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 - MMY3008 (MAIA) Study

Last Updated: 07/05/2024

A screenshot of a computer

Description automatically generated

Click on the following links to related sections within the document: MAIA (MMY3008) Study Overview, Prespecified Interim Results, and Updated Results (64.5 Month Follow-up).
Abbreviations
: AE, adverse event; CI, confidence interval; CR, complete response; CrCl, creatinine clearance; Dara, daratumumab; D-Rd, daratumumab + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; HR, hazard ratio; IRR, infusion-related reaction; IV, intravenous; MRD, minimal residual disease; NA, not applicable; NDMM, newly diagnosed multiple myeloma; NR, not reached; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; PFS2, PFS on the next line of therapy; Rd, lenalidomide + dexamethasone; sCR, stringent complete response; SPM, second primary malignancy; TEAE, treatment-emergent adverse event; VGPR, very good partial response.
aFacon (2019)1. bFacon (2018)2. cIn either treatment arm. dFacon (2024)4. eAll events were related to the general health condition of the patient.

SUMMARY  

  • MAIA is a phase 3 study evaluating the safety and efficacy of DARZALEX in combination with lenalidomide and dexamethasone (D-Rd) compared to lenalidomide and dexamethasone (Rd) in transplant-ineligible newly diagnosed multiple myeloma (NDMM) patients.1,2
    • Facon et al (2019)1 reported the pre-specified interim results of this ongoing study which indicated a 44% reduction in the risk of progression or death with D-Rd (hazard ratio [HR], 0.56; 95% confidence interval [CI], 0.43-0.73; P<0.001). Refer to Table: Most Common Adverse Events and Second Primary Cancers in the Safety Population for reported adverse event (AE) information.
    • Facon et al (2024)4 presented (at the European Hematology Association [EHA] Hybrid Congress) the results of the updated efficacy and safety analysis of the MAIA study. At a median follow-up of 89.3 months (range, 0-102.2), a 33% reduction in the risk of death was observed with the D-Rd vs Rd arms. Median OS was reached for the D-Rd arm and was prolonged for patients in the D-Rd vs Rd arms (90.3 vs 64.1 months [HR, 0.67; 95% CI, 0.55-0.82; nominal P<0.0001]). The median time to subsequent antimyeloma therapy was 42.4 months in the Rd arm and not reached (NR) in the D-Rd arm (HR, 0.51; 95% CI, 0.41-0.63; nominal P<0.0001). Deaths were reported for 47.5% (n=173) of patients in the D-Rd arm and 59.7% (n=218) of patients in the Rd arm, mostly due to disease progression.
    • Kumar et al (2022)3 presented (at the 64th American Society of Hematology [ASH] Annual Meeting and Exposition) the results of the updated efficacy and safety analysis of the MAIA study. At a median follow-up of 64.5 months, progression-free survival (PFS) improved with D-Rd vs Rd treatment. The overall response rate (ORR) and minimal residual disease (MRD)-negativity rate were also higher in the D-Rd vs Rd treatment. At a median follow-up of 73.6 months, a 35% reduction in the risk of death was observed with D-Rd vs Rd treatment. The most common grade 3/4 treatment-emergent adverse events (TEAEs; ≥20%) in any arm were neutropenia (DRd, 54.1%; Rd, 37.0%) and anemia (D-Rd, 17.0%; Rd, 21.6%).
  • Moreau et al (2022)5 presented (at the 64th ASH Annual Meeting and Exposition) the efficacy and safety results in clinically important subgroups of patients from the MAIA study. The PFS duration was longer in the D-Rd vs Rd arm in the majority of subgroups. Treatment with D-Rd generally resulted in a greater improvement in ORR, MRDnegativity (10-5) rate, and sustained MRD-negativity (10-5) rate for ≥12 months across subgroups, compared with Rd. Among patients aged ≥75 years, grade 3/4 TEAEs were reported in 95.5% vs 95.0% 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 (DRd, 21.0%; Rd, 12.6%), anemia (D-Rd, 20.4%; Rd, 25.2%), and pneumonia (DRd, 20.4%; Rd, 14.5%).
  • Facon et al (2022)6 reported a post hoc subgroup analysis of frailty status in patients with NDMM ineligible for transplant in the MAIA study. Median PFS benefit was observed in the D-Rd vs Rd arm across all frailty subgroups. The most common serious TEAE was pneumonia and the most common reasons for treatment discontinuation across all frailty subgroups were progressive disease (PD) and AEs.
  • Facon et al (2022)7 presented (at the 64th ASH Annual Meeting and Exposition) the efficacy results of the MAIA study according to age group (<70 years, ≥70 to <75 years, and <75 years) at a median follow-up of 64.5 months. All evaluated survival and response endpoints (including PFS, overall survival [OS], ORR, complete response [CR] or better, stringent complete response [sCR], very good partial response [VGPR] or better, and MRDnegativity [10-5]) were more favorable in the D-Rd arm than the Rd arm in patients aged <75 years, with the most pronounced PFS and OS benefit observed in patients aged <70 years.
  • Jakubowiak et al (2022)8 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 high-risk cytogenetic abnormalities (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. ≥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).
  • Perrot et al (2022)9 presented (at the 64th ASH Annual Meeting and Exposition) a post hoc analysis of patient-reported outcomes (PROs) in a subgroup of frail patients from the MAIA study (median follow-up, 64.5 months). In the D-Rd arm, larger (≥20 point) reductions in least square (LS) mean pain scores, moderate reductions in fatigue scores, and similar improvements in global health status (GHS) scores were reported compared with the Rd arm. Overall, the median time to worsening of the health-related quality of life (HRQoL) scores were longer in the D-Rd vs Rd arm.
  • Facon et al (2022)10 presented (at the 58th American Society of Clinical Oncology [ASCO] Annual Meeting) the results of a subgroup analysis of the phase 3 MAIA study that evaluated the efficacy and PROs of D-Rd vs Rd in subgroups of patients with NDMM who had renal impairment and/or high-risk cytogenetics.
  • San-Miguel et al (2022)11 presented an analysis of the predictive and prognostic role of MRD negativity and durability in patients treated in the MAIA study. Patients receiving the D-Rd regimen achieved higher rates of MRD-negativity and durability compared with those receiving Rd.
  • Moreau et al (2022)12 presented (at the 19th International Myeloma Society [IMS] Annual Meeting) the results of a post hoc analysis of the phase 3 MAIA study to evaluate the impact of treatment duration on long-term clinical outcomes. The median OS in the D-Rd arm was NR vs 20.5 months in patients who received treatment for ≥18 months vs <18 months, respectively. In patients who received treatment for ≥18 months, PFS benefit (HR, 0.57; 95% CI, 0.43-0.76; P<0.0001) and OS benefit (HR, 0.68; 95% CI, 0.47-0.98; P=0.0379) was observed in D-Rd vs Rd arms. Grade 3/4 TEAEs were reported in 96.5% vs 91.2% patients who received treatment for ≥18 months in the D-Rd vs Rd arm, respectively. The most common (≥15% in either treatment arm) grade 3/4 TEAEs were neutropenia (D-Rd, 56.9%; Rd, 41.2%), anemia (D-Rd, 17%; Rd, 18.1%), lymphopenia (D-Rd, 17%; Rd, 12.3%), pneumonia (D-Rd, 19.8%; Rd, 10.8%), and cataract (D-Rd, 13.8%; Rd, 18.6%).
  • Perrot et al (2021)13 presented (at the 63rd ASH Annual Meeting & Exposition) updated PRO analyses from the MAIA study at a median follow-up of 56.2 months.
  • Perrot et al (2021)14 presented (at the 63rd ASH Annual Meeting & Exposition) results of post hoc analyses of the MAIA study, which assessed the association between key clinical efficacy endpoints and PROs.
  • Usmani et al (2021)15 presented (at the 63rd ASH Annual Meeting & Exposition) on the impact of impaired renal function and lenalidomide starting dose in patients treated in the MAIA study.
  • Moreau et al (2020)16 presented (at the 8th Annual Meeting of the Society of Hematologic Oncology [SOHO]) safety and efficacy results of patients on DARZALEX for intravenous (IV) use that discontinued lenalidomide with or without dexamethasone (R±d) in the MAIA study. This analysis is listed in the References section below.
  • Other relevant literature has been identified in addition to the data summarized above.17-19

PRODUCT LABELING

CLINICAL DATA

MAIA (MMY3008; clinicaltrials.gov identifier: NCT02252172) is an ongoing, international, phase 3, randomized, open-label, active-controlled, multicenter study in patients with NDMM not eligible for high-dose chemotherapy and autologous stem cell transplantation (ASCT; N=737).1,2 Facon et al (2019)1 reported the prespecified interim results of this ongoing study, which is summarized below. Bahlis et al (2019)20 presented updated safety and efficacy results from an additional 9-month follow-up in the MAIA study. Kumar et al (2020)21 presented updated safety and efficacy results after approximately 4 years of follow-up in the MAIA study. Facon et al (2021)22 reported the updated safety and efficacy results of the MAIA study at the pre-specified interim OS analysis with a median follow-up of 56.2 months. Kumar et al (2022)3 presented the results of an updated efficacy and safety analysis at a median follow-up of 64.5 months. Facon et a (2024)4 presented updated efficacy and safety results at a median follow-up of 89.3 months.

Study Design/Methods

  • Phase 3, multicenter, randomized, open-label, active-controlled (Rd arm), international study.
  • Key eligibility criteria: documented NDMM, ineligible for high-dose chemotherapy with ASCT due to age (≥65 years) or due to the presence of comorbidities impacting ASCT tolerability, Eastern Cooperative Oncology Group performance status (ECOG PS) 0-2, creatinine clearance (CrCl) ≥30 mL/min.
  • Patients were randomized 1:1 to D-Rd (n=368) or Rd (n=369) and received 28-day cycles of:
    • Patients in the Rd arm were administered until PD or unacceptable safety event:
      • Lenalidomide 25 mg orally (PO) daily on days 1-21 until PD (10 mg daily if CrCl between 30-50 mL/min).
      • Dexamethasone: 40 mg PO or IV weekly on days 1, 8, 15, and 22, until PD
        (20 mg weekly in patients >75 years of age or with a body mass index [BMI] <18.5 kg/m2).
    • Patients in the D-Rd arm were administered until PD or unacceptable safety event the same as above along with:
      • DARZALEX 16 mg/kg IV weekly during cycles 1-2, every 2 weeks during cycles 36, then every 4 weeks during cycle 7+. Following a protocol amendment (2020), patients in the D-Rd arm were given the option to switch from DARZALEX IV to DARZALEX FASPRO for subcutaneous (SC) use. DARZALEX FASPRO was to be administered at a fixed dose of 1800 mg SC over 3-5 minutes in the abdominal SC tissue once every 4 weeks.
  • Dose modifications:  
    • Delays in DARZALEX treatment were permissible if patients experienced any of the following AEs that could not be attributed to lenalidomide: grade 4 hematologic events, grade 3 thrombocytopenia with bleeding, febrile neutropenia, neutropenia with infection (any grade), and grade ≥3 nonhematologic AEs (excluding: grade 3 nausea, vomiting, diarrhea that responded to either antiemetics or antidiarrheal treatment within a week and grade 3 fatigue/asthenia that was existing at baseline or that lasted for <7 days after the last DARZALEX administration).22
  • Patients in the D-Rd arm were administered the following pre-infusion medications:22
    • Acetaminophen 650-1000 mg IV/PO
    • Diphenhydramine 25-50 mg (or equivalent) IV/PO
    • Dexamethasone 40 mg IV/PO approximately 1 hours prior to DARZALEX infusion
      (20 mg once weekly for patients >75 years of age or with a BMI <18.5 kg/m2)
  • For patients with a higher risk of respiratory complications such as mild asthma or chronic obstructive pulmonary disease who have forced expiratory volume in
    1 second <80% (FEV1); could receive the following post-infusion mediations: diphenhydramine (or equivalent), short-acting ß2 adrenergic receptor agonists, control medications for lung disease.22
  • Serum and urine samples were collected at screening, on day 1 of every cycle for
    2 years and every 8 weeks thereafter until PD.
  • Safety was monitored throughout the study until 30 days after the last dose of the study treatment.22
  • Cytogenetic risk was evaluated locally by fluorescence in situ hybridization (FISH) or karyotype testing. High-risk cytogenetics was determined by a deletion (del)17p and/or translocations t(14;16), or t(4;14).
  • Primary endpoint: PFS
  • Secondary endpoints: ≥CR rate, duration of response, ≥VGPR rate; MRD-negativity rate, ORR, OS, time from randomization to disease progression on the next line of therapy or death (PFS2), safety, sCR, time to next (2nd-line) treatment, time to response, and time to progression

Results

Efficacy
  • Median follow-up: 28.0 months (range, 0.0-41.4)
  • Patients (52% male) ranged in age from 45-90 years (median, 73 years; 70-<75 years, 35%; ≥75 years, 44%), with key baseline characteristics being well-balanced between the arms.
  • HR for the primary endpoint of PFS: 0.56 (95% CI, 0.43-0.73; P<0.001) indicating a 44% reduction in the risk of progression or death with D-Rd.
  • Median time to first response was 1.05 months in both arms.
  • Median PFS: 31.9 months for Rd (95% CI, 28.9-NR); NR for D-Rd
    • PFS was maintained among patients ≥75 years (HR, 0.63; 95% CI, 0.44-0.92).
  • ORR: 92.9% (95% CI, 89.8-95.3) for D-Rd vs 81.3% (95% CI, 76.9-85.1) for Rd (P<0.001)
  • ≥CR was achieved in 175 (47.6%) patients in the D-Rd arm vs 92 (24.9%) patients in the Rd arm (P<0.001).
  • Median time to ≥CR was 10.4 months for D-Rd and 11.2 months for Rd.
  • ≥VGPR of 79.3% was reported for D-Rd vs 53.1% for Rd (P<0.001).
  • MRD-negativity was >3 times higher with D-Rd than with Rd (89 [24.2%] vs 27 [7.3%]; P<0.001).
  • HR for OS: 0.78 (95% CI, 0.56-1.1); data immature after a median follow-up of
    28.0 months
Safety
  • Serious AEs: 62.9% for D-Rd; 62.7% for Rd
  • AEs leading to discontinuation: 7.1% for D-Rd; 15.9% for Rd
  • TEAEs with outcome of death: 6.9% for D-Rd; 6.3% for Rd
    • Pneumonia was the most common AE that resulted in death (D-Rd, 0.5%; Rd, 0.8%).

Most Common Adverse Events and Second Primary Cancers in the Safety Populationa, 1
Event, n (%)
D-Rd (n=364)
Rd (n=365)
Any Grade
Grade 3/4
Any Grade
Grade 3/4
Hematologic
   Neutropenia
207 (56.9)
182 (50.0)
154 (42.2)
129 (35.3)
   Anemia
126 (34.6)
43 (11.8)
138 (37.8)
72 (19.7)
   Leukopenia
68 (18.7)
40 (11.0)
34 (9.3)
18 (4.9)
   Lymphopenia
66 (18.1)
55 (15.1)
45 (12.3)
39 (10.7)
Nonhematologic
   Infections
314 (86.3)
117 (32.1)
268 (73.4)
85 (23.3)
      Pneumonia
82 (22.5)
50 (13.7)
46 (12.6)
29 (7.9)
   Diarrhea
207 (56.9)
24 (6.6)
168 (46.0)
15 (4.1)
   Constipation
149 (40.9)
6 (1.6)
130 (35.6)
1 (0.3)
   Fatigue
147 (40.4)
29 (8.0)
104 (28.5)
14 (3.8)
   Peripheral edema
140 (38.5)
7 (1.9)
107 (29.3)
2 (0.5)
   Back Pain
123 (33.8)
11 (3.0)
96 (26.3)
11 (3.0)
   Asthenia
117 (32.1)
16 (4.4)
90 (24.7)
13 (3.6)
   Nausea
115 (31.6)
5 (1.4)
84 (23.0)
2 (0.5)
Second primary cancerb
32 (8.8)
NA
26 (7.1)
NA
   Invasive second primary cancer
12 (3.3)
NA
13 (3.6)
NA
Any infusion-related reaction
149 (40.9)
10 (2.7)
NA
NA
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; NA, not applicable; Rd, lenalidomide + dexamethasone.
aThe safety population included all patients who received at least one dose of the trial treatment. Adverse events of any grade that were reported in >30% of patients in either treatment arm and grade 3/4 adverse events that were reported in >10% of patients in either treatment arm are listed.
bThe presence of a second primary cancer was prespecified in the statistical analysis plan as an adverse event of clinical interest.

Final Survival Analysis of the MAIA Study

Facon et al (2024)4 presented the updated OS results for the D-Rd vs Rd arm at a long-term median follow-up of around 7.5 years.

Results

Patient Characteristics

Demographic and Baseline Disease Characteristics of the ITT Populationa,4
Characteristic
D-Rd
(n=368)

Rd
(n=369)

Age
   Median age (range), years
73 (50-90)
74 (45-89)
   ≥75 years, n (%)
160 (43.5)
161 (43.6)
Male, n (%)
189 (51.4)
195 (52.8)
ECOG PS, n (%)
   0
127 (34.5)
123 (33.3)
   1
178 (48.4)
187 (50.7)
   ≥2
63 (17.1)
59 (16.0)
ISS disease stage, n (%)
   I
98 (26.6)
103 (27.9)
   II
163 (44.3)
156 (42.3)
   III
107 (29.1)
110 (29.8)
Type of measurable disease, n (%)
   IgG
225 (61.1)
231 (62.6)
   IgA
65 (17.7)
66 (17.9)
   Otherb
9 (2.4)
10 (2.7)
   Detected in urine only
40 (10.9)
34 (9.2)
   Detected as serum FLC only
29 (7.9)
28 (7.6)
Cytogenetic riskc, n (%)
   n
319
323
   Standard risk
271 (85.0)
279 (86.4)
   High risk
48 (15.0)
44 (13.6)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; FLC, free light chain; Ig, immunoglobulin; ISS, International Staging System; ITT, intent-to-treat; Rd, lenalidomide/dexamethasone.
aThe ITT population included all randomized patients.
bInclusive of IgD, IgE, IgM, and biclonal disease.
cCytogenetic risk was based on fluorescence in situ hybridization or karyotype analysis; patients who had a high-risk cytogenetic profile had ≥1 of the following high-risk abnormalities: del(17p), t(14;16), or t(4;14).

Efficacy
  • At a median follow-up of 89.3 months (range, 0-102.2), the 7-year OS rate was 53.1% for the D-Rd arm and 39.3% for the Rd arm.
  • Median OS was reached for the D-Rd group and was prolonged for patients in the D-Rd vs Rd arm (90.3 vs 64.1 months [HR, 0.67; 95% CI, 0.55-0.82; nominal P<0.0001]). See Table: Analysis of OS in Pre-Specified Patient Subgroups.
  • Median time to subsequent antimyeloma therapy was 42.4 months in the Rd arm and NR in the D-Rd arm (HR, 0.51; 95% CI, 0.41-0.63; nominal P<0.0001). See Table: Summary of First Subsequent Antimyeloma Therapy in the Safety Population.
    • In the D-Rd vs Rd arm, 10.7% (15/140) vs 24.4% (49/201) of patients received DARZALEX-containing regimens as their first subsequent therapy.
    • Among treated patients, 38.5% (140/364) vs 55.1% (201/365) of patients in the D-Rd vs Rd arm received ≥1 subsequent antimyeloma therapy.
    • Across subsequent therapy lines, the most common antineoplastic agents in the D-Rd vs Rd arms were bortezomib (27.7% vs 41.9%), DARZALEX (6.3% vs 28.8%), and carfilzomib (7.7% vs 12.3%).
    • In patients evaluable for best response to first subsequent antimyeloma therapy, ≥CR was achieved by 4.6% (6/130) vs 4.1% (8/193) in the D-Rd vs Rd arm, and ≥VGPR was achieved by 13.8% (18/130) vs 23.8% (46/193) of patients in the D-Rd vs Rd arm.
    • No patient in either group reported the use of B-cell maturation antigen (BCMA)- or G protein–coupled receptor class C group 5 member D (GPRC5D)-targeted therapy.
    • Investigational drug was given to 2 patients in the D-Rd group and 2 patients in the Rd group in subsequent therapy lines.

Analysis of OS in Pre-Specified Patient Subgroupsa,4
Subgroup
D-Rd
Rd
HR (95% CI)b
n/N
Median OS, Months
n/N
Median OS, Months
Sex
   Male
95/189
82.5
120/195
60.6
0.72 (0.55-0.94)
   Female
80/179
NE
98/174
67.8
0.66 (0.49-0.89)
Age
   <75 years
84/208
NE
107/208
79.6
0.69 (0.52-0.92)
   ≥75 years
91/160
72.3
111/161
54.8
0.67 (0.51-0.88)
Race
   White
161/336
92.7
197/339
65.5
0.71 (0.57-0.87)
   Other
14/32
90.3
21/30
49.1
0.50 (0.25-0.99)
Region
   North America
46/101
92.7
64/102
54.8
0.57 (0.39-0.83)
   Other
129/267
90.3
154/267
66.8
0.74 (0.58-0.93)
Baseline renal function (CrCl)
   >60 mL/min
99/206
92.7
123/227
69.9
0.78 (0.60-1.01)
   ≤60 mL/min
76/162
90.3
95/142
54.4
0.57 (0.42-0.77)
Baseline hepatic function
   Normal
156/335
NE
203/340
63.8
0.65 (0.53-0.80)
   Impaired
19/31
63.5
15/29
87.4
1.31 (0.66-2.58)
ISS disease stage
   I
34/98
NE
42/103
NE
0.79 (0.50-1.24)
   II
77/163
92.7
95/156
61.7
0.63 (0.46-0.85)
   III
64/107
65.2
81/110
47.3
0.68 (0.49-0.95)
Type of MM
   IgG
111/225
87.2
132/231
69.3
0.78 (0.60-1.00)
   Non-IgG
35/74
86.4
49/76
53.7
0.58 (0.37-0.89)
Cytogenetic risk at study entryc
   High risk
31/48
55.6
36/44
42.5
0.65 (0.40-1.06)
   Standard risk
122/271
NE
160/279
65.5
0.66 (0.52-0.84)
ECOG PS
   0
48/127
NE
56/123
NE
0.76 (0.52-1.12)
   1
86/178
92.7
118/187
58.3
0.64 (0.48-0.84)
   ≥2
41/63
62.8
44/59
39.0
0.68 (0.44-1.04)
Abbreviations: CI, confidence interval; CrCl, creatinine clearance; del, deletion; D-Rd, DARZALEX + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; HR, hazard ratio; IgG, immunoglobulin G; ISS, International Staging System; MM, multiple myeloma; NE, not estimable; OS, overall survival; Rd, lenalidomide + dexamethasone; t, translocation.
aIn the ITT population, which included all randomized patients.
bHR and 95% CI from a Cox proportional hazards model with treatment as the sole explanatory variable. HR <1 indicates an advantage for D-Rd.
cCytogenetic risk was based on fluorescence in situ hybridization or karyotype analysis; patients who had a high-risk cytogenetic profile had ≥1 of the following high-risk abnormalities: del(17p), t(14;16), or t(4;14).


Summary of First Subsequent Antimyeloma Therapy in the Safety Populationa,4
n (%)
D-Rd
(n=140)

Rd
(n=201)

First subsequent therapy classb,c
   PI only
69 (49.3)
101 (50.2)
   IMiD only
22 (15.7)
25 (12.4)
   PI + IMiD
25 (17.9)
16 (8.0)
   DARZALEX monotherapy or combination
15 (10.7)
49 (24.4)
   Other
9 (6.4)
10 (5.0)
Most common first subsequent therapy regimensb,d
   Bortezomib/cyclophosphamide/dexamethasone
19 (13.6)
29 (14.4)
   Bortezomib/dexamethasone
20 (14.3)
28 (13.9)
   Bortezomib/melphalan/prednisone
14 (10.0)
28 (13.9)
   DARZALEX/bortezomib/dexamethasone
4 (2.9)
27 (13.4)
   Lenalidomide/dexamethasone
13 (9.3)
16 (8.0)
   Bortezomib/pomalidomide/dexamethasone
9 (6.4)
3 (1.5)
   Bortezomib/lenalidomide/dexamethasone
8 (5.7)
3 (1.5)
   DARZALEX/lenalidomide/dexamethasone
4 (2.9)
6 (3.0)
   Pomalidomide/dexamethasone
2 (1.4)
6 (3.0)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; IMiD, immunomodulatory drugs; PI, proteasome inhibitor; Rd, lenalidomide + dexamethasone.
aThe safety population included all randomized patients who received ≥1 dose of study treatment.
bPercentages were calculated with the number of patients who received subsequent therapy in each treatment group as the denominator.
cTherapy classes are mutually exclusive. Patients in any therapy class subgroup may have received additional agents (other than PI, IMiD, or DARZALEX), such as dexamethasone.
dRegimens received by ≥3% of patients in either treatment group.

Safety and Tolerability
  • Among the safety population, 78.3% (n=285) of patients in the D-Rd arm and 94.5% (n=345) in the Rd arm discontinued study treatment.
    • Progressive disease was the primary reason for discontinuation in both the D-Rd (32.7%) and Rd arms (38.6%).
    • A lower proportion of patients in the D-Rd (16.5%) and Rd arms (25.8%) discontinued study treatment due to AEs.
  • In the D-Rd vs Rd arm, 33% reduction in the risk of death was reported.

Summary of Death and Causes of Death in the Safety Populationa,4
n (%)
D-Rd
(n=364)

Rd
(n=365)

Total number of patients who died during the study
173 (47.5)
218 (59.7)
   Primary cause of death
      Disease progression
76 (20.9)
88 (24.1)
      Adverse events
44 (12.1)
40 (11.0)
         Related to study treatmentb
14 (3.8)
10 (2.7)
         Unrelated to study treatment
28 (7.7)
29 (7.9)
         Othersc
53 (14.6)
90 (24.7)
         Infections/infestations
9 (2.5)
30 (8.2)
         General disorders/administration site conditionsd
11 (3.0)
5 (1.4)
         Neoplasms (benign, malignant, or unspecified)
11 (3.0)
4 (1.1)
         Cardiac disorders
1 (0.3)
8 (2.2)
         Nervous system disorders
3 (0.8)
5 (1.4)
         Unknown
13 (3.6)
27 (7.4)
Deaths within 30 days of last study treatment dose
31 (8.5)
35 (9.6)
   Primary cause of death
      Disease progression
1 (0.3)
1 (0.3)
      Adverse events
29 (8.0)
32 (8.8)
         Related to study treatmentb
11 (3.0)
10 (2.7)
         Unrelated to study treatment
18 (4.9)
22 (6.0)
         Othere
1 (0.3)
2 (0.5)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; Rd, lenalidomide/dexamethasone.aThe safety population included all randomized patients who received ≥1 dose of study treatment.bAdverse events were related to ≥1 of the 3 components of study treatment: DARZALEX, lenalidomide, and dexamethasone.
cOther reasons were reported in ≥1% of patients in either treatment group.
dAll events were related to the general health condition of the patient.
eIncludes a nervous system disorder in 1 patient in the D-Rd group and a blood and lymphatic system disorder and general disorder/administration site condition in 1 patient each in the Rd group.

Kumar et al (2022)3 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

Patient Characteristics

Summary of Patient Disposition in the ITT Population (MAIA)3
Parameter
D-Rd (n=368)a
Rd (n=369)a
Patients treated, n (%)b
364 (98.9)
365 (98.9)
Patients who discontinued treatment, n (%)c
233 (64.0)
311 (85.2)
Reason for discontinuation, n (%)
   Progressive disease
107 (29.4)
131 (35.9)
   Adverse event
57 (15.7)
89 (24.4)
   Death
24 (6.6)
25 (6.8)
   Noncompliance with study drug
21 (5.8)
30 (8.2)
   Patient withdrawal
2 (0.5)
8 (2.2)
   Physician decision
16 (4.4)
24 (6.6)
   Loss to follow-up
1 (0.3)
2 (0.5)
   Other
5 (1.4)
2 (0.5)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; ITT, intent-to-treat; Rd, lenalidomide + dexamethasone.
aData are based on a median follow-up of 64.5 months.
bPercentages are based on the number of patients randomized.
cPercentages are based on the number of patients treated.

Efficacy
  • At a median follow-up of 64.5 months, the median PFS in the D-Rd vs Rd arm was 61.9 vs 34.4 months (HR, 0.55; 95% CI, 0.45-0.67; P<0.0001).
    • The 60-month PFS rate in the D-Rd vs Rd arm was 52.1% vs 29.6%.
  • 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%.
    • OS benefit with D-Rd vs Rd treatment was generally consistent across the prespecified patient subgroups, see Table: OS in Prespecified Subgroups (MAIA).

OS in Prespecified Subgroups (MAIA)3
Subgroup
D-Rda
Rda
HR (95% CI)a, b
n/N
Median OS, Months
n/N
Median OS, Months
Sex
   Male
86/189
NE
114/195
60.6
0.70 (0.53-0.93)
   Female
64/179
NE
88/174
67.8
0.62 (0.45-0.85)
Age
   <75 years
71/208
NE
101/208
77.6
0.64 (0.47-0.86)
   ≥75 years
79/160
73.5
101/161
54.8
0.67 (0.50-0.90)
Race
   White
138/336
NE
181/339
65.4
0.69 (0.55-0.86)
   Other
12/32
NE
21/30
49.1
0.43 (0.21-0.88)
Region
   North America
40/101
NE
61/102
54.8
0.54 (0.36-0.80)
   Other
110/267
NE
141/267
66.4
0.71 (0.55-0.91)
Baseline renal function (CrCl)
   >60 mL/min
83/206
NE
113/227
69.7
0.72 (0.55-0.96)
   ≤60 mL/min
67/162
NE
89/142
54.8
0.56 (0.41-0.77)
Baseline hepatic function
   Normal
132/335
NE
188/340
63.8
0.62 (0.49-0.77)
   Impaired
18/31
63.5
14/29
73.8
1.29 (0.64-2.60)
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)
Type of MM
   IgG
93/225
NE
120/231
68.6
0.74 (0.56-0.97)
   Non-IgG
29/74
NE
46/76
53.7
0.54 (0.34-0.85)
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)
ECOG PS
   0
37/127
NE
49/123
NE
0.69 (0.45-1.06)
   1
76/178
NE
110/187
58.3
0.62 (0.47-0.84)
   ≥2
37/63
61.9
43/59
39.0
0.64 (0.41-1.00)
Abbreviations: CI, confidence interval; CrCl, creatinine clearance; del, deletion; D-Rd, DARZALEX + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; HR, hazard ratio; IgG, immunoglobulin G; ISS, International Staging System; MM, multiple myeloma; NE, not estimable; OS, overall survival; Rd, lenalidomide + dexamethasone; t, translocation.
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).

  • At a median follow-up of 64.5 months, the ORR, MRD-negativity rate, and rates of sustained MRD-negativity lasting ≥12 months and ≥18 months were higher with D-Rd vs Rd treatment; see Table: Summary of ORR, MRD-Negativity Rate, and Sustained MRD-Negativity Rates in the ITT Population (MAIA).
  • At a median follow-up of 73.6 months, the OS was improved in patients who were MRDnegative (D-Rd, 88.9%; Rd, 78.0%) vs patients who were MRD-positive (D-Rd, 55.9%; Rd, 50.4%) in both treatment arms; however, more patients in the D-Rd arm achieved MRD-negativity.

Summary of ORR, MRD-Negativity Rate, and Sustained MRD-Negativity Rates in the ITT Population (MAIA)3
Parameter
D-Rd (n=368)a
Rd (n=369)a
P Valuea
ORR, n (%)
342 (92.9)
301 (81.6)
<0.0001
   ≥CR
188 (51.1)
111 (30.1)
<0.0001
      sCR
131 (35.6)
58 (15.7)
<0.0001
      CR
57 (15.5)
53 (14.4)
-
   ≥VGPR
300 (81.5)
210 (56.9)
<0.0001
      VGPR
112 (30.4)
99 (26.8)
-
   PR
42 (11.4)
91 (24.7)
-
MRD-negative, n (%)
118 (32.1)
41 (11.1)
<0.0001
Sustained MRD-negative, n (%)
   Lasting ≥12 months
69 (18.8)
15 (4.1)
<0.0001
   Lasting ≥18 months
62 (16.8)
12 (3.3)
<0.0001
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.
aData are based on a median follow-up of 64.5 months.

  • At a median follow-up of 64.5 months, a total of 128 patients in the D-Rd arm and 194 patients in the Rd arm received subsequent therapy.
    • In the D-Rd vs Rd arm, 9.4% vs 23.2% of patients received DARZALEXcontaining treatment as the first subsequent therapy and 14.1% vs 48.5% of patients received DARZALEX-containing treatment in any subsequent line of therapy.

Safety


Summary of Any-Grade (≥30%) and Grade 3/4 (≥20%) TEAEs in the Safety Population (MAIA)a, 3
Event, n (%)
D-Rd (n=364)
Rd (n=365)
Any-Grade
Grade 3/4
Any-Grade
Grade 3/4
Hematologic
   Neutropenia
224 (61.5)
197 (54.1)
166 (45.5)
135 (37.0)
   Anemia
154 (42.3)
62 (17.0)
150 (41.1)
79 (21.6)
Nonhematologic
   Diarrhea
240 (65.9)
33 (9.1)
188 (51.5)
22 (6.0)
   Fatigue
164 (45.1)
33 (9.1)
114 (31.2)
17 (4.7)
   Constipation
157 (43.1)
6 (1.6)
137 (37.5)
2 (0.5)
   Peripheral edema
155 (42.6)
10 (2.7)
117 (32.1)
3 (0.8)
   Back pain
155 (42.6)
14 (3.8)
109 (29.9)
14 (3.8)
   Asthenia
136 (37.4)
19 (5.2)
101 (27.7)
18 (4.9)
   Nausea
133 (36.5)
7 (1.9)
88 (24.1)
2 (0.5)
   Insomnia
125 (34.3)
11 (3.0)
116 (31.8)
14 (3.8)
   Bronchitis
124 (34.1)
12 (3.3)
87 (23.8)
7 (1.9)
   Pneumonia
113 (31.0)
71 (19.5)
66 (18.1)
39 (10.7)
   Cough
123 (33.8)
2 (0.5)
65 (17.8)
0 (0.0)
   Dyspnea
119 (32.7)
12 (3.3)
63 (17.3)
4 (1.1)
   Weight decreased
112 (30.8)
10 (2.7)
69 (18.9)
11 (3.0)
   Muscle spasms
111 (30.5)
2 (0.5)
86 (23.6)
5 (1.4)
   Peripheral sensory neuropathy
111 (30.5)
9 (2.5)
66 (18.1)
2 (0.5)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; Rd, lenalidomide + dexamethasone; TEAE, treatment-emergent adverse event.
aData are based on a median follow-up 64.5 months.

Analysis of Clinically Important Subgroups of MAIA

Moreau et al (2022)5 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 subgroups based on the following patient characteristics:
    • Age ≥75 years
    • International Staging System (ISS) stage III disease
    • Renal insufficiency (baseline CrCl ≤60 mL/min)
    • Extramedullary plasmacytomas
    • 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

Subgroup Analysis of PFS in the ITT Population of MAIA5
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
   Age ≥75 years
87/160
54.3
106/161
31.4
0.59 (0.44-0.79)
   ISS stage III
61/107
42.4
73/110
24.2
0.61 (0.43-0.86)
   Renal insufficiency
82/162
56.7
92/142
29.7
0.55 (0.41-0.75)
   Extramedullary plasmacytomas
7/15
57.5
5/9
19.4
0.47 (0.15-1.50)
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
56.0
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
40.0
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
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
24.0
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.


Subgroup Analysis of ORR in the ITT Population of MAIA5
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
   Age ≥75 years
144/160 (90.0)
131/161 (81.4)
2.06 (1.07-3.95)
   ISS stage III
93/107 (86.9)
86/110 (78.2)
1.85 (0.90-3.81)
   Renal insufficiency
146/162 (90.1)
112/142 (78.9)
2.44 (1.27-4.70)
   Extramedullary plasmacytomas
13/15 (86.7)
3/9 (33.3)
13.00 (1.70-99.37)
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 (75.0)
3.67 (1.07-12.55)
      Revised standard cytogenetic risk
162/176 (92.0)
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 (85.0)
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 (80.0)
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 (86.0)
3.36 (1.17-9.60)
      Gain(1q21) or amp(1q21) plus
      ≥1HRCA

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.


Subgroup Analysis of MRD-negativity (10-5) Rates in the ITT Population of MAIA5
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
   Age ≥75 years
43/160 (26.9)
16/161 (9.9)
3.33 (1.79-6.21)
   ISS stage III
29/107 (27.1)
12/110 (10.9)
3.04 (1.46-6.34)
   Renal insufficiency
48/162 (29.6)
11/142 (7.7)
5.01 (2.49-10.11)
   Extramedullary plasmacytomas
5/15 (33.3)
0/9 (0)
NE (NE-NE)
Cytogenetic risk
      Standard cytogenetic risk
93/271 (34.3)
33/279 (11.8)
3.89 (2.50-6.06)
      High cytogenetic risk
12/48 (25.0)
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.


Subgroup Analysis of Rates of Sustained MRD-Negativity (10-5) Lasting ≥12 Months in the ITT Population5
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
   Age ≥75 years
22/160 (13.8)
5/161 (3.1)
4.97 (1.83-13.49)
   ISS stage III
17/107 (15.9)
3/110 (2.7)
6.74 (1.91-23.73)
   Renal insufficiency
30/162 (18.5)
2/142 (1.4)
15.91 (3.73-67.89)
   Extramedullary plasmacytomas
2/15 (13.3)
0/9 (0)
NE (NE-NE)
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 95.0% 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, 21.0%; 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.

Impact of Frailty on Safety and Efficacy in the MAIA Study

Facon et al (2022)6 reported a post hoc subgroup analysis of frailty status in patients with NDMM ineligible for transplant in the MAIA study.

Results

Patient Characteristics
  • Patients (D-Rd, n=368; Rd, n=369) were stratified based on frailty scores (assessed using age, Charlson comorbidity index, and baseline ECOG PS scores):
    • Frail patients: 172 (46.7%) in the D-Rd arm vs 169 (45.8%) in the Rd arm
    • Total non-frail (fit and intermediate combined) patients: 196 (53.3%) in the D-Rd arm vs 200 (54.2%) in the Rd arm
    • Intermediate patients: 128 (34.8%) in the D-Rd arm vs 122 (33.1%) in the Rd arm
    • Fit patients: 68 (18.5%) in the D-Rd arm vs 78 (21.1%) in the Rd arm
  • Median duration of follow-up was 36.4 months.
Efficacy
  • Median relative dose intensity (RDI) of DARZALEX was consistent across all frailty subgroups (≥98.0%).
  • Median RDI of lenalidomide was higher in the D-Rd arm (77.2% vs 65.4%) and lower in the Rd arm (86.4% vs 92.9%) for patients in the total non-frail vs frail subgroup, respectively.
  • Median PFS benefit was observed in all subgroups:
    • For frail patients, PFS was NR in the D-Rd arm vs 30.4 months in the Rd arm (HR, 0.62; 95% CI, 0.45-0.85; P=0.003).
    • For total non-frail patients, PFS was NR in the D-Rd arm vs 41.7 months in the Rd arm (HR, 0.48; 95% CI, 0.34-0.68; P<0.0001).
    • For intermediate patients, PFS was NR in both the D-Rd and Rd arms (HR, 0.53; 95% CI, 0.35-0.80; P=0.0024).
    • For fit patients, PFS was NR in the D-Rd arm vs 41.7 months in the Rd arm (HR, 0.41; 95% CI, 0.22-0.75; P=0.0028).
  • The 36-month PFS rate was higher in the D-Rd vs Rd arm, respectively, in all subgroups:
    • Frail patients: 61.5% vs 39.5%
    • Total non-frail patients: 73.2% vs 52.1%
    • Intermediate patients: 70.4% vs 51.7%
    • Fit patients: 78.3% vs 53.6%
  • ORR was 87.2% vs 78.1% (P=0.0265) in frail patients, 98.0% vs 84.5% (P<0.0001) in total non-frail patients, 96.9% vs 85.2% (P=0.0012) in intermediate patients, and 100.0% vs 83.3% (P=0.0004) in fit patients receiving D-Rd vs Rd, respectively.
  • The rate of achieving ≥CR was 43.6% vs 30.8% (P=0.0144) in frail patients, 54.6% vs 24.0% (P<0.0001) in total nonfrail patients, 53.9% vs 25.4% (P<0.0001) in intermediate patients, and 55.9% vs 21.8% (P<0.0001) in fit patients receiving D-Rd vs Rd, respectively.
  • The rate of achieving ≥VGPR was 74.4% vs 53.8% (P<0.0001) in frail patients, 85.2% vs 56.0% (P<0.0001) in total nonfrail patients, 84.4% vs 57.4% (P<0.0001) in intermediate patients, and 86.8% vs 53.8% (P<0.0001) in fit patients receiving D-Rd vs Rd, respectively.
  • The rate of MRD-negativity was 23.8% vs 10.1% (P=0.0008) in frail patients, 33.2% vs 8.5% (P<0.0001) in total non-frail patients, 32.8% vs 9.0% (P<0.0001) in intermediate patients, and 33.8% vs 7.7% (P<0.0001) in fit patients receiving D-Rd vs Rd, respectively.
Safety
  • Incidence of serious TEAE was higher in the frail subgroup vs all other subgroups. The most common serious TEAE in patients receiving D-Rd vs Rd, respectively, was pneumonia across all subgroups: frail, 17.9% vs 8.4%; total non-frail, 10.7% vs 9.0%; intermediate, 10.2% vs 9.8%; and fit, 11.8% vs 7.8%.
  • The most common (≥10%) grade 3/4 TEAEs and TEAEs with an outcome of death (>1 patient, safety population) are summarized in Table: Summary of Safety Data.

Summary of Safety Data6
Parameter, n (%)
Non-frail
Frail (n=334)
Fit (n=145)
Intermediate (n=250)
Total Non-frail (n=395)
D-Rd (n=
68)

Rd
(n=
77)

D-Rd
(n=
128)

Rd
(n=
122)

D-Rd
(n=
196)

Rd
(n=
199)

D-Rd
(n=
168)

Rd
(n=
166)

Total number of patients with grade 3/4 TEAEs
58 (85.3)
61 (79.2)
117 (91.4)
104 (85.2)
175 (89.3)
165 (82.9)
159 (94.6)
148 (89.2)
Hematologic TEAEs
   Neutropenia
30 (44.1)
22 (28.6)
59 (46.1)
52 (42.6)
89 (45.4)
74 (37.2)
97 (57.7)
55 (33.1)
   Lymphopenia
7 (10.3)
7 (9.1)
18 (14.1)
14 (11.5)
25 (12.8)
21 (10.6)
31 (18.5)
18 (10.8)
   Leukopenia
7 (10.3)
2 (2.6)
11 (8.6)
10 (8.2)
18 (9.2)
12 (6.0)
22 (13.1)
9 (5.4)
   Anemia
4 (5.9)
11 (14.3)
17 (13.3)
24 (19.7)
21 (10.7)
35 (17.6)
28 (16.7)
40 (24.1)
   Thrombocytopenia
4 (5.9)
3 (3.9)
8 (6.3)
12 (9.8)
12 (6.1)
15 (7.5)
17 (10.1)
18 (10.8)
Nonhematologic TEAEs
   Infections
16 (23.5)
22 (28.6)
46 (35.9)
30 (24.6)
62 (31.6)
52 (26.1)
70 (41.7)
46 (27.7)
   Pneumonia
7 (10.3)
5 (6.5)
13 (10.2)
11 (9.0)
20 (10.2)
16 (8.0)
33 (19.6)
17 (10.2)
   Cataract
10 (14.7)
8 (10.4)
11 (8.6)
9 (7.4)
21 (10.7)
17 (8.5)
13 (7.7)
19 (11.4)
   Pulmonary
   embolism

8 (11.8)
5 (6.5)
6 (4.7)
9 (7.4)
14 (7.1)
14 (7.0)
7 (4.2)
5 (3.0)
   Hypokalemia
7 (10.3)
5 (6.5)
12 (9.4)
10 (8.2)
19 (9.7)
15 (7.5)
18 (10.7)
20 (12.0)
   Hyperglycemia
2 (2.9)
2 (2.6)
13 (10.2)
4 (3.3)
15 (7.7)
6 (3.0)
12 (7.1)
8 (4.8)
Total number of patients with TEAE with an outcome of death
1 (1.5)
3 (3.9)
6 (4.7)
4 (3.3)
7 (3.6)
7 (3.5)
20 (11.9)
20 (12.0)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; Rd, lenalidomide + dexamethasone; TEAE, treatmentemergent adverse event.
  • The 2 most common reasons for treatment discontinuation across all subgroups were PD and AEs. Median time to treatment discontinuation was 13.4 vs 12.1 months in frail patients, 19.1 vs 12.0 months in total non-frail patients, 16.4 vs 10.4 months in intermediate patients, and 22.3 vs 17.3 months in fit patients receiving D-Rd vs Rd, respectively.
  • TEAEs that led to treatment discontinuation most frequently were fatigue and pulmonary embolism in the D-Rd and Rd arms, respectively. The rates of treatment discontinuation due to TEAEs were highest in the frail subgroup:
    • Frail patients: 10.1% and 19.3% receiving D-Rd and Rd, respectively
    • Total non-frail patients: 6.6% and 15.6% receiving D-Rd and Rd, respectively
    • Intermediate patients: 6.3% and 16.4% receiving D-Rd and Rd, respectively
    • Fit patients: 7.4% and 14.3% receiving D-Rd and Rd, respectively
  • The rates of death and TEAEs resulting in death were highest in the frail subgroup. Deaths were reported in:
    • Frail patients: 33.9% and 34.3% receiving D-Rd and Rd, respectively
    • Total non-frail patients: 13.3% and 23.1% receiving D-Rd and Rd, respectively
    • Intermediate patients: 16.4% and 27.9% receiving D-Rd and Rd, respectively
    • Fit patients: 7.4% and 15.6% receiving D-Rd and Rd, respectively
  • Deaths were reported due to TEAEs in frail (11.9% vs 12.0%), total non-frail (3.6% vs 3.5%), intermediate (4.7% vs 3.3%), and fit (1.5% vs 3.9%) patients receiving DRd vs Rd, respectively.

Subgroup Analysis of MAIA According to Age

Facon et al (2022)7 presented the efficacy results of the MAIA study according to age group.

Results

Patient Characteristics

Patient Disposition and Treatment Discontinuations According to Age Group in the ITT Population (MAIA)7
Parameter
Age <70 Years
Age ≥70 to
<75 Years

Age <75 Years
D-Rd (n=78)
Rd (n=77)
D-Rd (n=130)
Rd (n=131)
D-Rd (n=208)
Rd (n=208)
Patients treated, n (%)a
78 (100)
76 (98.7)
129 (99.2)
130 (99.2)
207 (99.5)
206 (99.0)
Patients who discontinued treatment, n (%)b
37 (47.4)
64 (84.2)
79 (61.2)
106 (81.5)
116 (56.0)
170 (82.5)
   Reasons for discontinuation, n (%)
      PD
17 (21.8)
34 (44.7)
42 (32.6)
47 (36.2)
59 (28.5)
81 (39.3)
      AE
9 (11.5)
12 (15.8)
20 (15.5)
33 (25.4)
29 (14.0)
45 (21.8)
      Noncompliance with study drug
5 (6.4)
5 (6.6)
7 (5.4)
7 (5.4)
12 (5.8)
12 (5.8)
      Death
5 (6.4)
2 (2.6)
5 (3.9)
7 (5.4)
10 (4.8)
9 (4.4)
      Physician’s decision
1 (1.3)
9 (11.8)
3 (2.3)
7 (5.4)
4 (1.9)
16 (7.8)
      Patient withdrawal
0
2 (2.6)
1 (0.8)
3 (2.3)
1 (0.5)
5 (2.4)
      Other
0
0
1 (0.8)
2 (1.5)
1 (0.5)
2 (1.0)
Abbreviations: AE, adverse event; D-Rd, DARZALEX + lenalidomide + dexamethasone; ITT, intent-to-treat; PD, progressive disease; Rd, lenalidomide + dexamethasone.
aPercentages are based on number of patients randomized.
bPercentages are based on number of patients treated.

Efficacy

Survival Outcomes According to Age Group in the ITT Population (MAIA)7
Parameter
Age <70 Years
Age ≥70 to
<75 Years

Age <75 Years
D-Rd
(n=78)

Rd
(n=77)

D-Rd
(n=130)

Rd
(n=131)

D-Rd
(n=208)

Rd
(n=208)

Median PFS, months
NR
39.2
61.9
37.5
NR
37.5
Median PFS HR (95% CI)
0.35 (0.21-0.56)
0.64 (0.45-0.89)
0.52 (0.39-0.68)
P value
<0.0001
0.0079
<0.0001
60-month PFS, %
67.2
28.7
51.6
36.6
57.4
33.6
OS HR (95% CI)
0.50 (0.27-0.90)
0.64 (0.43-0.96)
0.59 (0.43-0.83)
P value
0.0179
0.0274
0.0017
60-month OS, %
79.9
61.7
70.3
57.0
73.9
58.8
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; HR, hazard ratio; ITT, intent-to-treat; NR, not reached; OS, overall survival; PFS, progression-free survival; Rd, lenalidomide + dexamethasone.

Response Outcomes According to Age Group in the ITT Population (MAIA)7
Parameter, n (%)
Age <70 Years
Age ≥70 to
<75 Years

Age <75 Years
D-Rd
(n=78)

Rd
(n=77)

P
Value

D-Rd
(n=130)

Rd
(n=131)

P
Value

D-Rd
(n=208)

Rd
(n=208)

P
Value

ORR
73
(93.6)

62
(80.5)

0.0156
125
(96.2)

108
(82.4)

0.0004
198
(95.2)

170
(81.7)

<0.0001
   ≥CR
44
(56.4)

24
(31.2)

0.0016
73
(56.2)

41
(31.3)

<0.0001
117
(56.3)

65
(31.3)

<0.0001
      sCR
31
(39.7)

11
(14.3)

0.0004
50
(38.5)

23
(17.6)

0.0002
81
(38.9)

34
(16.3)

<0.0001
      CR
13
(16.7)

13
(16.9)

-
23
(17.7)

18
(13.7)

-
36
(17.3)

31
(14.9)

-
   ≥VGPR
64
(82.1)

45
(58.4)

0.0013
111
(85.4)

76
(58.0)

<0.0001
175
(84.1)

121
(58.2)

<0.0001
      VGPR
20
(25.6)

21
(27.3)

-
38
(29.2)

35
(26.7)

-
58
(27.9)

56
(26.9)

-
   PR
9
(11.5)

17
(22.1)

-
14
(10.8)

32
(24.4)

-
23
(11.1)

49
(23.6)

-
SD
1
(1.3)

14
(18.2)

-
3
(2.3)

20
(15.3)

-
4
(1.9)

34
(16.3)

-
PD
0
0
-
0
0
-
0
0
-
NE
4
(5.1)

1
(1.3)

-
2
(1.5)

3
(2.3)

-
6
(2.9)

4
(1.9)

-
MRD-negative (10–5)
28
(35.9)

9
(11.7)

0.0006
47
(36.2)

16
(12.2)

<0.0001
75
(36.1)

25
(12.0)

<0.0001
Abbreviations: CR, complete response; D-Rd, DARZALEX + lenalidomide + dexamethasone; ITT, intent-to-treat; MRD, minimal residual disease; NE, not evaluable; ORR, overall response rate; PD, progressive disease; PR, partial response; Rd, lenalidomide + dexamethasone; sCR, stringent complete response; SD, stable disease; VGPR, very good partial response.

Pooled Analysis of Transplant-Ineligible, High-Risk Patients with NDMM from MAIA and ALCYONE Studies

Jakubowiak et al (2022)8 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, ≥CR, ≥VGPR, MRD-negative CR

Results

Patient Characteristics
  • Overall, 101 patients (D-Rd [MAIA], n=48; DARZALEX + bortezomib + melphalan + prednisone [D-VMP; ALCYONE], n=53) were included in the pooled DARZALEX cohort and 89 patients (Rd [MAIA], n=44; bortezomib + melphalan + prednisone [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 12.0 months, respectively.
  • Baseline characteristics of patients in the MAIA study are summarized in Table: Summary of Baseline Characteristics in MAIA.

Summary of Baseline Characteristics in MAIA8
Characteristic, n (%)
D-Rd (n=48)
Rd (n=44)
Standardized Differencea
Median age, years
74.5
74.0
-
   <75 years
24 (50.0)
24 (54.5)
9.1%
   ≥75 years
24 (50.0)
20 (45.5)
9.1%
Male
24 (50.0)
20 (45.5)
9.1%
ECOG PS score
   0
17 (35.4)
18 (40.9)
11.3%
   1
18 (37.5)
17 (38.6)
2.3%
   ≥2
13 (27.1)
9 (20.5)
15.6%
Type of MM by immunofixation or serum FLC
   IgG
35 (72.9)
28 (63.6)
20.0%
   Non-IgG
13 (27.1)
16 (36.4)
20.0%
ISS stageb
   I
6 (12.5)
8 (18.2)
15.8%
   II
21 (43.8)
15 (34.1)
19.9%
   III
21 (43.8)
21 (47.7)
8.0%
Cytogenetic riskc
   del(17p)
25 (52.1)
29 (65.9)
28.4%
   t(4;14)
21 (43.8)
12 (27.3)
35.0%
   t(14;16)
4 (8.3)
5 (11.4)
10.2%
Renal impairmentd
25 (52.1)
19 (43.2)
17.9%
Abbreviations: del, deletion; D-Rd, DARZALEX + lenalidomide + dexamethasone; 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; t, translocation.
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.
dRenal impairment was defined as having baseline creatinine clearance <60 mL/minute.

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, 0.33-1.00) in the MAIA study. See Table: HRs for PFS in Individual Studies.
  • 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 Individual Studies8
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 (0.33-1.00)
Pooleda
64/101
64/89
0.59 (0.41-0.85)
Abbreviations: CI, confidence interval; del, deletion; ECOG PS, Eastern Cooperative Oncology Group performance status; HR, hazard ratio; IgG, immunoglobulin G; ISS, International Staging System; PFS, progression-free survival; t, translocation.
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 PS, 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.
  • 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 Rates8
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)

5.50
(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 (3.0%)
19 (21.3%)
-
-
-
-
-
   PD
0 (0.0%)
0 (0.0%)
-
-
-
-
-
   NE
5 (5.0%)
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: CI, confidence interval; CR, complete response; del, deletion; ECOG PS, Eastern Cooperative Oncology Group performance status; 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; t, translocation; 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 PS, 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).

Impact of Frailty on PROs in the MAIA Study

Perrot et al (2022)9 presented a post hoc analysis of PROs in a subgroup of frail patients from the MAIA study.

Study Design/Methods

  • Patients were categorized as frail based on age, ECOG PS, and Charlson comorbidity index.
  • PROs were assessed using the following sub-scales of the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30-item (EORTC QLQ-C30) questionnaire:
    • GHS
    • Functional scales: physical, role, emotional, cognitive, and social
    • Symptom scales: fatigue, nausea and vomiting, and pain
  • The scores were transformed to values ranging from 0-100.
  • The questionnaires were completed at baseline; on day 1 of cycles 3, 6, 9, and 12 for year 1; and every 6 months thereafter until PD.

Results

Patient Characteristics

Baseline Patient and Disease Characteristics in Frail Patients (MAIA)9
Characteristic
D-Rd (n=172)
Rd (n=169)
Median age (range), years
77.0 (57-90)
77.0 (45-89)
Female, n (%)
79 (45.9)
79 (46.7)
ECOG PS score, n (%)
   0
20 (11.6)
18 (10.7)
   1
89 (51.7)
92 (54.4)
   ≥2
63 (36.6)
59 (34.9)
ISS stage, n (%)a
   I
34 (19.8)
35 (20.7)
   II
74 (43.0)
67 (39.6)
   III
64 (37.2)
67 (39.6)
Type of measurable disease, n (%)
   IgG
103 (59.9)
99 (58.6)
   IgA
32 (18.6)
34 (20.1)
Cytogenetic profile, n/N (%)b
High risk
25/153 (16.3)
23/147 (15.6)
Abbreviations: del, deletion; D-Rd, DARZALEX + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; IgA, immunoglobulin A; IgG, immunoglobulin G; ISS, International Staging System; Rd, lenalidomide + dexamethasone; t, translocation.
aBased on the combination of serum β2-microglobulin and albumin.
bPatients with high-risk cytogenetics had a del17p, t(14;16), or t(4;14) abnormality.


Baseline EORTC QLQ-30 Scores in Frail Patients (MAIA)9
EORTC QLQ-C30 Score, Mean (SD)
D-Rd (n=172)
Rd (n=169)
GHS
50.4 (26.1)
51.6 (24.3)
Functional scales
   Physical functioning
55.2 (29.3)
57.9 (26.1)
   Role functioning
49.5 (37.7)
54.6 (34.0)
   Emotional functioning
68.7 (26.2)
66.3 (25.6)
   Cognitive functioning
75.1 (26.9)
74.2 (24.9)
   Social functioning
62.1 (36.3)
66.8 (31.5)
Symptom scales
   Pain
54.5 (37.8)
51.8 (34.0)
   Fatigue
49.2 (31.4)
50.2 (27.3)
   Nausea/vomiting
9.0 (19.0)
9.7 (18.9)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; EORTC QLQ-C30, European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30-item; GHS, global health status; Rd, lenalidomide + dexamethasone; SD, standard deviation.
PROs
  • Larger reductions (≥20-point change) in LS mean pain score and a greater improvement in pain symptoms from baseline were reported in patients treated with D-Rd vs Rd.
  • Treatment with D-Rd did not increase fatigue compared with Rd.
  • Improvements in GHS scores from baseline were consistent over time both in patients treated with D-Rd and Rd.
  • Improvements in physical functioning from baseline was greater in the D-Rd vs Rd arm until cycle 42.
  • Improvements in emotional and social functioning from baseline was numerically higher in the D-Rd vs Rd arm at several timepoints.
  • Role functioning was improved from baseline in both treatment arms with no difference between D-Rd and Rd.
  • No meaningful changes from baseline were reported in nausea and vomiting in either treatment arm.
  • Overall, the median time to worsening of HRQoL scores were longer in the D-Rd vs Rd arm, particularly for physical functioning (D-Rd, 68.1 months; Rd, 39.6 months).
    • At data cutoff, the median time to worsening of pain symptoms was NR in the D-Rd arm.
    • Fatigue scores were maintained for a longer median duration in the Rd (35 months) vs D-Rd (22 months) arm.
    • In the D-Rd vs Rd arm, cognitive symptoms worsened at 8 vs 10 months.

Subgroup Analysis in Patients with Renal Impairment and/or High Cytogenetic Risk in the MAIA Study

Facon et al (2022)10 conducted a subgroup analysis of the phase 3 MAIA study that evaluated the efficacy and PROs of D-Rd vs Rd in subgroups of patients with NDMM who had renal impairment (CrCl ≤60 mL/min) and/or high-risk cytogenetics (del17p, t[4;14], or t[14;16] abnormality).

Study Design/Methods

  • Renal impairment was defined as CrCl ≤60 mL/min
  • Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality.

Results

Patient Characteristics

Baseline Renal Function and Cytogenetic Risk Status10
Characteristic
D-Rd (n=368)
Rd (n=369)
Baseline renal function (CrCl), n (%)
   >60 mL/min
206 (56.0)
227 (61.5)
   ≤60 mL/min
162 (44.0)
142 (38.5)
Cytogenetic risk,a n
319
323
   Standard risk, n (%)
271 (85.0)
279 (86.4)
   High risk, n (%)
48 (15.0)
44 (13.6)
Abbreviations: CrCl, creatinine clearance; del, deletion; D-Rd, DARZALEX + lenalidomide + dexamethasone; IMWG, International Myeloma Working Group; Rd, lenalidomide + dexamethasone; t, translocation.
aCytogenetic risk was evaluated based on local fluorescence in situ hybridization or karyotype testing. Patients with high cytogenetic risk had a del17p, t(4;14), or t(14;16) abnormality per IMWG 2016 recommendations. Patients with standard cytogenetic risk had an absence of high-risk cytogenetic abnormalities.

Efficacy

Efficacy Outcomes in the Overall Study Population and in Renal Function and Cytogenetic Risk Subgroups10
Parameter
D-Rd
Rd
HR (95% CI)
P Value
Median time to ≥VGPR, monthsa,b
   ITT population
3.8
9.4
2.08 (1.73-2.49)
<0.0001
   Renal function
      CrCl ≤60 mL/min
3.8
12.5
2.26 (1.69-3.02)
<0.0001
      CrCl >60 mL/min
3.8
8.5
1.82 (1.45-2.28)
<0.0001
   Cytogenetic risk status
      High risk
4.7
14.1
2.50 (1.44-4.36)
0.0008
      Standard risk
3.8
9.3
1.96 (1.59-2.40)
<0.0001
Median time to ≥CR, months
   Renal function
      CrCl ≤60 mL/min
23.3
54.6
1.58 (1.07-2.33)
0.0197
      CrCl >60 mL/min
17.6
43.8
1.80 (1.34-2.41)
<0.0001
   Cytogenetic risk status
      High risk
15.7
47.9
1.74 (0.83-3.63)
0.1372
      Standard risk
20.8
42.6
1.69 (1.29-2.21)
<0.0001
48-month EFS rate in patients achieving ≥CR, %
   ITT population c,d
81.8
57.8
0.38 (0.23-0.65)
0.0002
   Renal function
      CrCl ≤60 mL/min
81.0
61.5
0.45 (0.20-1.04)
0.0551
      CrCl >60 mL/min
82.2
55.3
0.41 (0.22-0.77)
0.0043
   Cytogenetic risk status
      High risk
74.7
NE
0.32 (0.09-1.16)
0.0694
      Standard risk
80.0
55.1
0.42 (0.24-0.73)
0.0015
48-month EFS rate in patients achieving ≥PR, %
   Renal function
      CrCl ≤60 mL/min
67.2
44.4
0.50 (0.34-0.74)
0.0003
      CrCl >60 mL/min
69.8
48.9
0.50 (0.36-0.70)
<0.0001
      Cytogenetic risk status
      High risk
48.8
29.9
0.65 (0.35-1.19)
0.1560
      Standard risk
72.3
45.7
0.43 (0.32-0.57)
<0.0001
Abbreviations: CI, confidence interval; CR, complete response; CrCl, creatinine clearance; D-Rd, DARZALEX + lenalidomide + dexamethasone; EFS, event-free survival; HR, hazard ratio; ISS, International Staging System; ITT, intent-to-treat; NE, not evaluable; PR, partial response; Rd, lenalidomide + dexamethasone; VGPR, very good partial response.
aHR and 95% CI are calculated from a Cox proportional hazards model, with treatment as the sole explanatory variable. HR >1 indicates an advantage for D-Rd.
bP value was based on the log-rank test.
cHR and 95% CI are calculated from a Cox proportional hazards model, with treatment as the sole explanatory variable and stratified by ISS stage (I, II, or III), region (North America vs other), and age (<75 years vs ≥75 years), as randomized. HR <1 indicates an advantage for D-Rd.
dP value was based on a stratified log-rank test.

  • Among patients with renal impairment, greater improvements from baseline in patientreported pain scores, patient-reported fatigue, and nausea and vomiting symptom scores were observed in the D-Rd vs Rd arm across most time points.
    • A greater meaningful reduction in pain score was reported in the D-Rd vs Rd arm, respectively, at cycle 6 day 1 (LS mean change from baseline, 14.9 vs 7.0; P=0.0241) in patients with renal impairment.
    • A greater reduction in fatigue symptom score was reported in the D-Rd vs Rd arm, respectively, at cycle 24 day 1 (LS mean change from baseline, -2.9 vs 8.1; P=0.0018), cycle 30 day 1 (LS mean change from baseline, -3.7 vs 4.4; P=0.0258), and cycle 36 day 1 (LS mean change from baseline, -2.6 vs 6.5; P=0.0183) in patients with renal impairment.
    • A greater reduction in nausea and vomiting symptom score was observed in the D-Rd vs Rd arm, respectively, at cycle 36 day 1 (LS mean change from baseline, 3.7 vs 3.3; P=0.0035) in patients with renal impairment.
  • Greater improvements from baseline in patient-reported symptoms were observed in the D-Rd vs Rd arm in patients without renal impairment (CrCl >60 mL/min).
  • Greater improvements from baseline in patient-reported pain score were observed in the D-Rd vs Rd arm across most time points in patients with standard and high cytogenetic risk.

Predictive and Prognostic Role of MRD-Negativity and Durability

San-Miguel et al (2022)11 evaluated the predictive and prognostic role of MRD-negativity and durability in patients with NDMM ineligible for transplant.

Results

Patient Characteristics

Demographics and Baseline Characteristics by MRD Durability23
Characteristic
D-Rd
Rd
ITT
(n=368)

MRD-Negative Patients
ITT
(n=369)

MRD-Negative Patients
At Any Time
(n=106)

≥12 Months
(n=40)

Not ≥12 Months
(n=66)

At Any Time
(n=34)

≥12 Months
(n=9)

Not ≥12 Months
(n=25)

Age, years
   Median (range)
73.0 (50-90)
72.0 (65-87)
71.0 (66-85)
73.5 (65-87)
74.0 (45-89)
72.5 (66-87)
71.0 (69-78)
73.0 (66-87)
   Distribution
   <75
208 (56.5)
68 (64.2)
31 (77.5)
37 (56.1)
208 (56.4)
20 (58.8)
6 (66.7)
14 (56.0)
   ≥75
160 (43.5)
38 (35.8)
9 (22.5)
29 (43.9)
161 (43.6)
14 (41.2)
3 (33.3)
11 (44.0)
ISS disease stagea
   I
98 (26.6)
24 (22.6)
10 (25.0)
14 (21.2)
103 (27.9)
11 (32.4)
5 (55.6)
6 (24.0)
   II
163 (44.3)
55 (51.9)
19 (47.5)
36 (54.5)
156 (42.3)
15 (44.1)
3 (33.3)
12 (48.0)
   III
107 (29.1)
27 (25.5)
11 (27.5)
16 (24.2)
110 (29.8)
8 (23.5)
1 (11.1)
7 (28.0)
Cytogenetic profileb
   Patients evaluated
319
96
34
62
323
27
8
19
   Standard-risk cytogenetic abnormality
271 (85.0)
85 (88.5)
29 (85.3)
56 (90.3)
279 (86.4)
26 (96.3)
8 (100.0)
18 (94.7)
   High-risk cytogenetic abnormalityc
48 (15.0)
11 (11.5)
5 (14.7)
6 (9.7)
44 (13.6)
1 (3.7)
0
1 (5.3)
      del(17p)
25 (7.8)
6 (6.3)
2 (5.9)
4 (6.5)
29 (9.0)
0
0
0
Abbreviations: del, deletion; D-Rd, DARZALEX + lenalidomide + dexamethasone; ISS, International Staging System; ITT, intention-to-treat; MRD, minimal residual disease; Rd, lenalidomide + dexamethasone; t, translocation.
All data are n (%) unless otherwise indicated.
aISS staging is derived based on the combination of serum β2-microglobulin and albumin.
bCytogenetic risk status was determined by fluorescence in situ hybridization or karyotype testing.
cHigh risk is defined as having a positive test for any of the del17p, t(14;16), or t(4;14) molecular abnormalities.

Efficacy
  • In the ITT population and among patients with ≥CR, patients receiving the D-Rd regimen achieved higher rates of MRD-negativity and durability compared with Rd. See Table: Rates of Sustained MRD-negativity Status in Transplant-ineligible NDMM.
  • In the ITT population, MRD-negative patients had improved PFS compared with MRD-positive patients (HR, 0.15; 95% CI, 0.09-0.26; P<0.0001).
  • Durable MRD-negativity lasting ≥6 months or ≥12 months improved PFS compared with MRD-negative patients who did not maintain MRD-negativity for ≥6 months or
    ≥12 months.
  • Estimated 36-month rates of time to subsequent anticancer therapy were mostly higher for patients who achieved MRD-negativity at any time vs MRD-positive patients, and for patients with sustained MRD-negativity lasting ≥6 months or ≥12 months compared with those who did not have durable MRD-negativity are presented in the Table: Estimated Rates of Patients Without Subsequent Therapy at 36 Months in ITT Population.
  • Estimated 36-month PFS2 rate (ITT; D-Rd, n=368; Rd, n=369):
    • MRD-negative (10-5) at ≥1 time point: D-Rd, 95.0% (n=106); Rd, 83.9% (n=34)
    • MRD-positive: D-Rd, 65.5% (n=262); Rd, 61.5% (n=335)

Rates of Sustained MRD-negativity Status in Transplant-ineligible NDMM11
MRD-Negativity (10-5)
(N=737)
D-Rd
Rd
P Valuea
ITT
n=368
n=369
   MRD-negative status, n (%)
106 (28.8)
34 (9.2)
<0.0001
      ≥6 months sustained
55 (14.9)
16 (4.3)
<0.0001
      ≥12 months sustained
40 (10.9)
9 (2.4)
<0.0001
≥CR
n=182
n=100
   MRD-negative status, n (%)
106 (58.2)
34 (34.0)
0.0001
      ≥6 months sustained
55 (30.2)
16 (16.0)
0.0097
      ≥12 months sustained
40 (22.0)
9 (9.0)
0.0053
Abbreviations: CR, complete response; D-Rd, DARZALEX + lenalidomide + dexamethasone; ITT, intention-to-treat; MRD, minimal residual disease; NDMM, newly diagnosed multiple myeloma; Rd, lenalidomide + dexamethasone.
aP value was calculated using Fisher’s exact test.


Estimated Rates of Patients Without Subsequent Therapy at 36 months in ITT Population11
Estimated 36-Month Time to Subsequent Anticancer Therapy Rate, n (%)
D-Rd (n=368)
Rd (n=369)
MRD-negative (10-5) at ≥1 time point
106 (96.9)
34 (90.5)
MRD-positive
262 (65.4)
335 (48.7)
Achieved and remained MRD-negative (10-5) for ≥6 months
55 (96.1)
16 (100.0)
MRD-negativity not ≥6 months
51 (98.0)
18 (78.7)
Achieved and remained MRD-negative (10-5) for ≥12 months
40 (94.6)
9 (100.0)
MRD-negativity not ≥12 months
66 (98.5)
25 (85.2)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; ITT, intention-to-treat; MRD, minimal residual disease; Rd, lenalidomide + dexamethasone.

Post Hoc Analysis of MAIA Study

Moreau et al (2022)12 analyzed PFS and OS by treatment duration within the MAIA study, with a median follow-up of 56.2 months.

Study Design/Methods

  • OS was evaluated in patients who received treatment with D-Rd for <18 months and ≥18 months.
  • PFS and OS was evaluated in the following subgroups:
    • Patients who received treatment with D-Rd and who discontinued either DARZALEX only or lenalidomide only with or without dexamethasone but continued remaining treatment.
    • Patients who received treatment with D-Rd or Rd for ≥9 months or for ≥18 months.
    • Patients who achieved VGPR by 6 months and ≥CR by 9 months or 18 months.
  • Patients were excluded from this analysis if the treatment with D-Rd was discontinued due to disease progression during the first 18 months.

Results


Baseline Characteristics and Demographics12
Characteristic, n (%)
ITT Population (D-Rd, n=368)
Discontinued only R±da
(D-Rd, n=48)

D-Rd treatment
<18 monthsb
(n=48)

D-Rd treatment ≥18 monthsb (n=283)
Age
   <65 years
4 (1.1)
0
3 (6.3)
1 (0.4)
   65-74 years
204 (55.4)
26 (54.2)
18 (37.5)
167 (59.0)
   ≥75 years
160 (43.5)
22 (45.8)
27 (56.3)
115 (40.6)
   Median (range)
73.0 (50-90)
74.0 (67-87)
75.0 (50-90)
73.0 (55-88)
Baseline ECOG PS score
   0
127 (34.5)
15 (31.3)
10 (20.8)
110 (38.9)
   1
178 (48.4)
25 (52.1)
25 (52.1)
132 (46.6)
   ≥2
63 (17.1)
8 (16.7)
13 (27.1)
41 (14.5)
  ISS disease stagec
   I
98 (26.6)
14 (29.2)
8 (16.7)
84 (29.7)
   II
163 (44.3)
26 (54.2)
19 (39.6)
129 (45.6)
   III
107 (29.1)
8 (16.7)
21 (43.8)
70 (24.7)
Cytogenetic abnormalitiesd
   N
319
45
44
242
      Standard risk
271 (85.0)
39 (86.7)
38 (86.4)
212 (87.6)
      High risk
48 (15.0)
6 (13.3)
6 (13.6)
30 (12.4)
Frailty status
   Frail
172 (46.7)
23 (47.9)
30 (62.5)
118 (41.7)
   Intermediate
128 (34.8)
19 (39.6)
13 (27.1)
105 (37.1)
   Fit
68 (18.5)
6 (12.5)
5 (10.4)
60 (21.2)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; ITT, intention-to-treat; R±d, lenalidomide ± dexamethasone.
aPatients discontinued R±d but continued remaining treatment.
bPatients who discontinued D-Rd due to disease progression during the first 18 months of treatment were excluded.
cISS staging is derived based on the combination of serum β2-microglobulin and albumin.
dCytogenetic abnormalities (del17p, t[14;16] or t[4;14]) were based on fluorescence in situ hybridization or karyotype testing. Percentages calculated with the number of patients in each treatment group as the denominator).

Patient Disposition

Reasons for Lenalidomide Discontinuation12
Event, n (%)
Discontinued only R±d
(D-Rd, n=48)a
Reasons for lenalidomide discontinuation
   Adverse Event
44 (91.7)
   Otherb
4 (8.3)
Most common (≥5%) reasons for discontinuation of lenalidomide due to adverse events
   Diarrhea
9 (18.8)
   Peripheral sensory neuropathy
5 (10.4)
   Neutropenia
4 (8.3)
   Constipation
6 (6.3)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; R±d, lenalidomide ± dexamethasone.
aPatients discontinued R±d but continued remaining treatment.
bOther includes patient decision to discontinue, interruption due to adverse events caused by progressive disease or other conditions, or cumulative low-grade adverse events.

Efficacy
  • Median duration of DARZALEX treatment in the D-Rd arm was 58.1 months in patients who discontinued R±d but continued remaining treatment.
    • The estimated 60-month PFS rate and OS rate was 97.9% and 100.0%, respectively.
  • Only 1 patient discontinued DARZALEX while continuing lenalidomide treatment.
    • This patient was alive and progression free at the time of analysis. No further analysis was performed on this patient.
  • The median OS in the D-Rd arm was NR vs 20.5 months (HR, 0.16; 95% CI, 0.1-0.25; P<0.0001) in patients who received treatment for ≥18 months vs <18 months, respectively.
  • In patients who received D-Rd vs Rd for ≥9 months, PFS (HR, 0.49; 95% CI, 0.38-0.62; P<0.0001) and OS (HR, 0.63; 95% CI, 0.47-0.85; P=0.0025) benefits were observed.
  • PFS benefit in the D-Rd vs Rd arm was observed in patients who received treatment for ≥18 months (HR, 0.57; 95% CI, 0.43-0.76; P<0.0001) vs ITT population (HR, 0.53; 95% CI, 0.43-0.66; P<0.0001).
  • OS benefit in the D-Rd vs Rd arm was observed in patients who received treatment for ≥18 months (HR, 0.68; 95% CI, 0.47-0.98; P=0.0379) vs ITT population (HR, 0.68; 95% CI, 0.53-0.86; P=0.0013).
  • PFS and OS benefits in the D-Rd vs Rd arm was observed in patients who achieved a best response of VGPR by 6 months and converted to ≥CR by 9 or 18 months as presented in Table: PFS and OS in Patients who Achieved VGPR by 6 Months and Converted to ≥CR by 9 or 18 Months.
  • Among patients who were treated for ≥18 months, the ≥CR rate was 9.2% vs 4.4% at 6 months, 19.1% vs 11.3% at 9 months, and 49.8% vs 30.4% at 18 months in the D-Rd vs Rd arm, respectively. See Table: Response Rates in Patients Treated for ≥18 Months.

PFS and OS in Patients who Achieved VGPR by 6 Months and Converted to ≥CR by 9 or 18 Months12
Parameter
D-Rd
Rd
Hazard Ratio (95% CI)
P value
Patients who achieved VGPR by 6 Months and converted to ≥CR by 9 months
   Median PFS, months
NR
38.4
0.15 (0.05-0.45)
<0.0001
   Median OS, months
NR
53.8
0.25 (0.07-0.86)
0.0175
Patients who achieved VGPR by 6 Months and converted to ≥CR by 18 months
   Median PFS, months
NR
53.6
0.34 (0.19-0.62)
0.0002
   Median OS, months
NR
NR
0.33 (0.17-0.65)
0.0006
Abbreviations: CI, confidence interval; ≥CR, complete response or better; D-Rd, DARZALEX + lenalidomide + dexamethasone; NR, not reached; OS, overall survival; PFS, progression-free survival; Rd, lenalidomide + dexamethasone; VGPR, very good partial response.

Response Rates in Patients Treated for ≥18 Months12
Response Rates, %
6 months
9 months
18 months
D-Rd
Rd
D-Rd
Rd
D-Rd
Rd
ORR
98.2
93.6
98.6
96.1
99.6
98.0
   P value
0.0079
0.0785
0.0828
≥CR
9.2
4.4
19.1
11.3
49.8
30.4
   P value
0.0443
0.0199
<0.0001
   sCR
2.5
2.9
7.1
5.4
27.2
13.7
   CR
6.7
1.5
12.0
5.9
22.6
16.7
VGPR
57.2
38.2
57.2
41.7
36.7
37.3
PR
31.8
51.0
22.3
43.1
13.1
30.4
Abbreviations: CR, complete response; D-Rd, DARZALEX + lenalidomide + dexamethasone; ORR, overall response rate; PR, partial response; Rd, lenalidomide + dexamethasone; sCR, stringent complete response; VGPR, very good partial response.
Safety

Most Common (≥10% in Either Treatment Arm) Grade 3/4 TEAEs in Patients Who Received ≥18 Months of Treatment12
Event, n (%)
D-Rd (n=283)
Rd (n=204)
≥1 grade 3/4 TEAE
273 (96.5)
186 (91.2)
Hematologic
   Neutropenia
161 (56.9)
84 (41.2)
   Anemia
48 (17.0)
37 (18.1)
   Lymphopenia
48 (17.0)
25 (12.3)
   Leukopenia
33 (11.7)
19 (9.3)
Non-Hematologic
   Pneumonia
56 (19.8)
22 (10.8)
   Hypokalemia
41 (14.5)
24 (11.8)
   Cataract
39 (13.8)
38 (18.6)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; Rd, lenalidomide + dexamethasone; TEAE, treatment-emergent adverse event.

HRQoL Outcomes from the MAIA study

Perrot et al (2021)24 evaluated the impact of treatment on PRO in the MAIA study at a median follow-up of 28.0 months. Perrot et al (2021)13 reported updated results of the HRQoL analysis of the MAIA study, with a follow-up of 56.2 months. Results from this updated analysis are summarized below.

Results

  • At a median follow-up of 56.2 months, patients treated with D-Rd experienced lower discontinuation rates vs those treated with Rd (56.8% vs 80.8%, respectively).
  • The threshold for clinically meaningful change from baseline was defined a priori for each PRO.
    • A greater proportion of D-Rd-treated patients achieved a meaningful improvement vs Rd-treated patients, particularly for physical functioning (OR, 1.49; 95% CI, 1.111.99), fatigue (OR, 1.54; 95% CI, 1.14-2.07), dyspnea (OR, 1.43; 95% CI, 1.06-1.93), and constipation (OR, 1.66; 95% CI, 1.23-2.24).
    • The number of patients achieving meaningful worsening was similar across arms.
  • Median time to meaningful improvement was shorter with D-Rd vs Rd for both physical functioning (HR, 1.13; 95% CI, 0.91-1.40) and pain (HR, 1.01; 95% CI, 0.84-1.21) and with Rd vs D-Rd for both GHS (HR, 0.94; 95% CI, 0.77-1.15) and fatigue (HR, 1.01; 95% CI, 0.84-1.21).
  • Median time to worsening was comparable between the D-Rd and Rd arms for fatigue, longer for D-Rd vs Rd for GHS, and significantly longer for D-Rd vs Rd for physical functioning (HR, 0.77; 95% CI, 0.62-0.96), pain (HR, 0.69; 95% CI, 0.55-0.86), and dyspnea (HR, 0.78; 95% CI, 0.63-0.96).
  • Differences between arms in LS mean change from baseline for select PROs favored D-Rd at most time points. For most scales, differences were significant at ≥1 time point.

PROs in the MAIA Study

Perrot et al (2021)14 reported results of post hoc analyses assessing the relationship between clinical efficacy endpoints and PROs in the MAIA study.

Results


Best Clinical Response in the MAIA Study (Pooled Treatment Arms)14
Best Clinical Response
n (%)
Stringent complete response
186 (26.2)
Complete response
113 (15.9)
Very good partial response
209 (29.4)
Partial response
135 (19.0)
Stable disease
66 (9.3)
Progressive disease
1 (0.1)
N=710; best clinical response data were missing for 27/37 patients in the intent-to-treat population.

Odds Ratio (95% CI) for Meaningful Improvement of GHS, Fatigue, and Pain at Any Time for Each Best Clinical Response Subgroup (vs Stable Disease)14
Odds Ratio (95% CI)
GHS
   Partial response
2.6 (1.4-5.0)
   Very good partial response
3.9 (2.1-7.2)
   Complete response
4.7 (2.4-9.2)
   Stringent complete response
5.9 (3.1-11.1)
Fatigue
   Partial response
3.0 (1.6-5.6)
   Very good partial response
4.8 (2.6-8.8)
   Complete response
5.3 (2.7-10.3)
   Stringent complete response
8.9 (4.7-16.8)
Pain
   Partial response
2.2 (1.2-4.1)
   Very good partial response
3.3 (1.8-5.8)
   Complete response
4.3 (2.3-8.3)
   Stringent complete response
5.0 (2.7-9.1)
Abbreviations: CI, confidence interval; GHS, global health status.

Meaningful Worsening or No Improvement of GHS, Fatigue, and Pain at or Post-Progression Among Patients Who Progressed14
Worsening of Select PROs, %
Patients Who Experienced Disease Progression (N=264)
GHS
   Meaningful worsening
38.6
   Meaningful worsening or no improvement
62.9
Fatigue
   Meaningful worsening
54.9
   Meaningful worsening or no improvement
69.7
Pain
   Meaningful worsening
39.4
   Meaningful worsening or no improvement
61.0
Abbreviations: GHS, global health status; PRO, patient-reported outcome.

Impact of Impaired Renal Function and Lenalidomide Starting Dose on Efficacy in the MAIA Study

Usmani et al (2021)15 evaluated the efficacy of D-Rd vs Rd in patients from the MAIA study that had impaired renal function, both overall and based on the starting dose of lenalidomide that was received.

Study Design/Methods

  • Lenalidomide 10 mg was recommended for patients with a CrCl of 30 to 50 mL/min.
  • Renal impairment was defined as having a baseline CrCl of ≤60 mL/min.

Results

Patient Characteristics
  • A total of 737 patients received either D-Rd (n=368) or Rd (n=369). Of these, 162 (44.0%) patients in the D-Rd arm and 142 (38.5%) patients in the Rd arm had baseline renal impairment.
  • Among patients with renal impairment, 60 (37.0%) in the D-Rd arm and 62 (43.7%) in the Rd arm received lenalidomide at a starting dose of 25 mg, and 98 (60.5%) in the DRd arm and 75 (52.8%) in the Rd arm received lenalidomide at a starting dose of <25 mg. See Table: Distribution of Patients with Renal Impairment According to Baseline CrCl and Lenalidomide Starting Dose.
    • All patients received a starting lenalidomide dose of 10 mg, except for 1 DRdtreated patient who received a lenalidomide starting dose of 5 mg.

Distribution of Patients with Renal Impairment According to Baseline CrCl and Lenalidomide Starting Dose15
Baseline CrCl, n (%)
D-Rd
Rd
Total
(n=162)a
R Starting Dose of 25 mg
(n=60)
R Starting Dose of <25 mg
(n=98)
Total
(n=142)a
R Starting Dose of 25 mg
(n=62)
R Starting Dose of <25 mg
(n=75)
>50 to ≤60 mL/min
64 (39.5)a
41 (68.3)
22 (22.4)
62 (43.7)a
41 (66.1)
20 (26.7)
30 to ≤50 mL/min
91 (56.2)a
18 (30.0)
70 (71.4)
76 (53.5)a
20 (32.3)
52 (69.3)
<30 mL/min
7 (4.3)
1 (1.7)
6 (6.1)
4 (2.8)
1 (1.6)
3 (4.0)
Abbreviations: CrCl, creatinine clearance; D-Rd, DARZALEX + lenalidomide + dexamethasone; R, lenalidomide; Rd, lenalidomide + dexamethasone.
aFour patients in the D-Rd arm and 5 patients in the Rd arm were randomized and had baseline CrCl results but did not receive ≥1 dose of lenalidomide.


Demographic and Baseline Disease Characteristics of Patients with Renal Impairment According to Baseline CrCl and Lenalidomide Starting Dose15
Characteristics
Baseline CrCl >50 to ≤60 mL/min
Baseline CrCl ≤50 mL/min
R Starting Dose of 25 mg
(n=82)
R Starting Dose of <25 mg
(n=42)
R Starting Dose of 25 mg
(n=40)
R Starting Dose of <25 mg
(n=131)
Age, years
   Median (range)
75.0 (65.0-85.0)
76.0 (57.0-85.0)
75.5 (67.0-88.0)
78.0 (55.0-90.0)
ISS disease stage, n (%)a
   I
19 (23.2)
4 (9.5)
7 (17.5)
9 (6.9)
   II
34 (41.5)
21 (50.0)
18 (45.0)
50 (38.2)
   III
29 (35.4)
17 (40.5)
15 (37.5)
72 (55.0)
Median (range) time from MM diagnosis to randomization, months
0.92 (0.1-8.1)
1.00 (0.3-6.1)
0.97 (0.2-7.1)
0.89 (0.0-8.7)
Frailty status, n (%)b
   Fit
14 (17.1)
6 (14.3)
6 (15.0)
10 (7.6)
   Intermediate
33 (40.2)
8 (19.0)
11 (27.5)
27 (20.6)
   Frail
35 (42.7)
28 (66.7)
23 (57.5)
94 (71.8)
Cytogenetic profilec
n=71
n=37
n=35
n=119
   Standard risk, n (%)
63 (88.7)
26 (70.3)
28 (80.0)
103 (86.6)
   High risk, n (%)
8 (11.3)
11 (29.7)
7 (20.0)
16 (13.4)
      del17p
6 (8.5)
3 (8.1)
6 (17.1)
11 (9.2)
      t(4;14)
2 (2.8)
7 (18.9)
1 (2.9)
4 (3.4)
      t(14;16)
0
1 (2.7)
0
1 (0.8)
Abbreviations: CrCl, creatinine clearance; del, deletion; ECOG PS, Eastern Cooperative Oncology Group performance status; ISS, International Staging System; MM, multiple myeloma; R, lenalidomide; t, translocation.
aBased on the combination of serum β2-microglobulin and albumin.
bFrailty status was determined based on scores derived from age, baseline ECOG PS score, and Charlson comorbidity index according to the frailty scale developed in a retrospective subgroup analysis of the FIRST trial.
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.

Efficacy
  • At a median follow-up of 56.2 months, PFS benefit (median, 56.7 months vs
    29.7 months; HR, 0.55; 95% CI, 0.40-0.74) and OS benefit (median, NR vs
    54.8 months; HR, 0.67; 95% CI, 0.47-0.96) was observed in the D-Rd arm vs Rd arm, respectively.
  • PFS and OS benefits were observed with D-Rd vs Rd in renally-impaired patients who received a lenalidomide starting dose of 25 mg. PFS and OS were also prolonged with DRd vs Rd in those with renal impairment who received a lenalidomide starting dose of <25 mg. See Table: PFS and OS in Patients with Renal Impairment Based on Lenalidomide Starting Dose.

PFS and OS in Patients with Renal Impairment Based on Lenalidomide Starting Dose15

D-Rd
Rd
HR (95% CI)
P Value
R starting dose of 25 mg
n=60
n=62
-
-
   PFS
      Median PFS, months
NR
35.4
0.42 (0.24-0.72)
0.0012
      5-year PFS rate, %
63.1
33.3
-
-
   OS
      Median OS, months
NR
NR
0.37 (0.19-0.73)
0.0028
      5-year OS rate, %
79.0
50.9
-
-
R starting dose of
<25 mg

n=98
n=75
-
-
   PFS
      Median PFS, months
49.1
24.9
0.56 (0.38-0.83)
0.0029
      5-year PFS rate, %
40.3
18.6
-
-
   OS
      Median OS, months
62.8
54.8
0.81 (0.52-1.26)
0.3468
      5-year OS rate, %
54.0
44.1
-
-
Abbreviations: CI, confidence interval; D-Rd, DARZALEX + lenalidomide + dexamethasone; HR, hazard ratio; NR, not reached; OS, overall survival; PFS, progression-free survival; R, lenalidomide; Rd, lenalidomide + dexamethasone.
Safety

Deaths Among Patients with Renal Impairment15
D-Rd
Rd
Totala
Deaths among patients with renal
impairment, n
58
67
125
R starting dose of 25 mg
n=60
n=62
n=122
   Total deaths, n (%)
12 (20.0)
29 (46.8)
41 (33.6)
      Deaths due to disease progression, n (%)
6 (10.0)
10 (16.1)
16 (13.1)
R starting dose of <25 mg
n=98
n=75
n=173
   Total deaths, n (%)
44 (44.9)
37 (49.3)
81 (46.8)
      Deaths due to disease progression, n (%)
16 (16.3)
11 (14.7)
27 (15.6)
Abbreviations: D-Rd, DARZALEX + lenalidomide + dexamethasone; R, lenalidomide; Rd, lenalidomide + dexamethasone.
aTwo patients in the D-Rd arm and 1 patient in the Rd arm who died were randomized and had renal impairment but did not receive ≥1 dose of lenalidomide.

Literature Search

A literature search of MEDLINE®, Embase®, BIOSIS Previews®, and Derwent Drug File (and/or other resources, including internal/external databases) was conducted on 03 July 2024.

 

References

1 Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med. 2019;380:2104-2115.  
2 Facon T, Kumar SK, Plesner T, et al. Phase 3 randomized study of daratumumab plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) in patients with newly diagnosed multiple myeloma (NDMM) ineligible for transplant (MAIA). Oral presentation presented at: 60th American Society of Hematology (ASH) Annual Meeting & Exposition; December 1-4, 2018; San Diego, CA.  
3 SK Kumar, P Moreau, N Bahlis, 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. 2022;(Poster).  
4 Facon T, Kumar SK, Orlowski RZ, et al. Final survival analysis of daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in transplant-ineligible patients with newly diagnosed multiple myeloma: MAIA study. Poster presented at: The European Hematology Association (EHA) Hybrid Congress; June 13-16, 2024; Madrid, Spain.  
5 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: The German Society for Hematology and Medical Oncology (DGHO) Annual Conference; October 13-16, 2023; Hamburg, Germany.  
6 Facon T, Cook G, Usmani SZ, et al. Daratumumab plus lenalidomide and dexamethasone in transplant-ineligible newly diagnosed multiple myeloma: frailty subgroup analysis of MAIA. Leukemia. 2022;36(4):1066-1077.  
7 Facon T, Kumar S, Weisel K, et al. Daratumumab plus lenalidomide and dexamethasone in patients with transplant-ineligible newly diagnosed multiple myeloma: MAIA age subgroup analysis. 2022;(Poster).  
8 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-.  
9 Perrot A, Facon T, Plesner T, et al. Health related quality of life for frail transplant ineligible patients with newly diagnosed multiple myeloma treated with daratumumab, lenalidomide, and dexamethasone: subgroup analysis of MAIA trial. 2022;(Oral presentation).  
10 Facon T, Kumar S, Plesner T, et al. Time to response, duration of response, and patient-reported outcomes (PROs) with daratumumab (DARA) plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) alone in transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM): subgroup analysis of the phase 3 MAIA study. Poster presented at: American Society of Clinical Oncology (ASCO) Annual Meeting; June 3-7, 2022; Chicago, IL/Virtual Meeting.  
11 San-Miguel J, Avet-Loiseau H, Paiva B, et al. Sustained minimal residual disease negativity with daratumumab in newly diagnosed multiple myeloma: MAIA and ALCYONE. Blood. 2022;139:492-501.  
12 P Moreau, T Facon, SZ Usmani, et al. Treatment duration and long-term outcomes with daratumumab in transplant-ineligible newly diagnosed multiple myeloma from the phase 3 MAIA study. 2022;(Oral Presentation).  
13 Perrot A, Facon T, Kumar S, et al. Sustained improvement in health-related quality of life in transplant-ineligible patients with newly diagnosed multiple myeloma treated with daratumumab, lenalidomide and dexamethasone versus lenalidomide and dexamethasone: update of the phase 3 MAIA trial. Poster presented at: 63rd American Society of Hematology (ASH) Annual Meeting & Exposition; December 11-14, 2021; Atlanta, GA/Virtual.  
14 Perrot A, Facon T, Kumar S, et al. Meaningful changes in patient-reported outcomes in relation to best clinical response and disease progression: post hoc analyses from MAIA. Poster presented at: 63rd American Society of Hematology (ASH) Annual Meeting & Exposition; December 11-14, 2021; Atlanta, GA/Virtual.  
15 Nahi H, Usmani SZ, Mateos MV, et al. Efficacy of daratumumab, lenalidomide, and dexamethasone in transplant-ineligible patients with newly diagnosed multiple myeloma and impaired renal function from the phase 3 MAIA study based on lenalidomide starting dose. Poster presented at: 63rd American Society of Hematology (ASH) Annual Meeting & Exposition; December 11-14, 2021; Atlanta, GA/Virtual Meeting.  
16 Moreau P, Facon T, Usmani S, et al. Effect of lenalidomide (R) ¬± dexamethasone (d) discontinuation on daratumumab efficacy in multiple myeloma (MM): subgroup analysis of the phase 3 MAIA and POLLUX studies. presented at: 8th Annual Meeting of the Society of Hematologic Oncology (SOHO); September 9-12, 2020; Virtual.  
17 Cavo M, San-Miguel J, Usmani SZ, et al. Prognostic value of minimal residual disease negativity in myeloma: combined analysis of POLLUX, CASTOR, ALCYONE, and MAIA. Blood. 2021;139(6):835-844.  
18 Cavo M, San-Miguel J, USmani S, et al. Supplement to: Prognostic value of minimal residual disease negativity in myeloma: combined analysis of POLLUX, CASTOR, ALCYONE, MAIA. Blood. 2022;139:835-844.  
19 Wang J, Arroyo-Suarez R, Dasari S, et al. Early versus late response to daratumumab-based triplet therapies in patients with multiple myeloma: a pooled analysis of trials POLLUX, CASTOR and MAIA. Leuk Lymphoma. 2022;63(7):1669-1677.  
20 Bahlis N, Facon T, S. Usmani, et al. Daratumumab plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) in patients with newly diagnosed multiple myeloma (NDMM) ineligible for transplant: updated analysis of MAIA. Poster presented at: 61st American Society of Hematology (ASH) Annual Meeting & Exposition; December 7-10, 2019; Orlando, FL.  
21 Kumar S, Facon T, Usmani S, et al. Updated analysis of daratumumab plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) in patients with transplant-ineligible newly diagnosed multiple myeloma (NDMM): the phase 3 MAIA study. Virtual 62nd American Society of Hematology (ASH) Annual Meeting & Exposition. 2020.  
22 Facon T, Kumar S, Plesner T, et al. Supplement to: Daratumumab, lenalidomide, and dexamethasone versus lenalidomide and dexamethasone alone in newly diagnosed multiple myeloma (MAIA): overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2021;22(11):1582-1596.  
23 San-Miguel J, Avet-Loiseau H, Paiva B, et al. Supplement to: Sustained minimal residual disease negativity with daratumumab in newly diagnosed multiple myeloma: MAIA and ALCYONE. Blood. 2022;139:492-501.  
24 Perrot A, Facon T, Plesner T, et al. Health-Related Quality of Life in Transplant-Ineligible Patients With Newly Diagnosed Multiple Myeloma: Findings From the Phase III MAIA Trial. J Clin Oncol. 2021;39(3):227-237.