You are on page 1of 7

Critical Reviews in Oncology / Hematology 159 (2021) 103211

Contents lists available at ScienceDirect

Critical Reviews in Oncology / Hematology


journal homepage: www.elsevier.com/locate/critrevonc

Daratumumab-based induction therapy for multiple myeloma: A systematic


review and meta-analysis
Lip Leong Chong a, 1, Yu Yang Soon b, 1, Cinnie Yentia Soekojo a, Melissa Ooi a, d,
Wee Joo Chng a, c, d, Sanjay de Mel a, *
a
Department of Haematology Oncology, National University Cancer Institute Singapore, National University Health System Singapore, Singapore
b
Department of Radiation Oncology, National University Cancer Institute Singapore, National University Health System Singapore, Singapore
c
Cancer Science Institute of Singapore, National University of Singapore, Singapore
d
Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore

A R T I C L E I N F O A B S T R A C T

Keywords: This study aims to evaluate the efficacy and safety of Daratumumab-based induction therapy (DBI) in newly
Daratumumab diagnosed multiple myeloma (MM). We identified four eligible RCTs including 2735 patients. The primary
Induction therapy outcomes of RCTs involving transplant eligible (TEMM) and non-transplant eligible MM (NTEMM) were stringent
Newly diagnosed multiple myeloma
complete response (sCR) and progression-free survival (PFS) respectively. Meta-analysis was performed using
High cytogenetic risk
Meta-analysis
random-effects models. DBI improved sCR rates for standard risk (SR) (OR 1.86, 95 % CI 1.41–2.46) but not HiR
(high risk) (OR 0.78, 95 % CI 0.41–1.48) (interaction P = 0.01) TEMM. In NTEMM, DBI improved PFS in SR (HR
0.44, 95 % CI 0.35–0.55) but not HiR patients. (HR 0.81, 95 % CI 0.52–1.27) (interaction P = 0.02). In
conclusion, while DBI is efficacious in SR patients, there is insufficient data to support a benefit in HiR-MM.

1. Introduction mechanisms (Plesner and Krejcik, 2018). Dara has demonstrated


remarkable efficacy both as a single agent and in combination with
Multiple myeloma (MM) is the second most common haematologic conventional therapies in relapsed or refractory MM (Lonial et al., 2016;
malignancy worldwide (Rajkumar, 2018). MM is characterized by a Dimopoulos et al., 2016; Bahlis et al., 2020; Palumbo et al., 2016). More
clonal proliferation of plasma cells usually associated with a monoclonal recently, Dara has been investigated in the upfront setting in combina­
protein and specific clinical features (Rajkumar, 2018). Proteasome in­ tion with standard induction regimens. In TEMM, Dara was combined
hibitor (PI) and immunomodulator (IMID) based triplet induction fol­ with bortezomib, thalidomide, dexamethasone (VTD) in the CASSIO­
lowed by high-dose melphalan and autologous stem-cell transplantation PEIA trial and with bortezomib, lenalidomide, dexamethasone (VRD) in
is the standard of care for transplant eligible MM (TEMM) (Rajkumar, the GRIFFIN trial (Moreau et al., 2019; Voorhees et al., 2020a). Both
2018; Laubach and Kumar, 2016). Depending on their fitness, phase III randomized controlled trials (RCT) showed a benefit for
non-transplant eligible MM (NTEMM) patients are treated with PI Dara-VTD (DVTD) and Dara-VRD (DVRD) over VTD and VRD respec­
and/or IMID based triplet or doublet regimens (Piechotta et al., 2019). tively in terms of stringent complete response (SCR) rate and
Despite the development of potent novel therapeutics, MM remains progression-free survival (PFS) (Moreau et al., 2019; Voorhees et al.,
incurable with the outlook for patients with high cytogenetic risk (HiR) 2020a).
MM being particularly poor (Rajkumar, 2018; Sonneveld et al., 2016). In NTEMM, Dara combined with bortezomib, melphalan, predniso­
There is hence a need to improve on currently available targeted lone (VMP) resulted in a superior PFS when compared to VMP (Mateos
therapy. et al., 2017; Mateos et al., 2020). The addition of Dara to lenalidomide
Daratumumab (Dara) is an anti-CD38 human IgGk monoclonal and dexamethasone (RD) also produced a significant PFS benefit
antibody which targets malignant plasma cells through a variety of compared to RD (Facon et al., 2019). These RCTs strongly suggest the

* Corresponding author at: Department of Haematology Oncology, National University Cancer Institute Singapore, National University Health System Singapore,
1E Kent Ridge Road, 119228, Singapore.
E-mail address: sanjay_widanalage@nuhs.edu.sg (S. de Mel).
1
These authors contributed equally.

https://doi.org/10.1016/j.critrevonc.2020.103211
Received 3 July 2020; Received in revised form 13 December 2020; Accepted 20 December 2020
Available online 30 December 2020
1040-8428/© 2020 Elsevier B.V. All rights reserved.
L.L. Chong et al. Critical Reviews in Oncology / Hematology 159 (2021) 103211

superiority of Dara-based induction over conventional regimens. The TEMM (CASSIOPEIA and GRIFFIN) and PFS for trials involving patients
benefit of Dara-based induction is however, less certain in patients with with NTEMM (ALCYONE and MAIA). Given the differing primary
HiR MM and these trials were not adequately powered to assess out­ outcome measures for the trials evaluating TEMM and NTEMM, we used
comes in this patient subset. We conducted a systematic review and sCR rate and PFS respectively when analysing the TEMM and NTEMM
meta-analysis to evaluate the efficacy and safety of Dara-based induc­ trials.
tion overall, and in specific subgroups of newly diagnosed MM (NDMM). The secondary outcomes for the CASSIOPEIA trial were OS, PFS,
response after induction (sCR, VGPR or better and overall response
2. Materials / subjects and methods rates), and response after consolidation (MRD negativity and CR). The
GRIFFIN trial considered OS, ORR and sCR rates at specific time points,
2.1. Research objective and study eligibility criteria time to response, duration of response as well as rates of PD, SD and
MRD-negativity after consolidation as its secondary outcomes.
This systematic review included RCTs comparing the effects of Dara- Meanwhile, the secondary outcomes for NTEMM studies from MAIA
based induction regimens versus standard treatment in patients with and ALCYONE were ORR (including rates of VGPR, CR and MRD
treatment naïve, TEMM and NTEMM. Inclusion criteria were as follows: negativity), time to response, duration of response, safety and side effect
patients with newly-diagnosed multiple myeloma (NDMM), comprising profiles.
either TEMM or NTEMM, who received standard induction regimens or
daratumumab-based induction treatment. The primary efficacy out­ 2.6. GRADE assessment
comes were stringent complete response (sCR) for TEMM and
progression-free survival (PFS) for NTEMM. Secondary efficacy out­ We assessed the overall certainty of the summarized evidence
comes were overall response rate (ORR) and the rates of very good focusing on the primary outcomes using the GRADE approach. The
partial response (VGPR), partial response (PR), complete response (CR), GRADE approach involves grading of five domains including study
negative minimal residual disease (MRD), stable disease (SD) and pro­ design, study’s overall risk of bias, inconsistency and imprecision of
gressive disease (PD). Safety and side effect profiles were additional studies’ results, and indirectness of the evidence. The GRADE system
secondary outcomes analysed in this study. classifies the certainty of the summarized evidence in one of the four
grades: High, Moderate, Low and Very Low. A high grade score suggests
2.2. Search strategy that further research is very unlikely to change our confidence in the
estimate of effect and very low grade score indicates that any estimate of
We identified eligible trials by searching MEDLINE, EMBASE and effect is very uncertain.
Cochrane Register of Controlled Trials from the date of inception on­
ward to April 2020. The search strategy included search terms such as 2.7. Subgroup analyses
“Multiple Myeloma” and “Daratumumab”. The results were then hand
searched for eligible trials. In addition, the reference list of selected trials Subgroup analyses were performed to determine if the estimates of
and conference proceedings from American Society of Clinical Oncology effect for the primary outcomes were influenced by cytogenetic profiles,
(ASCO), American Society of Hematology (ASH), European Hematology age, gender, race, international staging system (ISS) stage, heavy chain
Association (EHA) and International Myeloma Working Group (IMWG) subtype, ECOG performance status, baseline creatinine clearance and
between 2010 and 2019 were reviewed for any other eligible trials. hepatic function.

2.3. Selection of trials and data extraction 2.8. Statistical analyses

Three reviewers independently assessed the eligibility of the ab­ The log hazard ratios (HR) and their variances for time to event data
stracts identified by the search. The full text articles that appeared to (i.e. PFS and OS) were estimated using published methods when
meet the inclusion criteria was retrieved for closer review. Disagree­ appropriate summary statistics or Kaplan-Meier curves were reported.
ments were resolved by consensus. The individual trial log HR and their variances were combined using the
The same reviewers extracted the data independently using stan­ generic inverse variance method. A HR of less than 1 indicates an
dardized data collection forms. Data retrieved from the articles include advantage of using Dara-based treatment regimens.
publication details, methodological components, trial characteristics The restricted mean survival time (RMST) for both PFS and OS were
such as sample size, interventions, duration of follow up and outcome estimated at 6, 12, 24 and 36 months from randomisation. We recon­
measures. structed the individual patient data from the published Kaplan Meier
curves using methods developed by Wei and colleagues (Wei and Roy­
2.4. Risk of bias assessment ston, 2017). We estimated the RMST using the methods developed by
Cronin and colleagues (Cronin et al., 2016). We calculated the differ­
We assessed the risk of bias using the Cochrane RoB2 tool which ences in RMST and its standard deviation between the two treatment
assess the risk of bias in five domains, namely: randomization process, arms at the pre-specified time points. The individual trial differences in
deviations from the intended interventions, missing outcome data, RMST and its standard deviations were combined using the generic in­
measurement of the outcome and selection of the reported result. An verse variance method. A mean difference of more than zero indicates an
overall risk of bias was determined based on the reviewers’ judgement advantage of using Dara-based treatment.
for each of the domains. An overall “low risk of bias “score is given when The odd ratios for dichotomous data (i.e. disease response and
the study is judged to be at low risk of bias for all domains. An overall adverse events) were calculated from the number of patients who
“some concerns” score is given when the study is judged to raise some experienced the event and the number of patients who did not experi­
concerns in at least one domain, but not to be at high risk for any ence the event. The individual trial odds ratios were combined using the
domain. An overall “high risk of bias” score is given when the study is Mantel-Haenszel method. An odds ratio of more than 1 for disease
judged to be at high risk of bias in at least one domain. response outcomes and less than 1 for adverse events indicates an
advantage of using Dara-based treatment.
2.5. Outcome measures The chi-square Cochrane Q test was used to detect any heterogeneity
across the different trials and between subgroups. A P value of less than
The primary outcome was sCR for trials involving patients with 0.05 would indicate a statistically significant difference between the

2
L.L. Chong et al. Critical Reviews in Oncology / Hematology 159 (2021) 103211

subgroups i.e. a statistically significant heterogeneity among the trial studies was judged to be low (Supplementary Table 1).
results. The I2 statistics is used to judge the magnitude of heterogeneity.
An I2 statistic of more than 25 % would signify that at least moderate 3.3. Stringent Complete Response for trials on transplant eligible multiple
level of heterogeneity is present among the trial results. The random myeloma
effects meta-analysis model was used in the analysis.
Statistical analysis for reconstruction of individual patient data from Dara-based treatment was associated with a clinically and statisti­
the Kaplan Meier curves and restricted mean survival time were per­ cally significant improvement in sCR rate compared with standard
formed using Stata version 16.0 (Statacorp, TX). The meta-analysis was treatment (Odds ratio 1.59, 95 % CI 1.24–2.05, P value = 0.0003,
performed using the Cochrane Collaboration software (RevMan version Fig. 2). There was no statistically significant heterogeneity in the odd
5.30; http://www.cochrane.org) ratios for SCR from individual trials (chi square P value = 0.96, I2 = 0%).
The GRADE score was judged to be of high certainty (Supplementary
3. Results Table 2).

3.1. Results of search strategy 3.4. Subgroup analyses on Stringent Complete Response for trials on
transplant eligible multiple myeloma
We identified 267 records from our search strategy. After screening
through the titles and abstracts, we retrieved the full text articles of ten Patients with standard risk cytogenetic profiles treated with Dara-
records for further evaluation. We included a total of five articles on two based regimen demonstrated a statistically significant effect on sCR
RCTs that include TEMM and two RCTs including NTEMM (Fig. 1). compared to HiR MM. The pooled odds ratio was 1.86, 95 % CI
(1.41–2.46) for patients with the standard risk cytogenetic profile and
3.2. Characteristics of included studies was 0.78, 95 % CI (0.41–1.48) for patients with HiR MM. The test for
subgroup differences showed a P value of 0.01 (Fig. 3). The effects on
A total of 2735 patients were included from four RCTs analysed sCR were similar between subgroups defined by gender, ISS disease
(ALCYONE 2018, MAIA 2019, CASSIOPEIA 2019 and GRIFFIN 2020). stage, heavy chain isotype and ECOG performance status (Supplemen­
Patient demographics and baseline disease characteristics were gener­ tary Table 3).
ally well-balanced based on available data (Table 1). Dara-based in­
duction treatment was given for 50.1 % (n = 647) of TEMM and 49.8 % 3.5. Other efficacy outcomes for trials on transplant eligible multiple
(n = 718) of NTEMM, respectively. Overall, 388 patients (14 %) had myeloma
high-risk cytogenetics and were equally distributed between the Dara-
based induction and standard treatment groups. 1491 patients (55 %) Dara-based induction was associated with a clinically substantial and
were classified as IgG subtype and 681 patients (24 %) had ISS stage III statistically significant improvement in PFS compared to standard
disease. The average median follow-up time for all patients was 21.4 treatment (Hazard Ratio 0.47, 95 % CI 0.33–0.66, Fig. 4). The gain in
months (range 16.5–28.0 months). The risk of bias of the included RMST for PFS was observed at the 24-month time point (2.39 months, 95

Fig. 1. PRISMA flowchart for screening and identification of appropriate studies.

3
L.L. Chong et al. Critical Reviews in Oncology / Hematology 159 (2021) 103211

Table 1 1.74, 95 % CI 1.38–2.20), minimal residual disease (MRD) negative


Patient demographics and baseline disease characteristics among the patients regardless of response (odds ratio 2.83, 95 %CI 1.64–4.90) and MRD
included in the clinical trials (N = 2735). NDMM, newly diagnosed multiple negative with CR or better (odds ratio 2.66, 95 % CI 1.42–5.01)
myeloma. compared to standard treatment. There was no significant difference
Transplant Eligible NDMM Transplant Ineligible NDMM between Dara-based and standard treatment regimens for outcomes
Daratumumab- Standard Daratumumab- Standard including overall response, CR, VGPR or better, VGPR and PR. (Sup­
Characteristics
based Induction Induction based Induction Induction plementary Table 4).
(n = 647) (n = 645) (n = 718) (n = 725)

Age, years
Median, range 59 (22–70) 60 (26–70) 72 (40–93) 73 3.6. Adverse events for trials on transplant eligible multiple myeloma
(45–91)
Dara-based treatment was associated with increased odds of adverse
Gender events in terms of neutropenia (any grade) (odds ratio 2.17, 95 % CI
Male 374 (57.8 %) 379 (58.8 349 (48.6 %) 362 (49.9
%) %)
1.67–2.81), neutropenia (G3 or 4) (odds ratio 2.28, 95 % CI 1.74–3.00)
Female 273 (42.2 %) 266 (41.2 369 (51.4 %) 363 (50.1 and lymphopenia (any grade) (odds ratio 1.48, 95 % CI 1.10–1.98)
%) %) compared with standard treatment. There was no difference between the
two arms in terms of lymphopenia (G3 or 4), peripheral neuropathy (any
Performance score grades), peripheral neuropathy (G3 or 4), constipation (any grades),
Total 644 644 718 725
constipation (G3 or 4), nausea (any grades), nausea (G3 or 4), pyrexia
0 304 (47.0 %) 297 (46.0 205 (28.6 %) 222 (30.6
%) %) (any grades), pyrexia (G3 or 4), peripheral oedema (any grades) and
1 276 (42.7 %) 282 (43.7 360 (50.1 %) 360 (49.7 peripheral oedema (G3 or 4). These data are summarised in Supple­
%) %) mentary Table 5.
2 64 (9.9 %) 65 (10.0 153 (21.3 %) 143 (19.7
%) %)

3.7. Progression-free survival for trials on non-transplant eligible multiple


Type of measurable disease
IgG 386 (59.7 %) 366 (56.7 368 (51.3 %) 371 (51.2 myeloma
%) %)
Non-IgG 261 (40.3 %) 279 (43.3 350 (48.7 %) 354 (48.8 Dara-based treatment was associated with a clinically substantial
%) %) and statistically significant improvement in PFS compared with stan­
dard treatment (Hazard Ratio 0.48, 95 % CI 0.36 – 0.63, P value <
ISS disease stage
Total 646 643 718 725
0.0001) (Fig. 4). There was moderate level of heterogeneity among the
I 253 (39.1 %) 278 (43.1 167 (23.2 %) 170 (23.4 results with chi-square P value of 0.09 and I2 statistic of 65 %. The
%) %) GRADE score was judged to be high certainty (Supplementary Table 1).
II 295 (45.6 %) 270 (41.9 302 (42.1 %) 316 (43.6 The gain in RMST in PFS was observed at the 12-month (0.49 months, 95
%) %)
% CI 0.04–0.94 months), 24-month (2.30 month, 95 % CI 1.09–3.51
III 98 (15.1 %) 95 (14.7 249 (34.7 %) 239 (33.0
%) %) months) and 36-month (5.16 months, 95 % CI 2.39–7.93 months) time
points but not at the 6-month time point (Supplementary Fig. 2).
Cytogenetics classification
High risk 98 (15.1 %) 100 (15.5 101 (14.1 %) 89 (12.3
%) %) 3.8. Subgroup analyses on progression-free survival for trials on non-
Standard risk 542 (83.8 %) 537 (83.2 532 (74.1 %) 536 (73.9
transplant eligible multiple myeloma
%) %)
Not done 7 (1.1 %) 8 (1.2 %) 85 (11.8 %) 100 (13.8
%) The effect on PFS was statistically significantly greater in the patients
with standard risk cytogenetic profile compared with HiR MM. The
Median time since initial diagnosis of myeloma pooled hazard ratio was 0.44, 95 % CI (0.35–0.55) for patients with the
Month, range 0.81 (0.0–12.0) 0.91 0.88 (0.1–13.3) 0.85
standard risk cytogenetic profile and was 0.81, 95 % CI (0.52–1.27) for
(0.0–61.0) (0.0–25.3)
HiR MM patients. The test of subgroup differences showed a P value of
0.02 (Fig. 5). However, these studies were not powered to detect dif­
% CI 0.97–3.81 months), but not at the 6- and 12-month time points ferences in efficacy for HiR MM. The effects on PFS were similar between
(Supplementary Fig. 1). Dara-based treatment was also associated with subgroups defined by gender, age, race, baseline creatinine clearance,
improvement of other efficacy outcomes for CR or better (odds ratio hepatic function, ISS disease stage and heavy chain isotype (Supple­
mentary Table 6).

Fig. 2. Odds ratios (ORs) for stringent complete response (sCR) in transplant eligible newly diagnosed MM (NDMM). The ORs for each trial are represented by the
squares, with the size of each square corresponding to the size of the individual study. The confidence interval (CI) is a function of the overall sample size. The
diamonds represent the estimated overall effect, based on the meta-analysis fixed-effect method. All statistical tests were 2-sided.

4
L.L. Chong et al. Critical Reviews in Oncology / Hematology 159 (2021) 103211

Fig. 3. Odds ratios (ORs) for stringent complete response (sCR) in transplant eligible newly diagnosed (NDMM), by cytogenetic subgroup.

Fig. 4. Progression-free survival (PFS) analysis (intent-to-treat population) in both transplant eligible and ineligible newly diagnosed MM (NDMM). (a) Hazard ratios
(HRs) for PFS by individual study. (b) Pooled Kaplan-Meier estimates of PFS combining the data from CASSIOPEIA and GRIFFIN trials for TEMM and ALCYONE and
MAIA trials for NTEMM.

3.9. Other efficacy outcomes for trials on non-transplant eligible multiple Fig. 3). The gain in RMST for OS was observed at 36 months (1.04
myeloma months, 95 % CI 0.06–2.03 months) but not at the 6-, 12- and 24-month
time points (Supplementary Fig. 4). Dara-based treatment was associ­
Dara-based treatment was associated with a clinically substantial ated with improvement in other efficacy outcomes for overall response
and statistically significant improvement in OS compared with standard (odds ratio 3.28, 95 % CI 2.38–4.53). CR or better (odds ratio 2.51, 95 %
treatment (Hazard Ratio 0.67, 95 % CI 0.52–0.86, Supplementary CI 2.00–3.14), sCR (odds ratio 3.01, 95 % CI 2.23–4.06), VGPR or better

5
L.L. Chong et al. Critical Reviews in Oncology / Hematology 159 (2021) 103211

Fig. 5. Hazard ratios (HRs) for progression free survival (PFS) in transplant ineligible newly diagnosed myeloma (NDMM), by cytogenetic subgroup.

(odds ratio 2.90, 95 % CI 2.14–3.92). Patients treated with Dara-based results of these NMAs are largely consistent with our findings showing
regimens had lower odds of having stable disease (odds ratio 0.20, 95 that Dara-based regimens are beneficial for NDMM patients on the
% CI 0.13− 0.30) compared to standard treatment. There was no dif­ whole. The impact of Dara-based induction by cytogenetic subgroups
ference between the two arms in terms of VGPR rates or progressive was not however evaluated in any of these studies.
disease (Supplementary Table 4). It is noteworthy that the benefit of Dara-based induction in our study
did not differ when patients were sub grouped by gender, age, ISS stage
and heavy chain isotype. This highlights the importance of high risk
3.10. Adverse events for trials on transplant ineligible multiple myeloma cytogenetics as a predictor of treatment response in MM. It is note­
worthy that rates of MRD negativity were lower in HiR patients. Given
Dara-based treatment was associated with increased odds of adverse the correlation of MRD negativity with long term outcomes in MM, this
events in terms of infections (any grade) (odds ratio 2.21, 95 % CI is likely to be a key factor explaining the discrepancy in outcomes (Paiva
1.74–2.81), infections (G3 or 4) (odds ratio 1.64, 95 % CI 1.27–2.10), et al., 2020).
pneumonia (any grade) (odds ratio 2.59, 95 % CI 1.48–4.53) and Our study has several limitations. Firstly, we did not have individual
pneumonia (G3 or 4) (odds ratio 2.29, 95 % CI 1.39–3.77). There was no patient data of each trial, thus we were unable to identify which patients
difference in terms of neutropenia (any grade), neutropenia (G3 or 4), with HiR MM will benefit from Dara-based induction or determine the
anaemia (any grade), diarrhoea (any grade), diarrhoea (G3 or 4), nausea impact of cytogenetic profiles on the secondary efficacy outcomes.
(any grade), nausea (G3 or 4) and G5 toxicities. These data are sum­ Secondly, as these RCTs excluded patients with ECOG performance
marised in Supplementary Table 7. status greater than 2 and multiple co-morbidities, these results should
not be extrapolated to this frail group of patients who are commonly
4. Discussion seen in real world practice. The relatively short median follow up time is
also an important limitation of our analysis and longer term results of
Our meta-analysis demonstrates that Dara-based induction is asso­ these trials would be of significant importance.
ciated with significant improvement in the primary outcomes of sCR rate It is also noteworthy that the maintenance randomisations of the
for TEMM and PFS for NTEMM compared with standard regimens. CASSIOPEIA and GRIFFIN studies were different (Moreau et al., 2019;
Although Dara-based induction led to marked improvements for pa­ Voorhees et al., 2020a). Specifically, CASSIOPEIA randomised patients
tients with standard risk cytogenetics, there was no clear benefit in terms between Dara maintenance and observation, while GRIFFIN compared
of sCR or PFS for patients with HiR MM. The key strengths of our study Dara-lenalidomide maintenance with single agent lenalidomide main­
include the use of the most up to date published data as well as validated tenance. The different maintenance randomisation means the mainte­
tools to evaluate the quality of the individual studies and summarized nance arms of these trials and the magnitude of PFS benefit may not be
evidence on this topic. Importantly, our meta-analysis has more statis­ directly comparable. The primary end point of both these studies was
tical power than individual trials for examining the impact of cytoge­ however SCR rate which was assessed before maintenance and therefore
netic profiles as an effect modifier for analysis of the primary outcomes. remains a valid point of comparison.
In a systematic review and network meta-analysis (NMA) comparing In conclusion, we propose that although Dara-based induction is
a variety of induction regimens to RD as a reference, D-RD and D-VMP clearly beneficial in standard risk patients, there is currently insufficient
emerged as the most potent combinations for NTEMM (Cao et al., 2019). data to demonstrate a benefit in HiR MM. Given that some HiR patients
Sekine and colleagues also demonstrated in a NMA that D-VMP is su­ do benefit from Dara-based induction, clinical trials focusing on this
perior to standard PI and IMID based triplet induction regimens in terms subgroup are required to determine which HiR patients may benefit
of PFS for NTEMM (Sekine et al., 2019). A NMA comparing induction from Dara-based induction and the basis for this response. A better
regimens reported in six clinical trials of NTEMM similarly demon­ understanding of the biology of HiR MM is required to design more
strated that Dara-based regimens were superior to standard treatment potent targeted therapeutics to address this unmet clinical need.
(Xu et al., 2019). Interestingly, this analysis suggested that DRD may be
superior to DVMP. Gil-Sierra and colleagues performed a similar NMA Funding sources
on NTEMM which also demonstrated a benefit for Dara-based induction
compared to conventional regimens (Gil-Sierra et al., 2020). This study This research did not receive any specific grant from funding
suggested that VRD may be equivalent to DRD in terms of efficacy. Given agencies in the public, commercial, or not-for-profit sectors.
that the GRIFFIN trial showed superiority of D-VRD over VRD, this
suggests that the incorporation of a PI contributes significantly to the
efficacy of induction therapy in NTEMM (Voorhees et al., 2020b). The

6
L.L. Chong et al. Critical Reviews in Oncology / Hematology 159 (2021) 103211

Ethical approval and consent to participate Lonial, S., et al., 2016. Daratumumab monotherapy in patients with treatment-refractory
multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 387
(10027), 1551–1560.
None required. Mateos, M.-V., et al., 2017. Daratumumab plus bortezomib, melphalan, and prednisone
for untreated myeloma. N. Engl. J. Med. 378 (6), 518–528.
Consent to publication Mateos, M.V., et al., 2020. Overall survival with daratumumab, bortezomib, melphalan,
and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised,
open-label, phase 3 trial. Lancet 395 (10218), 132–141.
All authors have provided consent for publication. Moreau, P., et al., 2019. 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
CRediT authorship contribution statement study. Lancet 394 (10192), 29–38.
Paiva, B., et al., 2020. Measurable residual disease by next-generation flow cytometry in
Lip Leong Chong: Conceptualization, Data curation, Formal anal­ multiple myeloma. J. Clin. Oncol. 38 (8), 784–792.
Palumbo, A., et al., 2016. Daratumumab, bortezomib, and dexamethasone for multiple
ysis, Methodology, Writing - original draft, Writing - review & editing. myeloma. N. Engl. J. Med. 375 (8), 754–766.
Yu Yang Soon: Conceptualization, Data curation, Formal analysis, Piechotta, V., et al., 2019. Multiple drug combinations of bortezomib, lenalidomide, and
Methodology, Writing - original draft, Writing - review & editing. Cin­ thalidomide for first-line treatment in adults with transplant-ineligible multiple
myeloma: a network meta-analysis. Cochrane Database Syst. Rev. (11).
nie Yentia Soekojo: Writing - review & editing. Melissa Ooi: Writing - Plesner, T., Krejcik, J., 2018. Daratumumab for the treatment of multiple myeloma.
review & editing, Supervision. Wee Joo Chng: Writing - review & Front. Immunol. 9, p. 1228–1228.
editing, Supervision. Sanjay de Mel: Conceptualization, Data curation, Rajkumar, S.V., 2018. Multiple myeloma: 2018 update on diagnosis, risk-stratification,
and management. Am. J. Hematol. 93 (8), 1091–1110.
Formal analysis, Methodology, Writing - original draft, Writing - review
Sekine, L., et al., 2019. Upfront treatment for newly diagnosed transplant-ineligible
& editing, Supervision. multiple myeloma patients: a systematic review and network meta-analysis of
14,533 patients over 29 randomized clinical trials. Crit. Rev. Oncol. Hematol. 143,
Declaration of Competing Interest 102–116.
Sonneveld, P., et al., 2016. Treatment of multiple myeloma with high-risk cytogenetics: a
consensus of the International Myeloma Working Group. Blood 127 (24),
The authors report no declarations of interest. 2955–2962.
Voorhees, P.M., et al., 2020a. Daratumumab, lenalidomide, bortezomib, &
dexamethasone for transplant-eligible newly diagnosed multiple myeloma: GRIFFIN.
Acknowledgements Blood.
Voorhees, P.M., et al., 2020b. Daratumumab, lenalidomide, bortezomib, &
None. dexamethasone for transplant-eligible newly diagnosed multiple myeloma: GRIFFIN.
Blood.
Wei, Y., Royston, P., 2017. Reconstructing time-to-event data from published Kaplan-
Appendix A. Supplementary data Meier curves. Stata J. 17 (4), 786–802.
Xu, W., et al., 2019. Daratumumab added to standard of care in patients with newly
diagnosed multiple myeloma: a network meta-analysis. Eur. J. Haematol. 103 (6),
Supplementary material related to this article can be found, in the 542–551.
online version, at doi:https://doi.org/10.1016/j.critrevonc.2020.10
3211. Lip Leong Chong: Senior resident in haematology at the national university cancer
institute Singapore.
References
Yu Yang Soon: Consultant radiation oncologist at the national university cancer institute
Bahlis, N.J., et al., 2020. Daratumumab plus lenalidomide and dexamethasone in Singapore.
relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a
randomized, open-label, phase 3 study. Leukemia. Cinnie Yentia Soekojo: Associate consultant haematologist at the national university
Cao, Y., et al., 2019. Treatment outcomes in patients with newly diagnosed multiple cancer institute Singapore.
myeloma who are ineligible for stem-cell transplantation: systematic review and
network meta-analysis. Clin. Lymphoma Myeloma Leuk. 19 (8), e478–e488.
Cronin, A., Tian, L., Uno, H., 2016. strmst2 and strmst2pw: new commands to compare Melissa Ooi: Consultant haematologist at the national university cancer institute
survival curves using the restricted mean survival time. Stata J. 16 (3), 702–716. Singapore.
Dimopoulos, M.A., et al., 2016. Daratumumab, lenalidomide, and dexamethasone for
multiple myeloma. N. Engl. J. Med. 375 (14), 1319–1331. Wee Joo Chng: Senior Consultant haematologist at the national university cancer institute
Facon, T., et al., 2019. Daratumumab plus lenalidomide and dexamethasone for Singapore.
untreated myeloma. N. Engl. J. Med. 380 (22), 2104–2115.
Gil-Sierra, M.D., et al., 2020. Network meta-analysis of first-line treatments in transplant-
ineligible multiple myeloma patients. Eur. J. Haematol. Sanjay de Mel: Consultant haematologist at the national university cancer institute
Laubach, J., Kumar, S., 2016. Management of transplant-eligible patients with newly Singapore
diagnosed multiple myeloma. Cancer Treat. Res. 169, 145–167.

You might also like