Abstract
The treatment of multiple myeloma has evolved markedly in the last decade, but mortality remains high, emphasizing the need for more effective therapies. Daratumumab, a fully human monoclonal antibody targeting CD38, has shown clinical efficacy in relapsed/refractory multiple myeloma both as monotherapy and in combination with other drugs, including novel agents. More recently, promising results have been reported in patients with untreated newly diagnosed multiple myeloma (NDMM). Clinical trials thus far have shown enhanced efficacy and tolerability of several daratumumab-based combinations in both transplant ineligible and eligible patients, without compromising transplant ability. However, benefit in high-risk subpopulations is still unclear. A subcutaneous formulation of daratumumab has been introduced to decrease the risk of infusion reactions, with preliminary results showing non-inferior efficacy. The antimyeloma activity of daratumumab is achieved through multiple mechanisms including direct, Fc-dependent, and immunomodulatory mechanisms. Enhanced efficacy of daratumumab in combination with immunomodulatory drugs and proteasome inhibitors is supported by preclinical data showing synergism. This review will focus on the role of daratumumab in untreated NDMM patients, highlighting the results of major clinical trials, and listing ongoing trials that are evaluating various daratumumab-based combinations in this setting.
Keywords: daratumumab, newly diagnosed multiple myeloma, untreated multiple myeloma
Introduction
Multiple myeloma (MM) is a neoplastic clonal plasma cell disorder characterized by secretion of a monoclonal protein in the blood or urine and organ dysfunction manifesting as anemia, hypercalcemia, renal failure, and lytic bone lesions.1 MM is more commonly diagnosed in men and older patients between the age of 65 and 74 years, and accounts for 1.8% of all new cancer cases in the US.2 Treatment strategies in MM have evolved markedly in the past decade with the introduction of novel treatment agents.3–5 This has expanded the treatment options for both newly diagnosed multiple myeloma (NDMM) and relapsed or refractory (R/R) disease. These include immunomodulatory drugs (IMiD) like thalidomide, lenalidomide, and pomalidomide; and proteasome inhibitors (PI) like bortezomib, carfilzomib, and ixazomib. The heterogeneity of drug classes and nonoverlapping mechanisms of action has allowed their use in combination to achieve greater responses.3–5 In the absence of direct comparisons among all available combinations, the choice of initial treatment is currently guided by physician and patient preference, taking into account disease stage, transplant eligibility, risk stratification based on cytogenetic abnormalities, comorbidities, frailty, and functional status.6 Despite advances in treatment, MM mortality remains high, with 5-year overall survival estimated at 52.2%.2 This highlights the need for more effective therapeutic options. The CD38 targeted monoclonal antibody (mAb) daratumumab has shown clinical efficacy in R/R MM both as monotherapy, and in combination with other drugs, including novel agents, leading to its approval for use in this setting. More recently, it has been explored in untreated NDMM patients, with promising results in both transplant eligible and ineligible patients. The role of daratumumab in untreated NDMM patients will be the primary focus of this review.
Mechanisms of daratumumab activity
Daratumumab is an IgG1κ fully human mAb that targets CD38, a type II transmembrane glycoprotein composed of extracellular, transmembrane, and intracellular domains.7 CD38 is widely expressed on nonhematopoietic and hematopoietic tissues, including plasma cells (PCs), natural killer (NK) cells, lymphoid cells, and myeloid cells. Its high expression in MM has made it an attractive target for treatment.8 CD38 serves as a receptor for CD31. Upon ligand binding, CD38 interacts with other cell surface receptors, resulting in activation of intracellular signaling pathways, leading to cellular responses ranging from proliferation to apoptosis.7,9 In addition, CD38 is considered an ectoenzyme using NAD+ and NADP+ as its main substrates.7,9 The antimyeloma activity of daratumumab is exerted through several mechanisms: direct, Fc-dependent, and immunomodulatory mechanisms.9 Direct effects are mediated by inhibition of intracellular signal transduction, and by inhibition of CD38 enzymatic activity, which leads to decreased levels of immunosuppressive adenosine. Fc-dependent mechanisms include complement-dependent cytotoxicity (CDC),8 antibody-dependent cellular cytotoxicity (ADCC),8 antibody-dependent cellular phagocytosis (ADCP),10 and induction of apoptosis through cross-linking via the Fcγ receptor.11 In addition, daratumumab exerts immunomodulatory effects by increasing the production of helper and cytotoxic T cells and depleting CD38-expressing immunosuppressive cells like regulatory T (Tregs) and B cells, and myeloid-derived suppressor cells (MDSCs).12
The rationale for daratumumab-based combinations
The benefit of adding daratumumab to other treatment regimens, particularly IMiD- and PI- based combinations, is supported by preclinical data. Daratumumab has exhibited synergistic effects in enhancing MM cell lysis ex vivo when combined with lenalidomide or bortezomib, particularly in the triplet combination. Interestingly, this effect was more pronounced in myeloma cells of patients who had low response or refractoriness to treatment with lenalidomide or bortezomib.13 This implies that daratumumab may have the potential to reinstate the host susceptibility to these agents through its immunomodulating effects.14 The ability of IMiDs, including thalidomide, lenalidomide, and pomalidomide, to activate immune effector cells like NK cells or reduce inhibitory T cell populations in vitro, may be a potential explanation for this synergism.15 Lenalidomide, in particular, has been shown to augment daratumumab-induced myeloma cell lysis by ADCC by inducing endogenous NK cells,14 and to potentiate ADCP by inducing endogenous macrophages through vitamin D pathway activation.16 Furthermore, lenalidomide has the potential to sensitize Tregs to the action of daratumumab by upregulating CD38 expression on Tregs, and by increasing the fraction of CD38-high Tregs.17 Lenalidomide has not been shown to influence complement-mediated cytotoxicity.18
Role of daratumumab in R/R MM
Since initial use in 2015,19 there has been clear evidence for daratumumab efficacy in pretreated MM patients both as monotherapy and in combination with other agents. The GEN501 and SIRIUS trials provided the earliest reports on efficacy and safety of daratumumab as monotherapy in heavily pretreated R/R MM patients. A combined analysis showed durable responses after 36.6 months of median follow up, with an overall response rate (ORR) of 30.4%, median overall survival (OS) of 20.5 months (CI 16.6–28.1), and 3-year OS rate of 36.5% (28.4–44.6).19–21 Results from these studies led to the approval of daratumumab monotherapy in patients with at least three lines of prior therapy including an IMiD and PI, or double refractory to an IMiD and PI. Evidence for clinical efficacy in combination with bortezomib and lenalidomide in R/R patients was provided by the CASTOR and POLLUX trials, respectively.22,23 In the CASTOR trial, the combination of daratumumab with bortezomib and dexamethasone (DVd) was superior to bortezomib and dexamethasone (Vd) alone, achieving significantly longer progression-free survival (PFS) (median PFS: 16.7 versus 7.1 months at 31.3 months median follow up) and superior overall and deeper responses, including minimal residual disease (MRD) negativity.24 After showing safety and efficacy in the phase I/II GEN503 trial,25,26 the combination of daratumumab with lenalidomide and dexamethasone (DRd) was found to be superior to lenalidomide and dexamethasone alone (Rd) in R/R MM in the phase III POLLUX trial.23 The addition of daratumumab to Rd was associated with longer PFS; at a median follow up of 39.5 months, median PFS was NR versus 17.5 months with hazard ratio (HR) 0.44, 0.35–0.55 p < 0.0001. Similarly, greater overall and deeper responses were achieved, including significantly improved and sustained MRD negativity rate.27 Together, these results lead to the approval of daratumumab in combination with Vd or Rd in R/R MM patients.28 The combination of daratumumab, pomalidomide, and dexamethasone (Dara-Pom-Dex) has also gained FDA approval for use in R/R patients based on results from the EQUULEUS trial, showing an ORR of 60% and MRD negativity of 29%.29 Interestingly, retreatment with the Dara-Pom combination in patients who were previously refractory to Dara or Pom was associated with response in a retrospective study.30 This regimen is not yet approved for use in Europe and other countries outside the US. Encouraging results have also been reported for daratumumab in combination with carfilzomib and dexamethasone in R/R patients following one to three lines of therapy.31
Role of daratumumab in untreated MM
The efficacy of daratumumab observed in R/R MM, particularly among patients with fewer lines of prior therapy,24 has been replicated in untreated MM, expanding its labeled use to include transplant-ineligible NDMM.32 This was based on results from the ALCYONE and MAIA trials, showing improved responses when daratumumab is combined with Rd or melphalan-bortezomib-prednisone (MVP) respectively.33,34 In addition, encouraging results are being reported in other combinations with standard-of-care regimens, and deeper responses are being achieved even in transplant-eligible patients, without compromising the ability to proceed to transplant. We discuss below the most important clinical trials evaluating daratumumab combinations in NDMM.
Daratumumab in transplant-ineligible NDMM
The dosing and safety of daratumumab, in combination with standard regimens in NDMM patients, were established in a phase Ib trial where four different daratumumab-based combinations were used, including melphalan, bortezomib, thalidomide, and pomalidomide.35 Subsequently, the phase III ALCYONE trial evaluated the efficacy of daratumumab in combination with bortezomib-melphalan-dexamethasone (D-VMP), compared with bortezomib-melphalan-dexamethasone alone (VMP), among 706 transplant ineligible NDMM patients. After a median follow up of 16.5 months, the D-VMP group had significantly improved PFS compared with the VMP group with a median PFS NR versus 18.1 months, respectively and HR 0.5 (95% CI 0.38–0.65 p < 0.001). The superiority of the daratumumab combination was maintained in patients who were ⩾75 years, had higher ISS stage, poor performance status, impaired hepatic or renal function. However, patients with high-risk cytogenetics (53 patients) appeared to have less benefit compared with the patients with standard-risk cytogenetics (261 patients) (HR: 0.78, 0.43–1.43 versus 0.39, 0.28–0.55, respectively). The overall response, very good partial response (VGPR), complete response (CR), and stringent complete response (sCR) rates, were all significantly higher in the daratumumab group. The MRD rate was also increased significantly in the D-VMP group (22.3% versus 6.2% p < 0.001).33 An updated analysis was recently reported after a median follow up of 27.8 months, showing continued responses in the D-VMP group (Median PFS NR versus 19.1 months in the D-VMP and control groups, respectively and HR 0.43, 0.35–0.54 p < 0.0001). The rate of grade 3–4 infection was higher in the D-VMP group compared with the control group (25.1% versus 14.7%, respectively). In particular, pneumonia was higher in the D-VMP group (12.4% versus 4%). However, these caused treatment discontinuation in only a small number of patients.36 The clinical efficacy of DRd, compared with Rd, was evaluated in the phase III MAIA trial among 737 transplant ineligible NDMM patients. Rd is one of the standard treatments used in this group of patients, particularly elderly patients unable to tolerate triplet regimens.37 Results from a preplanned interim analysis of the MAIA trial were recently reported. At a median follow up of 28 months, PFS was longer in the DRd group compared with the Rd group, with median PFS NR versus 31.9 months, respectively and HR 0.56 (95% CI 0.43 –0.73 p < 0.001). Superiority was maintained in patients ⩾75 years, but not in the subgroup of patients with high risk cytogenetics (HR: 0.85, 0.44–1.65).34 This is in contrast to results from the POLLUX study, comparing DRd with Rd in R/R patients, where PFS was longer in the subgroup with high-risk cytogenetics, albeit to a lesser extent that standard-risk patients (HR: 0.53, 0.25–1.13 versus 0.30, 0.20–0.47, respectively).27,38 Patients in the DRd group experienced a higher rate of deeper responses including ⩾CR (47.6% versus 24.9%). Similarly, the rates of overall response, ⩾VGPR and MRD negativity (24.2% versus 7.3%) were all higher in the DRd group. OS data is still immature. Overall, adverse events were manageable. The most common grade 3–4 adverse events in the DRd and Rd groups were neutropenia (50% versus 35.3% respectively) and infections (32.1% versus 23.3% respectively). Pneumonia was the most common infection, occurring in 13.7% versus 7.9%, and leading to death in 0.5% versus 0.8% of patients in the DRd and Rd groups, respectively.34 Based on results from these two trials, D-VMP has been approved in the US and Europe, and DRd has been approved only in the US, for use as first line treatments in transplant ineligible MM patients.
Given the lack of direct head-to-head comparisons, there is little evidence to guide treatment choice between the various standard regimens at this time. A recently published metanalysis by Cao and colleagues compared the efficacy of currently used treatment regimens in this group of patients using Rd as reference. In terms of PFS, three combinations showed superiority to Rd: DRd (HR: 0.57, 0.43–0.73), D-VMP (HR, 0.59, 0.36–0.91), and VRd (HR, 0.72, 0.56–0.90). VRd showed superior OS, but the analysis excluded daratumumab-based regimens given immaturity of OS data.39 The efficacy of daratumumab is also being evaluated in other PI-based doublet regimens, specifically in frail patients with NDMM. The phase II HOVON 143 study is evaluating the efficacy of daratumumab in combination with ixazomib and low-dose dexamethasone in this subset of patients. In its first planned safety analysis, the combination was tolerated with manageable side effects. A preliminary ORR of 70% has been reported, including 20% VGPR. Updated efficacy results are awaited.40 Ongoing trials of daratumumab-based combinations in transplant ineligible NDMM patients are listed in Table 1.41
Table 1.
Trials evaluating daratumumab combinations in transplant ineligible patients with NDMM.
| NCT number | Title | Conditions | Interventions | Dosing and schedule | Primary outcome | Phase | Status |
|---|---|---|---|---|---|---|---|
| NCT02252172 | Study comparing daratumumab, lenalidomide, and dexamethasone with lenalidomide and dexamethasone in participants with previously untreated multiple myeloma (MAIA)34 | Transplant ineligible with untreated NDMM | Arm 1: DRd Arm 2 (control): Rd |
28-day cycles: Len: 25 mg PO D1–21 Dex: 40 mg PO/IV D1,8,15,22 Dara (arm 1 only): 16 mg/kg IV Qw ×8w, then Q2w ×16w, then Q4w |
PFS Time | III | Active, not recruiting |
| NCT02195479 | A study of combination of daratumumab and velcade (bortezomib) melphalan-prednisone (DVMP) compared with velcade melphalan-prednisone (VMP) in participants with previously untreated multiple myeloma (ALCYONE)33 | Transplant ineligible with untreated NDMM | Arm 1 (control): VMP Arm 2: D-VMP |
Up to 9 cycles, 42 days each: Bort: 1.3 mg/m2 SC 2×/w in W1,2,4,5 of C1, then Qw in W1,2,4,5 of C2–9 Mel: 9 mg/m2 PO QD D1–4 of C1–9 Pred: 60 mg/m2 PO QD D1–4 of C1–9 Dara (arm 2 only): 16 mg/kg IV Qw ×6w in C1, then Q3w in C2-9, then Q4w |
PFS | III | Active, not recruiting |
| NCT03993912 | Compare lenalidomide and subcutaneous daratumumab versus lenalidomide and dexamethasone in frail subjects with previously untreated multiple myeloma who are ineligible for high dose therapy (IFM2017_03) | Transplant ineligible, elderly (⩾65) frail, with untreated NDMM | Arm 1: Dara SC + Len + Dex Arm 2 (control): Len + Dex |
28-day cycles: Arm 1: Len: 25 mg PO D1–21 Dex: 20 mg PO D1, 8,15,22 ×2 cycles only Dara: 1800 mg SC Qw ×8w, then Q2w ×16w, then Q4w Arm 2: Len: 25 mg PO D1–21 Dex: 20 mg PO D1, 8,15,22 |
PFS | III | Not yet recruiting |
| NCT03742297 | Treatment for elderly fit newly diagnosed multiple myeloma patients aged between 65 and 80 years | Elderly (65–80), fit transplant ineligible with untreated NDMM |
Arm 1 (control): induction with VMP×9 + Rd×9, consolidation with Rd + low dose-Dara ×4 Arm 2a: induction with KRd×18, consolidation with Rd + low dose-Dara ×4 Arm 2b: induction with D-KRd×18 Part 2: randomization to: Arm 1: no maintenance Arm 2: Dara + Len |
Induction: Arm 1: VMP: one 6-week cycle and eight 4-week cycles: Mel: 9 mg/m2 PO D1–4 Pred: 60 mg/m2 PO D1–4 Bort: 1.3 mg/m2 D1,4,8,11,22,25,29,32 of C1, then D1,8,15,22 of C2–9 Rd: Len: 25 mg PO D1–21 Dex: 40 mg D1,8,15,22 Arm 2a (28-day cycles): Car: C1: 20 mg/m2 D1 and 36 mg/m2 D2,8,9,15,16. C2: 36 mg/m2 D1,2,8,9,15,16. C3–18: 56 mg/m2 D1,8,15 Len: 25 mg D1–21 Dex: 40 mg D1,8,15,22 Arm 2b (28-day cycles): Car: C1: 20 mg/m2 D1 and 36 mg/m2 D2,8,9,15,16. C2: 36 mg/m2 D1,2,8,9,15,16. C3–18: 56 mg/m2 D1,8,15 |
Immunophenotypic CR rate at 18 months | III | Recruiting |
| Len: 25 mg PO D1–21 Dex: 40 mg D1,8,15,22 Dara: 16 mg/Kg IV D1,8,15,22 of C1–2, D1–15 of C3-4 and D1 of C5–18 Consolidation: arm 1&2a: Len: 25 mg PO D1–21 Dex: details not available Dara: low dose, details not available Part 2: Maintenance: Len and Dara up to 2 years, then Len until progression |
|||||||
| NCT03652064 | A study comparing daratumumab, Velcade (Bortezomib), lenalidomide, and dexamethasone (D-VRd) with Velcade, lenalidomide, and dexamethasone (VRd) in participants with untreated multiple myeloma and for whom hematopoietic stem cell transplant is not planned as initial therapy (CEPHEUS) | Untreated NDMM and SCT not planned as Initial Therapy | Arm 1 (control): VRd, then Rd Arm 2: D-VRd then DRd |
C1-8: 21-days each. C9+: 28 days each: Bort: 1.3 mg/m2 SC 2×/w on D1,4,8,11 of C1–8 Len: 25 mg PO D1–14 for C1–8 and D1–21 for C9+ Dex: 20 mg PO D1,2,4,5,8,9,11,12 of C1–8 and 40 mg PO D1,8,15,22 of C9+ Dara (arm 2 only): 1800 mg SC Qw for C1–2, then Q3w for C3–8, and Q4w for C9+ |
% with negative MRD status | III | Recruiting |
| NCT03217812 | A study of Velcade (Bortezomib) melphalan-prednisone (VMP) compared with daratumumab in combination with VMP (D-VMP), in participants with previously untreated multiple myeloma who are ineligible for high-dose therapy (Asia Pacific region) | Transplant ineligible with untreated NDMM |
Arm 1 (control): VMP Arm 2: D-VMP |
Up to 9 cycles, 42 days each: Bort: 1.3 mg/m2 SC 2×/w in W1,2,4,5 of C1, then Qw in W1,2,4,5 of C2-9 Mel: 9 mg/m2 (4.5 if Cr >2) PO QD D1-4 of C1-9 Pred: 60 mg/m2 PO QD D1-4 of C1-9 Dara (arm 2 only): 16 mg/kg IV Qw ×6w in C1, then Q3w in C2-9, then Q4w |
⩾VGPR rate at 6 months and 3 years after last participant 1st dose | III | Recruiting |
| NCT04052880 | Study of subQ Dara with dose-attenuated bortezomib, lenalidomide, dexamethasone in elderly NDMM | Transplant ineligible elderly (age ⩾70) with untreated NDMM | 1 arm: SC Dara with dose-attenuated VRd ×12. Maintenance with Dara with either Len or Ixa. Maintenance choice is based on cytogenetic risk at diagnosis: Dara-Ixa if high-risk, defined by FISH with t(4;14) only, and Dara-Len in all others | Induction (28-day cycles): Dara: 1800 mg SC Qw in C1–2, then Q2w in C3–6, then Q4w Bort: 1.3 mg/m2 SC D1,8,15 Len 15 mg (10 if CrCl 30–60 ml/min, 5 if CrCl 15–30 ml/min) PO QD D1–21 Dex: A total of 20 mg PO Qw Maintenance (28-day cycles): Dara: 1800 mg SC Q4w Len: 1 dose level below C1–12 dosing (10 mg QD if CrCl >60, 5 QD if CrCl 30–60 ml/min, 5 QOD if CrCl 15–30 ml/min) Ixa: 3 mg (2.3 Qw if CrCl 15–30 ml/min) PO QD D1,8,15 |
⩾VGPR rate after 8 cycles | II | Not yet recruiting |
| NCT04009109 | Study of lenalidomide/ixazomib/dexamethasone/daratumumab in transplant-ineligible patients with newly diagnosed MM | Transplant ineligible with untreated NDMM | Arm 1: induction with Len, Ixa, Dara and Dex ×12. Maintenance with Len Arm 2: induction with Len, Ixa, Dara and Dex ×12. Maintenance with Len, Ixa and Dara |
Induction (28-day cycles): Arm 1 and 2: Len: 15 mg PO QD D1–21 Ixa: 4 mg PO D1,8,15 Dara: 16 mg/kg IV Qw ×8w, then Q2w ×16w, then Q4w Dex: 20 mg PO D1,2,8,9,15,16 Maintenance (28-day cycles, up to 2 years): Len: 10 mg PO QD D1-21 Ixa (arm 2 only): 3 mg PO D1,8,15 Dara (arm 2 only): 16 mg/kg IV D1 |
PFS | II | Not yet recruiting |
| NCT03695744 | Daratumumab in combination with bortezomib and dexamethasone in newly diagnosed transplant ineligible multiple myeloma (AMN006) | Transplant ineligible with untreated NDMM | 1 arm: DVd ×9 months followed by Dara | Dara: 16 mg/kg IV Qw in W1-9, then Q3w in W10-24, then Q4w in W25+Bort: SC Qw Dex: 40 mg (20 mg if >75 years) PO Qw |
ORR | II | Not yet recruiting |
| NCT02918331 | A study of JNJ-54767414 (daratumumab) in combination with lenalidomide and dexamethasone in Japanese participants with previously untreated multiple myeloma who are ineligible for high-dose therapy and autologous stem cell transplantation | Transplant ineligible with untreated NDMM |
1 arm: DRd | 28-day cycles: Dara: 16 mg/kg IV Qw ×8w, then Q2w ×16w, then Q4w Len: 25 mg (10 if CrCl 30–60) PO D1-21 Dex: 40 mg D1,8,15,22 |
Safety: DLT | Ib | Active, not recruiting |
Bort, bortezomib; Car, carfilzomib; CR, complete response; D-KRd, daratumumab, carfilzomib, lenalidomide, dexamethasone; D-VMP, daratumumab, bortezomib, melphalan, prednisone; D-VRd, daratumumab, bortezomib, lenalidomide, dexamethasone; Dara, daratumumab; Dex, dexamethasone; DLT, dose-limiting toxicity; DRd, daratumumab, lenalidomide, dexamethasone; DVd, daratumumab, bortezomib, dexamethasone; IV, intravenous; Ixa, ixazomib; KRd, carfilzomib, lenalidomide, dexamethasone; Len, lenalidomide; Mel; melphalan; MRD, minimal residual disease; NCT, National Clinical Trial; NDMM, newly diagnosed multiple myeloma; ORR, overall response rate; PFS, progression-free survival; PO, by mouth; Pred, prednisone; QD, daily; QOD, every other day; Qw, every week; Rd, lenalidomide; dexamethasone; SC, subcutaneous; SCT, stem cell transplant; dexamethasone; VGPR, very good partial response; VMP, bortezomib, melphalan, prednisone; VRd, bortezomib, lenalidomide.
Daratumumab in transplant-eligible NDMM
Favorable results, including deeper responses, observed in transplant ineligible patients have stimulated efforts to evaluate daratumumab in transplant eligible patients with NDMM. In the phase III CASSIOPEIA trial,42 daratumumab was combined with bortezomib, thalidomide, and dexamethasone (DVTd) during induction and consolidation, showing increased rates of sCR at 100 days post-transplant; the sCR rate was 29% in the DVTd group compared with 20% in the bortezomib, thalidomide, and dexamethasone (VTd) group [odds ratio (OR): 1.60, 1.21–2.12 p: 0.0010]. In addition, the DVTd group achieved higher rates of ⩾CR, ⩾VGPR, and MRD negativity (64% versus 44% p < 0.0001). Superiority, in terms of sCR rate, was maintained in patients ⩾50 years, patients with poor performance status, baseline renal or hepatic dysfunction, but not in patients with ISS stage 3 MM. Patients with high-risk cytogenetics had lower odds for achieving sCR compared with standard risk MM (OR: 0.83, 0.42–1.66). However, both subgroups showed benefit with the daratumumab combination in terms of MRD negativity and PFS. The overall incidence of serious adverse events was similar in both groups (47% in both). There was a higher rate of grade 3–4 cytopenias in the DVTd group. Although infection rate was higher in the DVTd group, the rate of grade 3–4 infection was similar in both groups. The addition of daratumumab was associated with a lower yield of stem cells compared with VTd alone (median collected CD34+ cells 6.3 × 106/kg versus 8.9 × 106/kg) and increase rate of plerixafor use during mobilization. However, the rates of transplant and hematopoietic reconstitution were similar. The impact of daratumumab maintenance in patients achieving ⩾PR from both groups is currently being evaluated in another part of this study.42 The results of the CASSIOPEIA trial led to the FDA approval of Daratumumab in combination with VTd in transplant eligible patients with MM. Initial safety and efficacy reports on the combination of DVRd in transplant eligible NDMM patients were provided by the phase II GRIFFIN study. Patients received DVRd induction and post-transplant consolidation, followed by maintenance with daratumumab and lenalidomide. All 16 included patients proceeded to transplant and achieved ⩾VGPR, including 63% with ⩾CR after consolidation. Of 16 patients, 15 had not progressed at the median follow up of 15.6 months. Adverse events were manageable and did not lead to death or treatment discontinuation in any patient. The trial is ongoing to assess the impact of DVRd compared with VRd alone on sCR rate.43 Based on these initial reports, the phase III PERSUES trial was designed to compare the efficacy of DVRd to VRd in terms of PFS in this group of patients, utilizing the subcutaneous formulation of daratumumab to minimize toxicity.44 Ongoing trials of daratumumab-based combinations in transplant eligible NDMM patients are listed in Table 2.41 The effect of daratumumab-based induction chemotherapy on bone marrow recovery was studied by a group at Mayo Clinic.45 Neutrophil engraftment was found to be delayed among patients treated with daratumumab (12 patients) prior to stem cell mobilization, compared with patients who did not receive daratumumab (129 patients). Median time to engraftment was 19 and 16 days in the 2 groups, respectively (p value: 0.017).45 As daratumumab use in the front-line setting increases, more data is needed to delineate its effect on stem cell mobilization and engraftment, and the clinical implications of these effects.
Table 2.
Trials evaluating daratumumab combinations in transplant eligible patients with NDMM.
| NCT Number | Title | Conditions | Interventions | Dosing and schedule | Primary outcome | Phase | Status |
|---|---|---|---|---|---|---|---|
| NCT02541383 | A study to evaluate daratumumab in transplant eligible participants with previously untreated multiple myeloma (Cassiopeia)42 | Transplant eligible with untreated NDMM | Part 1: Arm 1 (control): VTd induction ×4, SCT, VTd consolidation ×2 Arm 2: D-VTd induction ×4, SCT, D-VTd consolidation ×2 Part 2: maintenance (randomization of patients with ⩾PR) Arm 1: observation Arm 2: Dara |
Part1: Induction and consolidation (28-day cycles): Bort: 1.3 mg/m2 D1,4 in W1, and D8,11 in W2 of each cycle Thal: 100 mg PO QD Dex: 40 mg PO/IV D1,2,8,9,15,16,22,23 of induction C1–2, D1–2 of induction C3–4, 20 mg D8,9,15,16 of induction C3–4, and D1,2,8,9,15, 16 of consolidation cycles Dara (arm 2 only): 16 mg/kg IV Qw in induction C1–2, then Q2w in induction C3–4 and consolidation cycles Part 2: Maintenance (up to 2 years): Dara (arm 2 only): 16 mg/kg Q8w |
sCR after consolidation PFS after maintenance |
III | Active, not recruiting |
| NCT03710603 | Daratumumab, velcade (bortezomib), lenalidomide and dexamethasone compared with velcade, lenalidomide and dexamethasone in subjects with previously untreated multiple myeloma (Perseus)44 | Transplant eligible with Untreated NDMM | Arm 1 (control): VRd induction ×4, SCT, VRd consolidation ×2. Len maintenance Arm 2: D-VRd induction, SCT, D-VRd consolidation ×2. Dara and Len maintenance |
Induction and consolidation (28-day cycles): Bort: 1.3 mg/m2 SC D1,4,8,11 in C1–6 Len: 25 mg PO D1–21 of C1–6 Dex: 40 mg PO QD D1–4 and D9-12 in C1–6 Dara (arm 2 only): 1800 mg SC Qw in C1–2, then Q2w in C3–6 Maintenance (28-day cycles): Len: 10 mg QD PO D1-28 Dara (arm 2 only): 1800 mg SC Q4w |
PFS | III | Recruiting |
| NCT03792620 | Daratumumab intensified treatment to eligible MM new patients CTD-Dara induction, follow by Dara consolidation (MAXDARA) | Transplant eligible with untreated NDMM | 1 arm: induction with CTD-Dara ×4, SCT, consolidation with Dara + Thal, maintenance with Dara | Induction (28-day cycles): Cyclo: 500 mg D1,8,15 Dex: 40 mg Qw Thal: 100–200 mg D1-28 Dara: 16 mg/Kg Qw of C1–2, then Q2w of C3–4 Consolidation: Thal: 100 mg D1-28 ×16w (full consolidation) Dara: 16 mg/Kg Q2w after D+30 (pre consolidation) and Q2w D+90−120 (full consolidation) for 4 doses total Maintenance: Dara: 16 mg/Kg Qm (28 doses) |
Incidence of >VGPR after transplant | III | Recruiting |
| NCT03896737 | Daratumumab-bortezomib-dexamethasone (Dara-VCd) versus bortezomib-thalidomide-dexamethasone (VTd), then maintenance with ixazomib (IXA) or IXA-Dara | Young (⩽65) transplant eligible with untreated NDMM | Part 1: Arm 1: D-VCd induction ×4, SCT, D-VCd consolidation ×2–4, Arm 2: VTd induction ×4, SCT, VTd consolidation ×2–4 Part 2: maintenance (Randomization of patients with ⩾PR) Arm 1: Ixa Arm 2: Ixa + Dara |
Part 1: Induction and consolidation (28-day cycles): Arm 1: Bort: 1.3 mg/m2 SC D1,8,15,22 Cyclo: 300 mg/m2 PO/IV D1,8,15,22 Dex: 40 mg PO/IV D1,8,15,22 Dara: 16 mg/Kg IV D1,8,15,22 of C1-2, D1,15 of C3-4 and during consolidation Arm 2: Bort: 1.3 mg/m2 SC D1,4,8,11 Thal: 100 mg PO D1-28 Dex: 20 mg PO/IV D1,2,3,4,8,9,10,11 Part 2: Maintenance (28-day cycles, up to 24 months): Ixa: 3 mg D1,8,15 of C1–4, then 4 mg D1,8,15 of C5+Dara (arm 2 only): 16 mg/kg D1 |
PFS MRD negativity |
II | Recruiting |
| NCT03669445 | Study association of lenalidomide, ixazomib, dexamethasone and daratumumab in newly diagnosed standard risk multiple myeloma (IFM2018-01) | Transplant eligible with standard Risk NDMM | 1 arm: Len, Ixa, Dex and Dara induction, SCT, consolidation (all 4 drugs) and Len maintenance | Treatment details not available | MRD-negativity rate | II | Recruiting |
| NCT03606577 | An intensive program with quadruplet induction and consolidation plus tandem autologous stem cell transplantation in newly diagnosed high risk multiple myeloma patients (IFM 2018-04) | Transplant eligible with high risk untreated NDMM | 1 arm: D-KRd induction ×6, tandem SCT and D-KRd consolidation ×4 Maintenance with Len and Dara | Induction: Car: 20/36 mg/m² D1,2,8,9,15,16 (20 mg/m² D1,2 in C1) Dex: 20 mg QD D1,2,8,9,15,16,22,23 Dara: 8/16 mg/kg IV D1,8,15,22 in C1–2 and D1,15 in C3–6 (8 mg/kg D1,2 in C1) Len: 25 mg QD D1-21 Consolidation: Car: 20/36 mg/m² D1,2,8,9,15,16 (20 mg/m² D1,2 in C1) Dex: 40 mg QD D1,8,15,22 Dara: 16 mg/kg IV Day 1,15 Len: 15 mg QD D1–21 in C1 and 25 mg/day D1–21 in C2-4 Maintenance (up to 2 years) Dara: 16 mg/kg IV Q8w Len: 10 mg QD D1–21 |
% of patients receiving the second transplant | II | Recruiting |
| NCT03188172 | MUK nine b: optimum treatment protocol (MUKnineb) | Transplant eligible with high risk untreated NDMM or plasma cell leukemia | 1 arm: D-VRd + low-dose cyclo induction ×4–6, SCT, consolidation part 1 with D-VRd ×6, consolidation part 2 with Dara, Bort, Len ×12. Maintenance: Dara, Len | Induction: Cyclo: 500 mg D1,8 Dara: 16 mg/kg, D1,8,15 (C1,2) and D1 in C3+ Bort: 1.3 mg/m² D1,4,8,11 Len: 25 mg D1–14 Dex: 20–40 mg D1,4,8,11 Consolidation 1: Bort: 1.3 mg/m² D1,8,15,22 Len: 25 mg D1-21 Dex: 20–40 mg D1,8,15,22 Dara: 16 mg/kg D1 Consolidation 2: Dara: 16 mg/kg D1 Bort: 1.3 mg/m² D1,8,15 Len: 25 mg D1-21 Maintenance: Dara: 16 mg/kg D1 Len: 10 mg D1-21 |
PFS | II | Recruiting |
| NCT03004287 | 2015-12: A study exploring the use of early and late consolidation/maintenance therapy | Transplant eligible with untreated or ⩽4 cycles of systemic therapy, high-risk MM | 1 arm: KTd-Dara-PACE, SCT1, consolidation 1: Dara-Kd +/– SCT 2, consolidation 2: Dara. Maintenance: Dara-Kd alternating with Dara-Rd (alternating 3-month blocks) | Induction: Car: IV D1,2 Thal: PO D1–4 Dex: PO/IV D1–4 Dara: IV D1 Cisplatin: IV continuous infusion D1–4 Adriamycin: IV continuous infusion D1–4 Cyclo: IV continuous infusion D1–4 Etoposide: IV continuous infusion D1–4 Consolidation 1: Car: IV D1,8,15,22 Dara: IV D1,8 Dex: PO/IV D–4 and D–1 of SCT Consolidation 2: Dara: IV D1,8 Maintenance: Dara: IV D1 Car: IV D1,8,15,22 Len: PO D1–21 Dex: PO/IV D1,8,15,22 |
PFS | II | Recruiting |
| NCT02874742 | Study comparing daratumumab, lenalidomide, bortezomib, and dexamethasone (D-RVd) versus lenalidomide, bortezomib, and dexamethasone (RVd) in subjects with newly diagnosed multiple myeloma | Transplant eligible with untreated NDMM |
Arm 1: D-RVd induction ×4, SCT, consolidation ×2, then maintenance Arm 2: RVd induction ×4, SCT, consolidation ×2, maintenance |
Induction and consolidation (21-day cycles): Len: 25 mg PO D1–14 Bort: 1.3 mg/m² SC D1,4,8,11 Dex: 20 mg PO D1,2,8,9,15,16 Dara (arm 1 only): 16 mg/kg IV Qw in induction, then Q3w in consolidation Maintenance (28-day cycles, up to 24 months): Len: 10 mg PO D1-21 in C7-9 and 15 mg D1-21 in C10+ Dex: 20 mg PO D1,2,8,9,15,16 Dara (arm 1 only): 16 mg/kg IV Q4 or Q8w |
sCR rate by the end of post-SCT consolidation | II | Active, not recruiting |
| NCT02955810 | Cyclophosphamide-bortezomib-dexamethasone (CyBorD) with daratumumab (Dara) (CyBorD-Dara)46 | Transplant eligible with untreated NDMM | 1 arm: Dara-CyBorD induction ×4, SCT, Dara-CyBorD consolidation ×2, maintenance with Dara (and Bort if high-risk disease) | Induction and consolidation: Cyclo: PO D1,8,15,22 Bort: SC D1,8,15,22 Dex: 20 mg PO D1,2,8,9,15,16,22,23 Dara: 16 mg/kg IV D1,8,15,22 in C1,2 and D1,15 in C3,4 and consolidation cycles Cyclo and Bort dosing - 3 dose levels: L1: cyclo 150 mg/m2, bort 1.3 mg/m2 L2: cyclo 300 mg/m2, bort 1.3 mg/m2 L3: cyclo 300 mg/m2, bort 1.5 mg/m2 −1L: cyclo 100 mg/m2, bor 1.3 mg/m2 Maintenance (up to 2 years): Dara: 16 mg/kg IV D1 Q4w Bort (if high-risk disease): SC D1,15 |
MTD CR rate post SCT |
Ib | Active, not recruiting |
Bort, bortezomib; Car, carfilzomib; CR, complete response; CTD-Dara, cyclophosphamide, thalidomide, dexamethasone, daratumumab; Cyclo, cyclophosphamide; CYBORD, cyclophosphamide, bortezomib and dexamethasone; D-KRd, daratumumab, carfilzomib, lenalidomide, dexamethasone; D-RVd, daratumumab, lenalidomide, dexamethasone, bortezomib; D-VCd, daratumumab, bortezomib, cyclophosphamide, dexamethasone; D-VRd, daratumumab, bortezomib, lenalidomide, dexamethasone; D-VTd, daratumumab, bortezomib, thalidomide, dexamethasone; Dara, daratumumab; Dex, dexamethasone; IV, intravenous; Ixa, ixazomib; Kd, carfilzomib, dexamethasone; KTd, carfilzomib, thalidomide, dexamethasone; Len, lenalidomide; MRD, minimal residual disease; MTD, maximum tolerated dose; NCT, National Clinical Trial; NDMM, newly diagnosed multiple myeloma; PACE, cisplatin, doxorubicin, cyclophosphamide, etoposide; PFS, progression-free survival; PO, by mouth; PR, partial response; QD, daily; Qw, every week; Rd, lenalidomide, dexamethasone; RVd, lenalidomide, dexamethasone, bortezomib; SC, subcutaneous; sCR, stringent complete response; SCT, stem cell transplant; Thal, thalidomide; VGPR, very good partial response; VRd, bortezomib, lenalidomide, dexamethasone; VTd, bortezomib, thalidomide, dexamethasone.
Daratumumab in NDMM irrespective of transplant eligibility
In addition to the aforementioned studies, other trials are investigating daratumumab combinations among NDMM patients, irrespective of transplant eligibility. A phase II clinical trial at Mayo Clinic was designed to evaluate the efficacy of the daratumumab in combination with ixazomib, lenalidomide and dexamethasone (D-IRd) as an induction regimen in this group of patients with a median age of 62 years (41–81). Early reports have shown encouraging results. Among 38 patients, 90% achieved ⩾PR after only two cycles, and all patients (32 patients) who completed four cycles achieved ⩾PR, including VGPR in 50%. Stem cell yield was not adversely influenced by treatment, and there were no reports of treatment discontinuation related to adverse events. Updated analysis including results on MRD is awaited.47 The phase II Lyra trial studied the efficacy of daratumumab in combination with CYBORD in a heterogeneous group of MM patients including transplant eligible and ineligible NDMM patients and patients who relapsed after one line of therapy. Among patients with newly diagnosed MM (87 patients), 44% achieved ⩾VGPR after four induction cycles, including 5% with CR, with an ORR of 79%. Responses further improved by the end of induction with longer treatment; at a median of six induction cycles, ORR, ⩾VGPR rate and CR rate increased to 81%, 56%, and 9% respectively. At 12 months, PFS was 87% and OS 99%. Adverse events, including infusion reactions, were consistent with previous studies.48 Ongoing trials of daratumumab-based combinations in NDMM patients irrespective of transplant eligibility are listed in Table 3.41
Table 3.
Trials evaluating daratumumab combinations in NDMM irrespective of transplant eligibility.
| NCT Number | Title | Conditions | Interventions | Dosing and schedule | Primary Outcome | Phase | Status |
|---|---|---|---|---|---|---|---|
| NCT03942224 | Daratumumab, ixazomib, and dexamethasone or daratumumab, bortezomib, and dexamethasone in patients with newly diagnosed multiple myeloma (DeRIVE) | NDMM | Arm 1: induction with Did ×8 +/– SCT, maintenance with DId (up to 24 months) Arm 2: induction 1 with DVd ×3 + induction 2 with Did ×5 +/– SCT. Maintenance with DId |
Induction: Arm 1 (28-day cycles): Dara: IV D1,8,15,22 of C1-2 and D1,15 of C3-8 Ixa: PO D1,8,15 Dex: IV/PO D1,8,15,22 Arm 2: Induction 1 (21-day cycles): Dara: IV D1,8,15 Bort: SC D1,4,8,11 Dex: IV/PO D1,8,15 Induction 2 (28-day cycles): Dara: IV D1,15 Ixa: PO D1,8,15 Dex: IV/PO D1,8,15,22 Maintenance (up to 24 months) Arm 1 (28-day cycles): Dara: IV D1 Ixa: PO D1,8,15 Dex: IV/PO D1,8,15,22 Arm2 (28-day cycles): Dara: IV D1 Ixa: PO D1,8,15 Dex: IV D1 |
⩾VGPR rate | II | Recruiting |
| NCT03500445 | Daratumumab, carfilzomib, lenalidomide and low dose dexamethasone (DKRd) in newly diagnosed, multiple myeloma | Transplant eligible or ineligible untreated NDMM | 1 arm: D-KRd ×24 | Dara: 16 mg/kg IV. C1–2: Qw. C3-8: Q2w. C9–24: D1 Car: C1: 20 mg/m2 IV D1,2 and 36 mg/m2 D8,9,15,16. C2–9: 36 mg/m2 D1,2,8,9,15,16. C9-24: 36 mg/m2 D1,2,15,16 Len: 25 mg PO D1–21 in C1-24 Dex: 40 mg (20 if ⩾75 years) PO Qw in C1–9 and 20 mg Qw in C9–24 |
sCR rate MRD negative rate by NGS |
II | Recruiting |
| NCT03412565 | A study to evaluate subcutaneous daratumumab in combination with standard multiple myeloma treatment regimens | NDMM (for D-VRd & D-VMP) or R/R MM (DRd & DKd) | Arm 1: D-VRd Arm 2: D-VMP Arm 3: DRd Arm 4: DKd |
Dara: 1800 mg SC C1–4 in arm 1 and all cycles for arms 2–4 Bort: 1.3 mg/m2 SC C1–4 in arm 1 and C1–9 in arm 2 Len: 25 mg PO C1–4 in arm 1 and all cycles in arm 3 Dex: 20 mg PO/IV C1–4 in arm 1 and 40 mg PO/IV in all cycles in arms 3&4 Mel (arm 2 only): 9 mg/m2 PO C1–9 Pred (arm 2 only): 60 mg/m2 PO C1-9 Car (arm 4 only): 20 mg/m2 IV D1C1, then 70 mg/m2 IV D8,15 of C1, and D1,8,15 of C2+ |
⩾VGPR rate: For D-VRd Cohort ORR: For other cohorts |
II | Recruiting |
| NCT03290950 | A study of daratumumab in patients with newly diagnosed multiple myeloma | Untreated NDMM | 1 arm: D-KRd ×8 (2 cohorts) | 28-day cycles: Dara: 16 mg/kg D1,8,15,22 in C1–2, D1,15 in C3-6 and D1 in C7-8 Car: C1: 20 mg/m2 D2,3 and 36 mg/m2 D8,9,15,16. C2–8: 36 mg/m2 D1,2,8,9,15,16 Len: 25 mg PO QD D2–21 in C1 and D1-21 in C2-8 Dex (cohort 1): C1: 20 mg PO D2,3,8,9,15,16. C2: 20 mg D1,2,8,9,15,16,22. C3-4: 20 mg D1,2,8,9,15,16. C5-8: 10 mg PO D1,2,8,9,15,16 Dex (cohort 2): C1: 20 mg PO D1,2,22 and 40 mg D8,15. C2: 40 mg D1,8,15 and 20 mg D22. C3–4: 40 mg D1,8,15. C5–8: 20 mg PO D1,8,15 |
MRD negativity rate | II | Recruiting |
| NCT03224507 | Monoclonal antibody-based sequential therapy for deep remission in multiple myeloma (MASTER) | Transplant eligible and ineligible with untreated NDMM | Arm 1: D-KRd induction ×4, SCT, D-KRd consolidation (up to 2 blocks, 4 cycles each) Arm 2: D-KRd induction ×4 and D-KRd consolidation (up to 3 blocks, 4 cycles each) Maintenance (Both): MRD+: Len MRD-: observation |
Induction and consolidation: Arm 1: Dex: 40 mg PO D1,8,15,22 Len: 25 mg PO D1–21 Car: C1: 20 mg/m2 IV D8,9 and 36 mg/m2 IV D15,16. C2–4: 36 mg/m2 IV D1,2,8,9,15,16 Dara: 16 mg/kg IV D1,8,15,22 in C1–2 and D1,15 in C3–4 Arm 2: Dex: 40 mg PO D1,8,15,22 Len: 25 mg PO D1–21 Car: C1: 20 mg/m2 IV D8,9 and 36 mg/m2 IV D15,16. C2–4: 36 mg/m2 IV D1,2,8,9,15,16 Dara: 16 mg/kg IV D1,8,15,22 in C1-2 and D1,8,15 in C3-4 Maintenance: Len (MRD+ only) |
% of MRD negative remissions post consolidation | II | Recruiting |
| NCT03012880 | Ixazomib citrate, lenalidomide, dexamethasone, and daratumumab in treating patients with newly diagnosed multiple myeloma47 | Untreated NDMM | 1 arm: Ixa, Len, Dara, Dex induction ×12. Ixa and Dara maintenance (up to 36 months) | Induction: Ixa: 4 mg D1,8,15 Len: 25 mg D1–21 Dara: 16 mg/kg IV D1,8,15,22 in C1-2, D1,15 in C3-6 and D1 (Q4w) thereafter Dex: 40 mg D1,8,15,22 Maintenance: Ixa: D1,8,15 Dara: D1 Q4w |
confirmed CR rate | II | Active, not recruiting |
| NCT02951819 | A study to evaluate Dara-CyBorD in previously untreated and relapsed subjects with multiple myeloma (Lyra)48 | Untreated MM or Relapsed with 1 prior line of therapy | 1 arm: induction with Dara-CyBorD ×4–8, maintenance with Dara and Dex | Induction (28-day cycles): Cyclo: 300 mg/m2 PO D1,8,15,22 Bort: 1.5 mg/m2 SC D1,8,15 Dex: pre Dara infusion in all cycles. C1–8: pre + post-Dara infusion Dara: C1D1 and C1D2: 8 mg/kg. W2–8: 16 mg/kg IV Qw. W9–24: 16 mg/kg IV Q2w. W25+: Q4w Maintenance (12 28-day cycles): Dara: D1 Dex: D1 |
% with CR or VGPR | II | Active, not recruiting |
| NCT01998971 | A study of JNJ-54767414 (HuMax CD38) (anti-CD38 monoclonal antibody) in combination with backbone treatments for the treatment of patients with multiple myeloma | Newly diagnosed (for KRd) or R/R (for CFZ-dex) MM | Arm 1: DVd Arm 2: D-VMP Arm 3: D-VTd Arm 4: Dara-Pom-Dex Arm 5: DKd Arm 6: D-KRd |
Treatment details not available | Number affected by adverse events by MedDRA SOS and PT Number with DLT |
Ib | Active, not recruiting |
Bort, bortezomib; Car, carfilzomib; CR, complete response; CYBORD, cyclophosphamide, bortezomib and dexamethasone; D-KRd, daratumumab, carfilzomib, lenalidomide, dexamethasone; D-KRd, daratumumab, carfilzomib, lenalidomide, dexamethasone; D-VMP, daratumumab, bortezomib, melphalan, prednisone; D-VRd, daratumumab, bortezomib, lenalidomide, dexamethasone; D-VTd, daratumumab, bortezomib, thalidomide, dexamethasone; Dara, daratumumab; Dex, dexamethasone; DId, daratumumab, ixazomib, dexamethasone; DKd, daratumumab, carfilzomib, dexamethasone; DLT, dose-limiting toxicity; DRd, daratumumab, lenalidomide, dexamethasone; DVd, daratumumab, bortezomib, dexamethasone; IV, intravenous; Ixa, ixazomib; Len, lenalidomide; Mel; melphalan; MRD, minimal residual disease; NCT, National Clinical Trial; NGS, next generation sequencing; NDMM, newly diagnosed multiple myeloma; ORR, overall response rate; PO, by mouth; Pred, prednisone; Pom, pomalidomide; PT, preferred term; QD, daily; Qw, every week; SC, subcutaneous; sCR, stringent complete response; SCT, stem cell transplant; SOS, system organ class; VGPR, very good partial response.
Daratumumab dosing and formulations
The currently recommended dosing of intravenous (IV) daratumumab is 16 mg/kg administered on a weekly basis for 8 weeks, then biweekly for 16 weeks, and every 4 weeks thereafter, a schedule established based on efficacy and safety data from clinical trials.49 The pharmacokinetic properties of daratumumab were found to be unchanged when used in combination with other regimens, compared with monotherapy and regardless of the backbone regimen and of baseline patient or disease characteristics.50 Infusion reactions related to the IV formulation of daratumumab are very commonly encountered, especially with the first dose. They have been reported at a rate between 28% and 54% in major clinical trials, with the majority being grade 1–2.20,23,29,33,34,42,48 Thus, a subcutaneous (SC) formulation of daratumumab with recombinant human hyaluronidase enzyme (DARA + rHuPH20) has been developed in attempts to mitigate this reaction. Indeed, significantly decreased rates of infusion reactions have been reported with this formulation at a daratumumab dose of 1800 mg in a phase Ib safety study.51 Subsequently, the phase III COLUMBIA trial compared the DARA + rHuPH20 SC formulation (1800 mg) with the IV formulation (16 mg/kg) among 522 MM patients with at least 3 prior lines of therapy. The SC formulation was non-inferior to the IV formulation in terms of efficacy (ORR 41% versus 37% respectively) and pharmacokinetic properties, and exhibited a similar side effect profile. More importantly, it was associated with a significantly lower rate of infusion reactions (12.7% versus 34.5% p < 0.0001), at a lower administration time.52 As mentioned earlier, the phase III PERSUES trial is evaluating the efficacy of DVRd compared with VRd using SC daratumumab.44
Conclusion
Despite significant advances in MM treatment options, mortality remains high and more efficacious regimens are needed. Daratumumab has shown encouraging results both as monotherapy and in combination with other regimens in both R/R MM and untreated disease. Clinical trials so far have shown enhanced efficacy and tolerability of several daratumumab-based combinations in NDMM for both transplant ineligible and eligible patients, without hindering transplant. Although benefit in high-risk subpopulations is still unclear, data may be limited by the smaller number of participants in these subgroups, and thus more evidence is needed. Efforts are underway to explore other combinations with daratumumab, improve drug formulation, gain understanding of response predictors and mechanisms of resistance,53 and identify patient subgroups more likely to respond to these treatments.
Footnotes
Funding: The author(s) received no financial support for the research, authorship, and publication of this article.
Conflict of interest statement: S.K.K. served as a consultant for Celgene, Millennium Pharmaceuticals, Onyx Pharmaceuticals, Janssen, and Bristol-Myers Squibb and received research funding from Celgene, Millennium Pharmaceuticals, Novartis, Onyx Pharmaceuticals, AbbVie, Janssen, and Bristol-Myers Squibb. N.A. has no conflicts of interests to disclose.
ORCID iD: Shaji K. Kumar
https://orcid.org/0000-0001-5392-9284
Contributor Information
Nadine Abdallah, Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Shaji K. Kumar, Division of Hematology, Department of Internal Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA.
References
- 1. Palumbo A, Anderson K. Multiple myeloma. N Engl J Med 2011; 364: 1046–1060. [DOI] [PubMed] [Google Scholar]
- 2. Surveillance, Epidemiology, and End Results (SEER) Program. Cancer stat facts: myeloma, https://seer.cancer.gov/statfacts/html/mulmy.html (2019, accessed 12 September 2019).
- 3. Durie BG, Hoering A, Abidi MH, et al. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial. Lancet 2017; 389: 519–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Jakubowiak AJ, Dytfeld D, Griffith KA, et al. A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma. Blood 2012; 120: 1801–1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Moreau P, Hulin C, Macro M, et al. VTD is superior to VCD prior to intensive therapy in multiple myeloma: results of the prospective IFM2013-04 trial. Blood 2016; 127: 2569–2574. [DOI] [PubMed] [Google Scholar]
- 6. Mikhael J, Ismaila N, Cheung MC, et al. Treatment of multiple myeloma: ASCO and CCO joint clinical practice guideline. J Clin Oncol 2019; 37: 1228–1263. [DOI] [PubMed] [Google Scholar]
- 7. Deaglio S, Mehta K, Malavasi F. Human CD38: a (r)evolutionary story of enzymes and receptors. Leuk Res 2001; 25: 1–12. [DOI] [PubMed] [Google Scholar]
- 8. De Weers M, Tai YT, Van Der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol 2011; 186: 1840–1848. [DOI] [PubMed] [Google Scholar]
- 9. Van De Donk N, Richardson PG, Malavasi F. CD38 antibodies in multiple myeloma: back to the future. Blood 2018; 131: 13–29. [DOI] [PubMed] [Google Scholar]
- 10. Overdijk MB, Verploegen S, Bogels M, et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. MAbs 2015; 7: 311–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Overdijk MB, Jansen JH, Nederend M, et al. The therapeutic CD38 monoclonal antibody daratumumab induces programmed cell death via Fcγ receptor-mediated cross-linking. J Immunol 2016; 197: 807–813. [DOI] [PubMed] [Google Scholar]
- 12. Krejcik J, Casneuf T, Nijhof IS, et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood 2016; 128: 384–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Van Der Veer MS, De Weers M, Van Kessel B, et al. The therapeutic human CD38 antibody daratumumab improves the anti-myeloma effect of newly emerging multi-drug therapies. Blood Cancer J 2011; 1: e41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Nijhof IS, Groen RW, Noort WA, et al. Preclinical evidence for the therapeutic potential of CD38-targeted immuno-chemotherapy in multiple myeloma patients refractory to lenalidomide and bortezomib. Clin Cancer Res 2015; 21: 2802–2810. [DOI] [PubMed] [Google Scholar]
- 15. Görgün G, Calabrese E, Soydan E, et al. Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood 2010; 116: 3227–3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Bruns H, Jitschin R, Mougiakakos D, et al. Lenalidomide enhances MOR202 dependent macrophage-mediated effector functions via the vitamin D pathway. Blood 2015; 126: 2203. [DOI] [PubMed] [Google Scholar]
- 17. Feng X, Zhang L, Acharya C, et al. Targeting CD38 suppresses induction and function of T regulatory cells to mitigate immunosuppression in multiple myeloma. Clin Cancer Res 2017; 23: 4290–4300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Van Der Veer MS, De Weers M, Van Kessel B, et al. Towards effective immunotherapy of myeloma: enhanced elimination of myeloma cells by combination of lenalidomide with the human CD38 monoclonal antibody daratumumab. Haematologica 2011; 96: 284–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N Engl J Med 2015; 373: 1207–1219. [DOI] [PubMed] [Google Scholar]
- 20. Usmani SZ, Nahi H, Weiss BM, et al. Safety and efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed and refractory multiple myeloma: final results from GEN501 and sirius. Blood 2017; 130(Suppl. 1): 3107. [Google Scholar]
- 21. Lonial S, Weiss BM, Usmani SZ, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 2016; 387: 1551–1560. [DOI] [PubMed] [Google Scholar]
- 22. Palumbo A, Chanan-Khan A, Weisel K, et al. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med 2016; 375: 754–766. [DOI] [PubMed] [Google Scholar]
- 23. Dimopoulos MA, Oriol A, Nahi H, et al. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med 2016; 375: 1319–1331. [DOI] [PubMed] [Google Scholar]
- 24. Mateos MV, Sonneveld P, Hungria VTM, et al. Efficacy and safety of daratumumab, bortezomib, and dexamethasone (D-Vd) versus bortezomib and dexamethasone (Vd) in first relapse patients: two-year update of castor. Blood 2018; 132(Suppl. 1): 3270. [Google Scholar]
- 25. Plesner T, Arkenau HT, Gimsing P, et al. Phase 1/2 study of daratumumab, lenalidomide, and dexamethasone for relapsed multiple myeloma. Blood 2016; 128: 1821–1828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Plesner T, Arkenau HT, Gay F, et al. Enduring efficacy and tolerability of daratumumab in combination with lenalidomide and dexamethasone in patients with relapsed or relapsed/refractory multiple myeloma (GEN503): final results of an open-label, phase 1/2 study. Br J Haematol 2019; 186: e35–e39. [DOI] [PubMed] [Google Scholar]
- 27. Bahlis N, Dimopoulos MA, White DJ, et al. Three-year follow up of the phase 3 pollux study of daratumumab plus lenalidomide and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) alone in relapsed or refractory multiple myeloma (RRMM). Blood 2018; 132(Suppl. 1): 1996. [Google Scholar]
- 28. Bhatnagar V, Gormley NJ, Luo L, et al. FDA approval summary: daratumumab for treatment of multiple myeloma after one prior therapy. Oncologist 2017; 22: 1347–1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Chari A, Suvannasankha A, Fay JW, et al. Daratumumab plus pomalidomide and dexamethasone in relapsed and/or refractory multiple myeloma. Blood 2017; 130: 974–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Nooka AK, Joseph NS, Kaufman JL, et al. Clinical efficacy of daratumumab, pomalidomide, and dexamethasone in patients with relapsed or refractory myeloma: utility of re-treatment with daratumumab among refractory patients. Cancer 2019; 125: 2991–3000. [DOI] [PubMed] [Google Scholar]
- 31. Chari A, Martinez-Lopez J, Mateos MV, et al. Daratumumab plus carfilzomib and dexamethasone in patients with relapsed or refractory multiple myeloma. Blood 2019; 134: 421–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. DARZALEX® (daratumumab) injection [package insert on the internet]. Horsham, PA: Janssen Biotech, Inc., http://www.janssenlabels.com/package-insert/product-monograph/prescribing-information/DARZALEX-pi.pdf (2015, accessed 12 September). [Google Scholar]
- 33. Mateos MV, Dimopoulos MA, Cavo M, et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med 2018; 378: 518–528. [DOI] [PubMed] [Google Scholar]
- 34. Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med 2019; 380: 2104–2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Moreau P, Mateos MV, Bladé J, et al. An open-label, multicenter, phase 1b study of daratumumab in combination with backbone regimens in patients with multiple myeloma. Blood 2014; 124: 176.24859365 [Google Scholar]
- 36. Dimopoulos MA, Mateos MV, Cavo M, et al. One-year update of a phase 3 randomized study of daratumumab plus bortezomib, melphalan, and prednisone (D-VMP) versus bortezomib, melphalan, and prednisone (VMP) in patients (Pts) with transplant-ineligible newly diagnosed multiple myeloma (NDMM): alcyone. Blood 2018; 132(Suppl. 1): 156. [Google Scholar]
- 37. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med 2014; 371: 906–917. [DOI] [PubMed] [Google Scholar]
- 38. Dimopoulos MA, San-Miguel J, Belch A, et al. Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of POLLUX. Haematologica 2018; 103: 2088–2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Cao Y, Wan N, Liang Z, et al. Treatment outcomes in patients with newly diagnosed multiple myeloma who are ineligible for stem-cell transplantation: systematic review and network meta-analysis. Clin Lymphoma Myeloma Leuk 2019; 19: e478–e88. [DOI] [PubMed] [Google Scholar]
- 40. Stege CAM, Nasserinejad K, Levin MD, et al. Efficacy and tolerability of ixazomib, daratumumab and low dose dexamethasone (IDd) in unfit and frail newly diagnosed multiple myeloma (NDMM) patients; first interim safety analysis of the phase II HOVON 143 study. Blood 2018; 132(Suppl. 1): 596. [Google Scholar]
- 41. NIH U.S. National library of medicine, Clinicaltrials.gov. https://clinicaltrials.gov/ (2019, accessed 12 September 2019).
- 42. Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 2019; 394: 29–38. [DOI] [PubMed] [Google Scholar]
- 43. Voorhees PM, Rodriguez C, Reeves B, et al. Efficacy and updated safety analysis of a safety run-in cohort from griffin, a phase 2 randomized study of daratumumab (Dara), Bortezomib (V), lenalidomide (R), and dexamethasone (D; Dara-Vrd) Vs. Vrd in patients (Pts) with newly diagnosed (ND) multiple myeloma (MM) eligible for high-dose therapy (HDT) and autologous stem cell transplantation (ASCT). Blood 2018; 132(Suppl. 1): 151. [Google Scholar]
- 44. Sonneveld P, Broijl A, Gay F, et al. Bortezomib, lenalidomide, and dexamethasone (VRd) ± daratumumab (DARA) in patients (pts) with transplant-eligible (TE) newly diagnosed multiple myeloma (NDMM): a multicenter, randomized, phase III study (PERSEUS). J Clin Oncol 2019; 37(Suppl. 15): TPS8055. [Google Scholar]
- 45. Al Saleh AS, Sidiqi MH, Gertz MA, et al. Delayed neutrophil engraftment in patients receiving Daratumumab as part of their first induction regimen for multiple myeloma. Am J Hematol. Epub ahead of print 7 October 2019. DOI: 10.1002/ajh.25654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. O’dwyer M, Henderson R, Naicker SD, et al. CyBorD-DARA is potent initial induction for MM and enhances ADCP: initial results of the 16-BCNI-001/CTRIAL-IE 16-02 study. Blood Adv 2019; 3: 1815–1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Kumar SK, Kapoor P, Laplant B, et al. Phase 2 trial of ixazomib, lenalidomide, dexamethasone and daratumumab in patients with newly diagnosed multiple myeloma. Blood 2018; 132(Suppl. 1): 304. [Google Scholar]
- 48. Yimer H, Melear J, Faber E, et al. Lyra: a phase 2 study of daratumumab (Dara) plus cyclophosphamide, bortezomib, and dexamethasone (Cybord) in newly diagnosed and relapsed patients (Pts) with multiple myeloma (MM). Blood 2018; 132(Suppl. 1): 152. [Google Scholar]
- 49. Clemens PL, Yan X, Lokhorst HM, et al. Pharmacokinetics of daratumumab following intravenous infusion in relapsed or refractory multiple myeloma after prior proteasome inhibitor and immunomodulatory drug treatment. Clin Pharmacokinet 2017; 56: 915–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Xu XS, Dimopoulos MA, Sonneveld P, et al. Pharmacokinetics and exposure-response analyses of daratumumab in combination therapy regimens for patients with multiple myeloma. Adv Ther 2018; 35: 1859–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Chari A, Nahi H, Mateos MV, et al. Subcutaneous Delivery of daratumumab in patients (pts) with relapsed or refractory multiple myeloma (RRMM): pavo, an open-label, multicenter, dose escalation phase 1b study. Blood 2017; 130(Suppl. 1): 838. [Google Scholar]
- 52. Mateos MV, Nahi H, Legiec W, et al. Efficacy and safety of the randomized, open-label, non-inferiority, phase 3 study of subcutaneous (SC) versus intravenous (IV) daratumumab (DARA) administration in patients (pts) with relapsed or refractory multiple myeloma (RRMM): COLUMBA. J Clin Oncol 2019; 37(Suppl. 15): 8005. [Google Scholar]
- 53. Nijhof IS, Casneuf T, Van Velzen J, et al. CD38 expression and complement inhibitors affect response and resistance to daratumumab therapy in myeloma. Blood 2016; 128: 959–970. [DOI] [PubMed] [Google Scholar]
