficacy

OS in Pre-specified ISS Staging and Cytogenetic Risk Subgroups (CANDOR)43
Events/Patients
Median OS, months (95% CI)
HR for D-Kd vs Kd (95% CI)
D-Kd
Kd
D-Kd
Kd
Overall
148/312
80/154
50.8 (44.7-NE)
43.6 (35.3-NE)
0.784 (0.595-1.033)
ISS stage at screening
   I or II
104/252
58/127
NE (50.8-NE)
51.8 (41.9-NE)
0.870 (0.630-1.200)
   III
44/60
22/27
26.5 (21.4-38.3)
12 (4.9-17.8)
0.584 (0.345-0.989)
Cytogenetic risk group
   High risk
32/48
20/26
34.3 (22-46.5)
17.1 (8.5-35.3)
0.521 (0.288-0.942)
   Standard risk
44/108
30/56
NE (48.8-NE)
38.2 (32.9-NE)
0.621 (0.382-1.009)
   Unknown
72/156
30/72
NE (43.2-NE)
NE (42.9-NE)
1.062 (0.690-1.635)
Abbreviations: CI, confidence interval; D-Kd, DARZALEX + carfilzomib + dexamethasone; HR, hazard ratio; ISS, International Staging System; Kd, carfilzomib + dexamethasone; NE, not estimable; PFS, progression-free survival.
Safety
  • No new safety signals were identified with the longer follow-up.
  • In the safety population (D-Kd, n=308; Kd, n=153), any grade TEAEs occurred in 99.4% and 97.4% of patients in the D-Kd and Kd arm, respectively.
  • Grade ≥3 TEAEs occurred in 88.6% and 78.4% of patients in the D-Kd and Kd arm, respectively. The most common (≥15%) grade ≥3 TEAEs were thrombocytopenia (D-Kd, 24.7%; Kd, 16.3%), hypertension (D-Kd, 23.4%; Kd, 17.6%), pneumonia (D-Kd, 18.5%; Kd, 9.2%), and anemia (D-Kd, 17.5%; Kd, 16.3%).
  • Treatment-related fatal adverse events occurred in 2% of patients in the D-Kd arm and none in the Kd arm.

Daratumumab in Combination with Pomalidomide and Dexamethasone

APOLLO (MMY3013) is a randomized, open-label, multicenter, phase 3 study evaluating the safety and efficacy of D-Pd vs Pd with RRMM who received ≥1 prior treatment with both lenalidomide and a PI (N=304).44,45 Sonneveld et al (2021)46 presented updated efficacy and safety data at a median follow-up of 30.7 months. Results in the high-risk cytogenetic subgroup are summarized below.

Study Design/Methods

  • Primary endpoint: PFS
  • Secondary endpoints: ORR, ≥VGPR, ≥CR, MRD-negativity, OS, TTR, DOR, time to next therapy, safety, and health-related quality of life

Results


Key Baseline Characteristics in the ISS and Cytogenetic Profile Subgroups (APOLLO)46

D-Pd (n=151)
Pd (n=153)
ISS disease stagea, n (%)
   I
68 (45)
69 (45.1)
   II
50 (33.1)
51 (33.3)
   III
33 (21.9)
33 (21.6)
Cytogenetic profileb
   N
103
108
   Standard-risk, n (%)
64 (62.1)
73 (67.6)
   High-risk, n (%)
39 (37.9)
35 (32.4)
Abbreviations: D-Pd, DARZALEX FASPRO + pomalidomide + dexamethasone; ISS, International Staging System; Pd, pomalidomide + dexamethasone.
aBased on the combination of serum β2-microglobulin and albumin.
b
Based on fluorescence in situ hybridization. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality. Patients with standard cytogenetic risk had an absence of high-risk cytogenetic abnormalities.

Efficacy

PFS in the ISS Staging and Cytogenetic Profile Subgroups (APOLLO)46
Events/Patients
Median PFS, months
HR (95% CI)a
D-Pd
Pd
D-Pd
Pd
Overall
100/151
120/153
12.09
7.03
0.63 (0.48-0.83)
ISS disease staging
   I
40/68
50/69
19.32
10.12
0.66 (0.43-1)
   II
36/50
41/51
12.25
6.08
0.52 (0.33-0.82)
   III
24/33
29/33
6.05
5.03
0.67 (0.39-1.17)
Revised ISS disease staging
   I
14/26
19/25
17.54
11.17
0.61 (0.30-1.22)
   II
54/74
72/88
12.25
6.47
0.58 (0.41-0.83)
   III
16/19
12/14
2.83
3.38
1.17 (0.55-2.51)
Cytogenetic risk at study entry
   High-risk
32/39
28/35
5.78
3.98
0.88 (0.53-1.46)
   Standard-risk
39/64
58/73
17.54
8.54
0.56 (0.37-0.84)
Abbreviations: CI, confidence interval; D-Pd, DARZALEX FASPRO + pomalidomide + dexamethasone; HR, hazard ratio; ISS, International Staging System; Pd, pomalidomide + dexamethasone; PFS, progression-free survival.
aHRs and 95% CIs were calculated from a Cox proportional hazards model with treatment as the sole explanatory variable. An HR <1 indicates an advantage for D-Pd.

Safety
  • Grade ≥3 TEAEs were reported in 133 (89.3%) D-Pd-treated patients vs 123 (82%) Pd-treated patients.
  • Grade 3/4 TEAEs occurring in ≥15% of patients in the D-Pd vs Pd arm were neutropenia (69.1% vs 50.7%), infections (31.5% vs 23.3%), anemia (18.1% vs 21.3%), thrombocytopenia (18.1% vs 18.7%), and leukopenia (16.8% vs 4.7%).
  • Treatment discontinuation due to TEAEs was low among both treatment groups (D-Pd, 2%; Pd, 4%).

CLINICAL DATA - COMBINED NDMM AND RRMM ANALYSIS

Meta-Analysis ALCYONE, MAIA, CASSIOPEIA, CASTOR, POLLUX, CANDOR

Giri et al (2020)48 conducted a systematic review and meta-analysis of 6 randomized clinical studies (ALCYONE, MAIA, CASSIOPEIA, CASTOR, POLLUX, CANDOR) which evaluated the efficacy of DARZALEX with backbone MM regimens in newly diagnosed or relapsed/refractory patients with HRMM.

Study Design/Methods

  • The search strategy identified 6 phase 3 randomized clinical studies for the treatment of patients with NDMM or RRMM. The clinical studies included: ALCYONE, MAIA, CASSIOPEIA for NDMM and CASTOR, POLLUX, and CANDOR for RRMM.
  • HRMM was defined as the presence of t(4;14), t(14;16), or del(17p).
  • Primary outcome: PFS
  • All clinical studies included a preplanned subgroup analysis based on cytogenetic group, the latter was used as a stratification variable during randomization only in the CASSIOPEIA study; the distribution of patients with high-risk disease was well balanced between the 2 groups in all the other studies.

Results

  • The meta-analysis included 4061 patients from 6 phase 3 clinical studies.
    • NDMM: 358 patients with HRMM of 2528 patients from the ALCYONE, MAIA and CASSIOPEIA studies.
    • RRMM: 222 patients with HRMM of 1533 patients from the CASTOR, POLLUX, and CANDOR studies.
  • HRMM in patients with NDMM:
  • The HRs for PFS in the most recent follow-up data for ALCYONE, CASSIOPEIA, and MAIA were 0.78 (95% CI, 0.43-1.43), 0.67 (95% CI, 0.35-1.30), and 0.57 (95% CI, 0.32-1.04), respectively.
    • The pooled HR among NDMM patients with HRMM was 0.67 (95% CI, 0.47-0.95; P=0.02)
  • HRMM in patients with RRMM:
    • The HRs for PFS in the most recent follow-up data for CASTOR, POLLUX, and CANDOR were 0.41 (95% CI, 0.21-0.83), 0.37 (95% CI, 0.18-0.76), and 0.58 (95% CI, 0.3-1.12), respectively.
    • The pooled HR among RRMM patients with HRMM was 0.45 (95% CI, 0.30-0.67; P<0.001)
  • Mature OS data for patients with HRMM were only reported for the ALCYONE study (HR, 0.91; 95% CI, 0.50-1.65); pooled OS data was not reported because mature OS data cannot be stratified by cytogenetic group for the included studies at this time.

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 17 February 2025.

References

1 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.  
2 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.  
3 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.  
4 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.  
5 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.  
6 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 17]. Available from: https://clinicaltrials.gov/ct2/show/NCT03652064 NML Identifier: NCT03652064.  
7 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.  
8 Badros A, Foster L, Anderson LD Jr, et al. Daratumumab with lenalidomide as maintenance after transplant in newly diagnosed multiple myeloma: the AURIGA study. Blood. 2024;In press.  
9 Badros A, Foster L, Anderson LD Jr, et al. Subcutaneous daratumumab plus lenalidomide versus lenalidomide alone as maintenance therapy in newly diagnosed multiple myeloma after transplant: primary results from the phase 3 AURIGA study. Oral Presentation presented at: The 21st International Myeloma Society (IMS) Annual Meeting; September 25-28, 2024; Rio de Janeiro, Brazil.  
10 Janssen Research & Development, LLC. A study of daratumumab plus lenalidomide versus lenalidomide alone as maintenance treatment in participants with newly diagnosed multiple myeloma who are minimal residual disease positive after frontline autologous stem cell transplant (AURIGA). In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000- [cited 2025 February 17]. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03901963 NLM Identifier: NCT03901963.  
11 Badros A, Foster L, Anderson LD Jr, et al. Supplement to: Daratumumab with lenalidomide as maintenance after transplant in newly diagnosed multiple myeloma: the AURIGA study. Blood. 2024;In press.  
12 Foster L, Anderson LD Jr, Chung A, et al. Daratumumab plus lenalidomide (D-R) versus lenalidomide (R) alone as maintenance therapy in newly diagnosed multiple myeloma (NDMM) after transplant: analysis of the phase 3 AURIGA study among clinically relevant subgroups. Oral Presentation presented at: The 66th American Society of Hematology (ASH) Annual Meeting and Exposition; December 7-10, 2024; San Diego, CA.  
13 Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med. 2019;380:2104-2115.  
14 Kumar SK, Moreau P, Bahlis N, et al. Daratumumab plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) alone in transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM): updated analysis of the phase 3 MAIA study. Poster presented at: The 64th American Society of Hematology (ASH) Annual Meeting & Exposition; December 10-13, 2022; New Orleans, LA.  
15 Moreau P, Facon T, Usmani S, et al. Daratumumab plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) in transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM): clinical assessment of key subgroups of the phase 3 MAIA study. Poster presented at: 64th American Society of Hematology (ASH) Annual Meeting and Exposition; December 10-13, 2022; New Orleans, LA/Virtual meeting.  
16 Mateos MV, Dimopoulos MA, Cavo M, et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med. 2018;378(6):518-528.  
17 Mateos M, San-Miguel J, Cavo M, et al. Daratumumab plus bortezomib, melphalan, and prednisone (D-VMP) versus bortezomib, melphalan, and prednisone (VMP) alone in transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM): updated analysis of the phase 3 ALCYONE study. Poster presented at: The 64th American Society of Hematology (ASH) Annual Meeting & Exposition; December 10-13, 2022; New Orleans, LA.  
18 Jakubowiak AJ, Kumar S, Medhekar R, et al. Daratumumab Improves Depth of Response and Progression-free Survival in Transplant-ineligible, High-risk, Newly Diagnosed Multiple Myeloma. Oncol. 2022;27(7):oyac067-.  
19 Fu W, Bang SM, Huang H, et al. Bortezomib, melphalan, and prednisone with or without daratumumab in transplant-ineligible Asian patients with newly diagnosed multiple myeloma: the phase 3 OCTANS study. Clin Lymphoma Myeloma Leuk. 2023;23:446-455 e4.  
20 Fu W, Bang SM, Huang H, et al. Supplement to: Bortezomib, melphalan, and prednisone with or without daratumumab in transplant-ineligible Asian patients with newly diagnosed multiple myeloma: the phase 3 OCTANS study. Clin Lymphoma Myeloma Leuk. 2023;23(6):446-455 e4.  
21 Hou J, Fu W, Bang SM, et al. Daratumumab, bortezomib, melphalan, and prednisone versus bortezomib, melphalan, and prednisone in transplant-ineligible patients with newly diagnosed multiple myeloma: pooled analysis of OCTANS and ALCYONE. Poster presented at: 63rd American Society of Hematology (ASH) Annual Meeting & Exposition; December 11-14, 2021; Atlanta, GA/Virtual.  
22 Fu W, Bang SM, Huang H, et al. Subgroup analyses of progression-free survival from the phase 3 OCTANS and ALCYONE studies in transplant-ineligible patients with newly diagnosed multiple myeloma treated with bortezomib, melphalan, and prednisone with or without daratumumab. Poster presented at: 65th American Society of Hematology (ASH) Annual Meeting & Exposition; December 9-12, 2023; San Diego, CA.  
23 Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet. 2019;394(10192):29-38.  
24 Moreau P, Hulin C, Perrot A, et al. Maintenance with daratumumab or observation following treatment with bortezomib, thalidomide, and dexamethasone with or without daratumumab and autologous stem-cell transplant in patients with newly diagnosed multiple myeloma (CASSIOPEIA): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22(10):1378-1390.  
25 Sonneveld P, Attal M, Perrot A, et al. Daratumumab plus bortezomib, thalidomide, and dexamethasone (D-VTd) in transplant-eligible newly diagnosed multiple myeloma (NDMM): subgroup analysis of high-risk patients (pts) in CASSIOPEIA. Oral presentation presented at: 17th International Myeloma Workshop (IMW); September 12-15, 2019; Boston, MA.  
26 Voorhees PM, 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 59th American Society of Hematology (ASH) Annual Meeting & Exposition; December 9-12, 2017; Atlanta, GA.  
27 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.  
28 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.  
29 Chari A, Kaufman JL, Laubach J, et al. Daratumumab in transplant-eligible patients with newly diagnosed multiple myeloma: final analysis of clinically relevant subgroups in GRIFFIN. Blood Cancer J. 2024;14(1).  
30 Chari A, Kaufman JL, Laubach J, et al. Supplement to: Daratumumab in transplant-eligible patients with newly diagnosed multiple myeloma: final analysis of clinically relevant subgroups in GRIFFIN. Blood Cancer J. 2024;14(1).  
31 Costa LJ, Chhabra S, Medvedova E, et al. Minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma (MASTER): final report of the multicentre, single-arm, phase 2 trial. Lancet Haematol. 2023;10(11):e890-e901.  
32 Costa LJ, Chhabra S, Medvedova E, et al. Supplement to: Minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma (MASTER): final report of the multicentre, single-arm, phase 2 trial. [published online ahead of print september 27, 2023]. Lancet Haematol. doi:10.1016/s2352-3026(23)00236-3.  
33 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.  
34 Palumbo A, Chanan-Khan A, Weisel K, et al. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med. 2016;375(8):754-766.  
35 Sonneveld P, Chanan-Khan A, Weisel K, et al. Overall survival with daratumumab, bortezomib, and dexamethasone in previously treated multiple myeloma (CASTOR): a randomized, open-label, phase III trial. J Clin Oncol. 2023;41(8):1600-1609.  
36 Weisel K, Spencer A, Lentzsch S, et al. Daratumumab, bortezomib, and dexamethasone in relapsed or refractory multiple myeloma: subgroup analysis of CASTOR based on cytogenetic risk. J Hematol Oncol. 2020;13(1):115.  
37 Dimopoulos M, Oriol A, Nahi H, et al. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 2016;375(14):1319-1331.  
38 Dimopoulos M, Oriol A, Nahi H, et al. Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with previously treated multiple myeloma: overall survival results from the phase 3 POLLUX trial. Poster presented at: 3rd European Myeloma Network (EMN) Meeting; April 6-9, 2022; Virtual.  
39 Kaufman J, Dimopoulos M, White D, et al. Daratumumab, lenalidomide, and dexamethasone in relapsed/refractory myeloma: a cytogenetic subgroup analysis of POLLUX. Blood Cancer J. 2020;10(11):111.  
40 Mateos M, Richardson P, Weisel K, et al. Daratumumab (DARA) plus bortezomib and dexamethasone (D-Vd) or lenalidomide and dexamethasone (D-Rd) in relapsed or refractory multiple myeloma (RRMM): subgroup analyses of CASTOR and POLLUX. Poster presented at: 19th International Myeloma Society (IMS) Annual Meeting; August 25-27, 2022; Los Angeles, CA.  
41 Usmani S, Quach H, Mateos M, et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for the treatment of patients with relapsed or refractory multiple myeloma: results of the randomized phase 3 study CANDOR (NCT03158688). Oral presentation presented at: 61st American Society of Hematology (ASH) Annual Meeting and Exposition; December 7-10, 2019; Orlando, FL.  
42 Dimopoulos M, Quach H, Mateos MV, et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study. Lancet. 2020;396(10245):186-197.  
43 Usmani S, Quach H, Mateos M, et al. Supplement to: Final analysis of carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone in the CANDOR Study. Blood Adv. 2023;7(14):3739-3748.  
44 Sonneveld P, Terpos E, Dimopoulos M, et al. Pomalidomide and dexamethasone (Pom-Dex) with or without daratumumab (DARA) in patients (pts) with relapsed or refractory multiple myeloma (RRMM): a multicenter, randomized, phase 3 study (APOLLO). Poster presented at: The Annual Meeting of the American Society of Clinical Oncology (ASCO); June 1-5, 2018; Chicago, IL.  
45 Dimopoulos M, Terpos E, Boccadoro M, et al. Daratumumab plus pomalidomide and dexamethasone versus pomalidomide and dexamethasone alone in previously treated multiple myeloma (APOLLO): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22(6):801-812.  
46 Sonneveld P, Terpos E, Boccadoro M, et al. Pomalidomide and dexamethasone with or without subcutaneous daratumumab in patients with relapsed or refractory multiple myeloma: updated analysis of the phase 3 APOLLO study. Poster presented at: 63rd American Society of Hematology (ASH) Annual Meeting & Exposition; December 11-14, 2021; Atlanta, GA/Virtual.  
47 A D Meletios, Evangelos T, S D, et al. Subcutaneous daratumumab plus pomalidomide and dexamethasone (D-Pd) versus pomalidomide and dexamethasone (Pd) alone in patients with relapsed or refractory multiple myeloma (RRMM): overall survival results from the phase 3 APOLLO study. Poster presented at: 64th American Society of Hematology (ASH) Annual Meeting and Exposition; December 10-13, 2022; New Orleans, LA.  
48 Giri S, Grimshaw A, Bal S, et al. Evaluation of daratumumab for the treatment of multiple myeloma in patients with high-risk cytogenetic factors: a systematic review and meta-analysis. Published online September 24, 2020. JAMA Oncol. 2020;6(11):1759-1765.