Quadruplets in newly diagnosed transplant-ineligible multiple myeloma

IF 10.1 1区 医学 Q1 HEMATOLOGY American Journal of Hematology Pub Date : 2024-09-10 DOI:10.1002/ajh.27473
Rajshekhar Chakraborty, Hira Mian
{"title":"Quadruplets in newly diagnosed transplant-ineligible multiple myeloma","authors":"Rajshekhar Chakraborty, Hira Mian","doi":"10.1002/ajh.27473","DOIUrl":null,"url":null,"abstract":"<p>To the Editor:</p>\n<p>With the advent of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies (CD38mAbs), the outcomes of transplant-ineligible (TIE) patients with newly diagnosed multiple myeloma (MM) have substantially improved in the last decade.<span><sup>1-3</sup></span> Currently, there are two widely accepted standard-of-care regimens in newly diagnosed TIE MM: daratumumab-lenalidomide-dexamethasone (DRd) and bortezomib-lenalidomide-dexamethasone (VRd), based on the MAIA and S0777 trials, respectively, both of which showed significant progression-free survival (PFS) and overall survival (OS) benefit over Rd.<span><sup>1, 2, 4, 5</sup></span> The major toxicity of concern with the addition of CD38mAb is infection and with bortezomib is peripheral neuropathy. While these triplet regimens have been considered the standard of care, two RCTs have been recently reported that compared a quadruplet regimen to these triplet regimens.<span><sup>6, 7</sup></span> The IMROZ phase III trial evaluated the CD38mAb isatuximab on the backbone of bortezomib, lenalidomide, and dexamethasone (Isa-VRd) compared with VRd alone. The IFM-2020/BENEFIT (hereafter, referred to as BENEFIT) trials evaluated Isa-VRd compared with IsaRd alone. Additionally, a third RCT named GEM2017FIT compared the quadruplet regimen daratumumab-carfilzomib-lenalidomide-dexamethasone (Dara-KRd) against KRd and alternating VMP/Rd (VMP: bortezomib-melphalan-prednisone) in this patient population.<span><sup>8</sup></span> The study design and key outcomes of these trials are summarized in Table 1. Here, we will discuss three key aspects of these trials (inclusion criteria, toxicity, and efficacy) and future directions for optimizing therapy for this patient population.</p>\n<div>\n<header><span>TABLE 1. </span>Key randomized controlled trials comparing quadruplets versus triplets in transplant-ineligible multiple myeloma.</header>\n<div tabindex=\"0\">\n<table>\n<thead>\n<tr>\n<td></td>\n<th>IFM-2020/BENEFIT (<i>n</i> = 270)</th>\n<th>IMROZ (<i>n</i> = 446)</th>\n<th>GEM2017FIT (<i>n</i> = 462)</th>\n</tr>\n</thead>\n<tbody>\n<tr>\n<td colspan=\"4\">Inclusion criteria</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Age</td>\n<td>65–79 years AND non-frail (<i>IMWG Frailty Score &lt;2</i>)</td>\n<td>≥18–80 years AND TIE due to ≥65 years or comorbidities</td>\n<td>TIE with age &lt;80 years</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">ECOG or Frailty cut-off</td>\n<td>ECOG &gt;2 or Frail excluded</td>\n<td>ECOG &gt;2 excluded</td>\n<td>Geriatric Assessment Hematology score &gt;42</td>\n</tr>\n<tr>\n<td colspan=\"4\">Trial treatment</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Regimen</td>\n<td>Isa-VRd vs. IsaRd</td>\n<td>Isa-VRd vs. VRd</td>\n<td>VMP-Rd vs. KRd vs. Dara-KRd</td>\n</tr>\n<tr>\n<td colspan=\"4\">Results</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Age of enrolled patients (median, range unless otherwise specified)</td>\n<td>73 (IQR 71–76)</td>\n<td>72 (55–80)</td>\n<td>72 (65–80)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Age ≥ 75 years</td>\n<td>31% (IsaVRd) vs. 31% (IsaRd)</td>\n<td>75–80: 26% (IsaVRd) vs. 32% (VRd)</td>\n<td>~ one-third older than 75 years</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">% High-risk cytogenetics</td>\n<td>10% (IsaVRd) vs. 8% (IsaRd)<sup>b</sup>\n</td>\n<td>15% (Isa-VRd) vs. 19% (VRd)<sup>a</sup>\n</td>\n<td>~15%</td>\n</tr>\n<tr>\n<td colspan=\"4\">Primary and secondary endpoints</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">MRD negativity, 10<sup>−5</sup></td>\n<td>18 mo: 53% (IsaVRd) vs. 26% (IsaRd)</td>\n<td>58% (IsaVRd) vs. 44% (VRd)</td>\n<td>33% (VMP-Rd) vs. 75% (KRd) vs. 84% (Dara-KRd)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">MRD negativity, 10<sup>−6</sup></td>\n<td>18 mo.: 36% (IsaVRd) vs. 17% (IsaRd)</td>\n<td></td>\n<td>25% (VMP-Rd) vs. 66% (KRd) vs. 79% (Dara-KRd)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">≥VGPR</td>\n<td>82% (IsaVRd) vs. 70% (IsaRd)</td>\n<td>89% (Isa-VRD) vs. 83% (VRd)</td>\n<td>N/A</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Median follow-up</td>\n<td>23.5 months</td>\n<td>~60 months</td>\n<td>NA</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">PFS</td>\n<td>2-years PFS: 80% (IsaRd) vs. 85.2% (IsaRVd)</td>\n<td>5-years PFS: 63% (Isa-VRD) vs. 45% (VRd)</td>\n<td><b>2.5 year PFS</b>: 73% (VMP-Rd) vs. 83% (KRd) vs. 79% (Dara-KRd)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">OS</td>\n<td>2-years OS: 91.5% (IsaRd) vs. 91.1% (IsaVRd)</td>\n<td>5-years OS: 72% (IsaVRd) vs. 66% (VRd)</td>\n<td><b>18-month OS</b>: 91% (VMP-Rd) vs. 95% (KRD) vs. 90% (Dara-KRD)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Post-protocol therapy</td>\n<td>Not available</td>\n<td>70% of patients in VRd arm that progressed received anti-CD38mAb</td>\n<td>N/A</td>\n</tr>\n<tr>\n<td colspan=\"4\">Major toxicities</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Neutropenia (Grade ≥3)</td>\n<td>40% (IsaVRd) vs. 45% (IsaRd)</td>\n<td>54% (Isa-VRd) vs. 37% (VRd)</td>\n<td>50% (VMP-Rd) vs. KRd (24%) vs. Dara-KRd (47%)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Infections (Grade ≥3)</td>\n<td>36% (Isa-VRd) vs. 28% (IsaRd)</td>\n<td><b>45</b>% (Isa-VRd) vs. 38% (VRd)</td>\n<td>12% (VMP-Rd) vs. (15% (KRd) vs. 15% (Dara-KRd)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Peripheral neuropathy (all grade)</td>\n<td>52% (Isa-VRd) vs. 28% (Isa-Rd) vs</td>\n<td>~55%</td>\n<td>N/A</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Cardiac disorders (Grade ≥3)</td>\n<td>8% (Isa-VRd) vs. 0% (Isa-Rd) vs</td>\n<td>N/A</td>\n<td>5% (VMP-Rd) vs. 11% (KRd) vs. 14% (Dara-KRd)</td>\n</tr>\n<tr>\n<td style=\"padding-left:2em;\">Treatment discontinuation (first 18 months due to deaths)</td>\n<td>3.7% (Isa-VRd) vs. 2.2% (Isa-Rd)</td>\n<td>Overall treatment-related deaths: 11% (IsaVRd) vs. 5.5% (VRd)</td>\n<td>Toxicity-related death: 4.5% (VMP-Rd) vs. 3.2% (KRd) vs. 8.5% (Dara-KRd)</td>\n</tr>\n</tbody>\n</table>\n</div>\n<div>\n<ul>\n<li title=\"Footnote 1\"><span><sup>a</sup> </span> del(17p) or HR translocations t(4;14), t (14;16). </li>\n<li title=\"Footnote 2\"><span><sup>b</sup> </span> Perrot et al, JCO 2019 criteria. </li>\n<li> Abbreviations: AE, adverse event; Dara-KRD, daratumumab, carfilzomib, lenalidomide, and dexamethasone; ECOG, Eastern Cooperative Oncology Group; IMWG, International Myeloma Working Group; IsaRd, isatuximab, lenalidomide, and dexamethasone; IsaRVd, isatuximab, lenalidomide, bortezomib, and dexamethasone; KRD, carfilzomib, lenalidomide, and dexamethasone; MRD, minimal residual disease; NA, not available; OS, overall survival; PFS, progression-free survival; RVd, lenalidomide, bortezomib, and dexamethasone; TIE, transplant-ineligible; VGPR, very good partial response; VMP-Rd, bortezomib, melphalan, prednisone, lenalidomide, and dexamethasone. </li>\n</ul>\n</div>\n<div></div>\n</div>\n<p>The first key aspect relates to eligibility criteria and the included patient population in these trials. Even though these RCTs were designed for a TIE population, they all had an upper age limit of 80 years, and approximately two-thirds of patients were within the age brackets of 65–75.<span><sup>6-8</sup></span> Additionally, the BENEFIT and GEM2017FIT trials specifically excluded frail patients as defined by the IMWG or Geriatric Assessment in Hematology (GAH) frailty scores, respectively. Objective assessment of frailty is critical in older adults, since frailty predicts treatment-related toxicities and OS.<span><sup>9</sup></span> Hence, the external validity of safety and efficacy of quadruplet regimens in patients who are frail or &gt;80 years of age, which constitutes at least ~20% of TIE population,<span><sup>9</sup></span> remain unclear. An ongoing RCT comparing Dara-VRd vs. VRd (CEPHEUS) in TIE patients does not have an upper age limit or exclusion based on frailty and will potentially generate data on the benefit of quadruplets for older patients (NCT03652064). However, it remains to be seen as to what proportion of patients in CEPHEUS trial are frail based on objective criteria. Future RCTs in this population should target specific subgroups such as fit, intermediate fit, and frail rather than arbitrarily defined category based on transplant-eligibility, which may include both patients that are truly not fit for transplant and those in whom either a transplant is deferred or not done due to patient/physician preference.</p>\n<p>Second, early mortality and cumulative treatment-related toxicity is an important concern in older adults when adding drugs to current backbones, with real-world data showing substantial early mortality even in the modern era.<span><sup>10</sup></span> Reassuringly, both IMROZ and BENEFIT trials demonstrated that there was no increase in the risk of early mortality with the addition of 4th drug (isatuximab and bortezomib, respectively).<span><sup>6, 7</sup></span> However, in IMROZ, the cumulative incidence of treatment-related deaths at a median follow-up of ~5 years was approximately 2-fold higher in Isa-VRd versus VRd arm (11% vs. 5.5% respectively), with the most common cause of these deaths being infection.<span><sup>6</sup></span> The increase in these treatment-related deaths in the Isa-VRd arm was largely driven by events beyond 6 months from treatment initiation. This highlights the deleterious impact of prolonged immunosuppression with continuous CD38mAb and dexamethasone until progression. Furthermore, it reiterates the urgent need for de-escalation strategies in frail older adults who particularly value minimizing treatment toxicity and optimizing health-related quality of life (HRQoL).<span><sup>11</sup></span> De-escalation strategies are currently being explored in two RCTs that are randomizing patients to continuous versus fixed-duration daratumumab in the DRd regimen (NCT05561387 and NCT06182774). In the BENEFIT trial, addition of once-weekly subcutaneous bortezomib increased the risk of Grade ≥2 peripheral neuropathy from 10% in the Isa-Rd arm to 27% in Isa-VRd arm.<span><sup>7</sup></span> However, the incidence of all-grade peripheral neuropathy with Isa-VRd in BENEFIT trial was almost half of that in IMROZ (28% vs. 54.4%, respectively), which is likely due to once-weekly bortezomib in BENEFIT versus once-weekly in IMROZ.<span><sup>6, 7</sup></span> Since bortezomib-induced neuropathy can be reversible in about two-thirds of patients with dose-reduction or dose-discontinuation,<span><sup>12</sup></span> it remains to be seen what proportion of these patients develop permanent neuropathy. Nevertheless, close monitoring for new-onset peripheral neuropathy while on bortezomib with prompt dose modification remains paramount if using a bortezomib-containing quadruplet regimen in this age group. Notably, in the GEM2017FIT trial, a substantial increase in toxicity-related deaths in the first 18 months was noted in the quadruplet arm (toxic deaths-4.5%, 3.2%, and 8.5% in VMP/Rd, KRd, and Dara-KRd arms, respectively).<span><sup>8</sup></span> Furthermore, a substantial proportion of patients in the carfilzomib-containing arms (11% in KRd and 14% in Dara-KRd) developed Grade 3–4 cardiac toxicities. This is in line with the GMMG-CONCEPT trial (Isa-KRd) where the incidence of Grade 3–5 cardiac toxicities was ~10× higher in the TIE cohort compared with the TE cohort (20% vs. 2.1%, respectively).<span><sup>13</sup></span> Together, these data imply caution with CD38mAb and carfilzomib-containing quadruplet regimens in older adults, including those who are fit and &lt;80 years old.</p>\n<p>Third, all three RCTs met their primary efficacy endpoint (PFS in IMROZ and MRD negativity at 10<sup>−5</sup> by NGS in BENEFIT and GEM2017FIT). However, IMROZ trial has the most mature data thus far with a median follow-up of ~5 years.<span><sup>6</sup></span> Importantly, in the GEM2017FIT trial, the efficacy of Dara-KRd was modulated by frailty status, with 30-month PFS in fitter patients (GAH &lt;20) being 88% and that in patients with GAH &gt;20 being just 71% with Dara-KRd.<span><sup>8</sup></span> Notably, the BENEFIT trial demonstrated a ~2× increase in MRD negativity at 10<sup>−6</sup> with the addition of bortezomib to Isa-Rd. An important caveat is that the MRD-negativity rates at 12 and 18 months were comparable in the Isa-VRd arm of BENEFIT trial, which raises the question whether bortezomib can be limited to 12 months to reduce toxicity without compromising efficacy. Additionally, the time to achievement of very good partial response or better was significantly shorter with Isa-VRd compared with Isa-Rd (2.1 vs. 3.7 mo., respectively; <i>p</i> = 0.0002), which highlights the importance of addition of bortezomib in situations where a rapid reduction in paraprotein is desirable such as myeloma cast nephropathy.<span><sup>14</sup></span> Of note, the MRD-negativity rate at 10<sup>−5</sup> (NGS) in the intention-to-treat population was comparable in the Isa-VRd arms of IMROZ and BENEFIT trials (58% vs. 53%, respectively) despite twice-weekly bortezomib in IMROZ and once-weekly in BENEFIT.<span><sup>6, 7</sup></span> Thus far, no difference in PFS or OS has been noted with the addition of bortezomib to Isa-Rd. Unlike IMROZ trial where addition of CD38mAb led to early separation of PFS curves, addition of bortezomib has not led to the separation of PFS curves yet at a median follow-up of ~2 years. In a recent meta-analysis, MRD negativity was shown to have a strong trial-level correlation with PFS (<i>R</i><sup>2</sup> = 0.85) and medium trial-level correlation with OS (<i>R</i><sup>2</sup> = 0.79) in the TIE cohort,<span><sup>15</sup></span> which makes it likely that the benefit in MRD negativity will translate into benefit in PFS and possibly OS with longer follow-up. Nevertheless, due to the trade-off of increased peripheral neuropathy, which has important HRQoL implications in frail older adults, mature data are needed to understand the magnitude of PFS/OS benefit to enable shared decision-making in this population. Of note, IMROZ was the only RCT that had HRQoL as an endpoint, and failed to show superiority of Isa-VRd over VRd in the Global Health Status/Quality of Life domain of EORTC-QLQ-C30 instrument.<span><sup>6</sup></span> In a prospective cohort study in adults with MM over the age of 50, compared with younger adults (50–69 years), older adults (≥70 years) were significantly more likely to prioritize QoL, functional independence, maintaining cognitive ability, and living free from pain compared with longer OS.<span><sup>11</sup></span> Hence, clinical trials in this population should rigorously measure HRQoL, and clinicians should be cognizant of unique patient preferences in this age group during treatment decision-making.</p>\n<p>In summary, the outcomes of patients with newly diagnosed TIE MM are poised to improve in coming years with the introduction of effective drugs in the frontline setting; however, it remains unclear how we should translate the results of these trials in our routine clinical setting. For healthcare systems that are using VRd as the current standard of care in this population, we believe IMROZ trial provides convincing evidence to add CD38mAb to VRd backbone and should lead to a change in the standard of care. This is particularly important in fit patients and those under the age of 80. Further trials are eagerly awaited investigating the optimal duration of CD38mAb and whether routine quadruplet therapy is feasible and beneficial in frail adults including over the age of 80. However, in settings where CD38mAb + Rd is being currently used as a standard of care, it remains unclear whether addition of bortezomib is essential for all patients since enthusiasm from the benefit in terms of MRD-negativity rate is tempered by the rise in clinically significant peripheral neuropathy, and long-term follow-up will be crucial to understand the magnitude of PFS and potentially OS benefit. We believe that future trials should move away from transplant “ineligibility” as an inclusion criterion given the heterogeneity of this large category and rather focus on frailty-specified subgroups—fit, intermediate fit, and frail. Additionally, incorporating frailty-adapted attenuated dosing strategies and more dynamic fitness measures may allow us to both decrease potential toxicity and improve efficacy, allowing for more personalized treatment strategies in routine clinical practice for this heterogenous group of TIE patients.</p>","PeriodicalId":7724,"journal":{"name":"American Journal of Hematology","volume":null,"pages":null},"PeriodicalIF":10.1000,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"American Journal of Hematology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1002/ajh.27473","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

To the Editor:

With the advent of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies (CD38mAbs), the outcomes of transplant-ineligible (TIE) patients with newly diagnosed multiple myeloma (MM) have substantially improved in the last decade.1-3 Currently, there are two widely accepted standard-of-care regimens in newly diagnosed TIE MM: daratumumab-lenalidomide-dexamethasone (DRd) and bortezomib-lenalidomide-dexamethasone (VRd), based on the MAIA and S0777 trials, respectively, both of which showed significant progression-free survival (PFS) and overall survival (OS) benefit over Rd.1, 2, 4, 5 The major toxicity of concern with the addition of CD38mAb is infection and with bortezomib is peripheral neuropathy. While these triplet regimens have been considered the standard of care, two RCTs have been recently reported that compared a quadruplet regimen to these triplet regimens.6, 7 The IMROZ phase III trial evaluated the CD38mAb isatuximab on the backbone of bortezomib, lenalidomide, and dexamethasone (Isa-VRd) compared with VRd alone. The IFM-2020/BENEFIT (hereafter, referred to as BENEFIT) trials evaluated Isa-VRd compared with IsaRd alone. Additionally, a third RCT named GEM2017FIT compared the quadruplet regimen daratumumab-carfilzomib-lenalidomide-dexamethasone (Dara-KRd) against KRd and alternating VMP/Rd (VMP: bortezomib-melphalan-prednisone) in this patient population.8 The study design and key outcomes of these trials are summarized in Table 1. Here, we will discuss three key aspects of these trials (inclusion criteria, toxicity, and efficacy) and future directions for optimizing therapy for this patient population.

TABLE 1. Key randomized controlled trials comparing quadruplets versus triplets in transplant-ineligible multiple myeloma.
IFM-2020/BENEFIT (n = 270) IMROZ (n = 446) GEM2017FIT (n = 462)
Inclusion criteria
Age 65–79 years AND non-frail (IMWG Frailty Score <2) ≥18–80 years AND TIE due to ≥65 years or comorbidities TIE with age <80 years
ECOG or Frailty cut-off ECOG >2 or Frail excluded ECOG >2 excluded Geriatric Assessment Hematology score >42
Trial treatment
Regimen Isa-VRd vs. IsaRd Isa-VRd vs. VRd VMP-Rd vs. KRd vs. Dara-KRd
Results
Age of enrolled patients (median, range unless otherwise specified) 73 (IQR 71–76) 72 (55–80) 72 (65–80)
Age ≥ 75 years 31% (IsaVRd) vs. 31% (IsaRd) 75–80: 26% (IsaVRd) vs. 32% (VRd) ~ one-third older than 75 years
% High-risk cytogenetics 10% (IsaVRd) vs. 8% (IsaRd)b 15% (Isa-VRd) vs. 19% (VRd)a ~15%
Primary and secondary endpoints
MRD negativity, 10−5 18 mo: 53% (IsaVRd) vs. 26% (IsaRd) 58% (IsaVRd) vs. 44% (VRd) 33% (VMP-Rd) vs. 75% (KRd) vs. 84% (Dara-KRd)
MRD negativity, 10−6 18 mo.: 36% (IsaVRd) vs. 17% (IsaRd) 25% (VMP-Rd) vs. 66% (KRd) vs. 79% (Dara-KRd)
≥VGPR 82% (IsaVRd) vs. 70% (IsaRd) 89% (Isa-VRD) vs. 83% (VRd) N/A
Median follow-up 23.5 months ~60 months NA
PFS 2-years PFS: 80% (IsaRd) vs. 85.2% (IsaRVd) 5-years PFS: 63% (Isa-VRD) vs. 45% (VRd) 2.5 year PFS: 73% (VMP-Rd) vs. 83% (KRd) vs. 79% (Dara-KRd)
OS 2-years OS: 91.5% (IsaRd) vs. 91.1% (IsaVRd) 5-years OS: 72% (IsaVRd) vs. 66% (VRd) 18-month OS: 91% (VMP-Rd) vs. 95% (KRD) vs. 90% (Dara-KRD)
Post-protocol therapy Not available 70% of patients in VRd arm that progressed received anti-CD38mAb N/A
Major toxicities
Neutropenia (Grade ≥3) 40% (IsaVRd) vs. 45% (IsaRd) 54% (Isa-VRd) vs. 37% (VRd) 50% (VMP-Rd) vs. KRd (24%) vs. Dara-KRd (47%)
Infections (Grade ≥3) 36% (Isa-VRd) vs. 28% (IsaRd) 45% (Isa-VRd) vs. 38% (VRd) 12% (VMP-Rd) vs. (15% (KRd) vs. 15% (Dara-KRd)
Peripheral neuropathy (all grade) 52% (Isa-VRd) vs. 28% (Isa-Rd) vs ~55% N/A
Cardiac disorders (Grade ≥3) 8% (Isa-VRd) vs. 0% (Isa-Rd) vs N/A 5% (VMP-Rd) vs. 11% (KRd) vs. 14% (Dara-KRd)
Treatment discontinuation (first 18 months due to deaths) 3.7% (Isa-VRd) vs. 2.2% (Isa-Rd) Overall treatment-related deaths: 11% (IsaVRd) vs. 5.5% (VRd) Toxicity-related death: 4.5% (VMP-Rd) vs. 3.2% (KRd) vs. 8.5% (Dara-KRd)
  • a del(17p) or HR translocations t(4;14), t (14;16).
  • b Perrot et al, JCO 2019 criteria.
  • Abbreviations: AE, adverse event; Dara-KRD, daratumumab, carfilzomib, lenalidomide, and dexamethasone; ECOG, Eastern Cooperative Oncology Group; IMWG, International Myeloma Working Group; IsaRd, isatuximab, lenalidomide, and dexamethasone; IsaRVd, isatuximab, lenalidomide, bortezomib, and dexamethasone; KRD, carfilzomib, lenalidomide, and dexamethasone; MRD, minimal residual disease; NA, not available; OS, overall survival; PFS, progression-free survival; RVd, lenalidomide, bortezomib, and dexamethasone; TIE, transplant-ineligible; VGPR, very good partial response; VMP-Rd, bortezomib, melphalan, prednisone, lenalidomide, and dexamethasone.

The first key aspect relates to eligibility criteria and the included patient population in these trials. Even though these RCTs were designed for a TIE population, they all had an upper age limit of 80 years, and approximately two-thirds of patients were within the age brackets of 65–75.6-8 Additionally, the BENEFIT and GEM2017FIT trials specifically excluded frail patients as defined by the IMWG or Geriatric Assessment in Hematology (GAH) frailty scores, respectively. Objective assessment of frailty is critical in older adults, since frailty predicts treatment-related toxicities and OS.9 Hence, the external validity of safety and efficacy of quadruplet regimens in patients who are frail or >80 years of age, which constitutes at least ~20% of TIE population,9 remain unclear. An ongoing RCT comparing Dara-VRd vs. VRd (CEPHEUS) in TIE patients does not have an upper age limit or exclusion based on frailty and will potentially generate data on the benefit of quadruplets for older patients (NCT03652064). However, it remains to be seen as to what proportion of patients in CEPHEUS trial are frail based on objective criteria. Future RCTs in this population should target specific subgroups such as fit, intermediate fit, and frail rather than arbitrarily defined category based on transplant-eligibility, which may include both patients that are truly not fit for transplant and those in whom either a transplant is deferred or not done due to patient/physician preference.

Second, early mortality and cumulative treatment-related toxicity is an important concern in older adults when adding drugs to current backbones, with real-world data showing substantial early mortality even in the modern era.10 Reassuringly, both IMROZ and BENEFIT trials demonstrated that there was no increase in the risk of early mortality with the addition of 4th drug (isatuximab and bortezomib, respectively).6, 7 However, in IMROZ, the cumulative incidence of treatment-related deaths at a median follow-up of ~5 years was approximately 2-fold higher in Isa-VRd versus VRd arm (11% vs. 5.5% respectively), with the most common cause of these deaths being infection.6 The increase in these treatment-related deaths in the Isa-VRd arm was largely driven by events beyond 6 months from treatment initiation. This highlights the deleterious impact of prolonged immunosuppression with continuous CD38mAb and dexamethasone until progression. Furthermore, it reiterates the urgent need for de-escalation strategies in frail older adults who particularly value minimizing treatment toxicity and optimizing health-related quality of life (HRQoL).11 De-escalation strategies are currently being explored in two RCTs that are randomizing patients to continuous versus fixed-duration daratumumab in the DRd regimen (NCT05561387 and NCT06182774). In the BENEFIT trial, addition of once-weekly subcutaneous bortezomib increased the risk of Grade ≥2 peripheral neuropathy from 10% in the Isa-Rd arm to 27% in Isa-VRd arm.7 However, the incidence of all-grade peripheral neuropathy with Isa-VRd in BENEFIT trial was almost half of that in IMROZ (28% vs. 54.4%, respectively), which is likely due to once-weekly bortezomib in BENEFIT versus once-weekly in IMROZ.6, 7 Since bortezomib-induced neuropathy can be reversible in about two-thirds of patients with dose-reduction or dose-discontinuation,12 it remains to be seen what proportion of these patients develop permanent neuropathy. Nevertheless, close monitoring for new-onset peripheral neuropathy while on bortezomib with prompt dose modification remains paramount if using a bortezomib-containing quadruplet regimen in this age group. Notably, in the GEM2017FIT trial, a substantial increase in toxicity-related deaths in the first 18 months was noted in the quadruplet arm (toxic deaths-4.5%, 3.2%, and 8.5% in VMP/Rd, KRd, and Dara-KRd arms, respectively).8 Furthermore, a substantial proportion of patients in the carfilzomib-containing arms (11% in KRd and 14% in Dara-KRd) developed Grade 3–4 cardiac toxicities. This is in line with the GMMG-CONCEPT trial (Isa-KRd) where the incidence of Grade 3–5 cardiac toxicities was ~10× higher in the TIE cohort compared with the TE cohort (20% vs. 2.1%, respectively).13 Together, these data imply caution with CD38mAb and carfilzomib-containing quadruplet regimens in older adults, including those who are fit and <80 years old.

Third, all three RCTs met their primary efficacy endpoint (PFS in IMROZ and MRD negativity at 10−5 by NGS in BENEFIT and GEM2017FIT). However, IMROZ trial has the most mature data thus far with a median follow-up of ~5 years.6 Importantly, in the GEM2017FIT trial, the efficacy of Dara-KRd was modulated by frailty status, with 30-month PFS in fitter patients (GAH <20) being 88% and that in patients with GAH >20 being just 71% with Dara-KRd.8 Notably, the BENEFIT trial demonstrated a ~2× increase in MRD negativity at 10−6 with the addition of bortezomib to Isa-Rd. An important caveat is that the MRD-negativity rates at 12 and 18 months were comparable in the Isa-VRd arm of BENEFIT trial, which raises the question whether bortezomib can be limited to 12 months to reduce toxicity without compromising efficacy. Additionally, the time to achievement of very good partial response or better was significantly shorter with Isa-VRd compared with Isa-Rd (2.1 vs. 3.7 mo., respectively; p = 0.0002), which highlights the importance of addition of bortezomib in situations where a rapid reduction in paraprotein is desirable such as myeloma cast nephropathy.14 Of note, the MRD-negativity rate at 10−5 (NGS) in the intention-to-treat population was comparable in the Isa-VRd arms of IMROZ and BENEFIT trials (58% vs. 53%, respectively) despite twice-weekly bortezomib in IMROZ and once-weekly in BENEFIT.6, 7 Thus far, no difference in PFS or OS has been noted with the addition of bortezomib to Isa-Rd. Unlike IMROZ trial where addition of CD38mAb led to early separation of PFS curves, addition of bortezomib has not led to the separation of PFS curves yet at a median follow-up of ~2 years. In a recent meta-analysis, MRD negativity was shown to have a strong trial-level correlation with PFS (R2 = 0.85) and medium trial-level correlation with OS (R2 = 0.79) in the TIE cohort,15 which makes it likely that the benefit in MRD negativity will translate into benefit in PFS and possibly OS with longer follow-up. Nevertheless, due to the trade-off of increased peripheral neuropathy, which has important HRQoL implications in frail older adults, mature data are needed to understand the magnitude of PFS/OS benefit to enable shared decision-making in this population. Of note, IMROZ was the only RCT that had HRQoL as an endpoint, and failed to show superiority of Isa-VRd over VRd in the Global Health Status/Quality of Life domain of EORTC-QLQ-C30 instrument.6 In a prospective cohort study in adults with MM over the age of 50, compared with younger adults (50–69 years), older adults (≥70 years) were significantly more likely to prioritize QoL, functional independence, maintaining cognitive ability, and living free from pain compared with longer OS.11 Hence, clinical trials in this population should rigorously measure HRQoL, and clinicians should be cognizant of unique patient preferences in this age group during treatment decision-making.

In summary, the outcomes of patients with newly diagnosed TIE MM are poised to improve in coming years with the introduction of effective drugs in the frontline setting; however, it remains unclear how we should translate the results of these trials in our routine clinical setting. For healthcare systems that are using VRd as the current standard of care in this population, we believe IMROZ trial provides convincing evidence to add CD38mAb to VRd backbone and should lead to a change in the standard of care. This is particularly important in fit patients and those under the age of 80. Further trials are eagerly awaited investigating the optimal duration of CD38mAb and whether routine quadruplet therapy is feasible and beneficial in frail adults including over the age of 80. However, in settings where CD38mAb + Rd is being currently used as a standard of care, it remains unclear whether addition of bortezomib is essential for all patients since enthusiasm from the benefit in terms of MRD-negativity rate is tempered by the rise in clinically significant peripheral neuropathy, and long-term follow-up will be crucial to understand the magnitude of PFS and potentially OS benefit. We believe that future trials should move away from transplant “ineligibility” as an inclusion criterion given the heterogeneity of this large category and rather focus on frailty-specified subgroups—fit, intermediate fit, and frail. Additionally, incorporating frailty-adapted attenuated dosing strategies and more dynamic fitness measures may allow us to both decrease potential toxicity and improve efficacy, allowing for more personalized treatment strategies in routine clinical practice for this heterogenous group of TIE patients.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
新诊断的符合移植条件的多发性骨髓瘤中的四联症
此外,与Isa-Rd相比,Isa-VRd达到非常好的部分应答或更好的部分应答的时间明显更短(分别为2.1个月对3.7个月;p = 0.0002),这凸显了在骨髓瘤铸型肾病等需要快速降低副蛋白的情况下加用硼替佐米的重要性。值得注意的是,尽管硼替佐米在 IMROZ 试验中为每周两次,在 BENEFIT 试验中为每周一次,但在 IMROZ 和 BENEFIT 试验的 Isa-VRd 两组中,意向治疗人群中 MRD 阴性率为 10-5(NGS)(分别为 58% 和 53%)。在 IMROZ 试验中,添加 CD38mAb 可使 PFS 曲线早期分开,但与 IMROZ 试验不同的是,在中位随访约 2 年时,添加硼替佐米尚未使 PFS 曲线分开。最近的一项荟萃分析表明,在 TIE 队列中,MRD 阴性与 PFS(R2 = 0.85)具有较强的试验水平相关性,与 OS(R2 = 0.79)具有中等的试验水平相关性15,这使得 MRD 阴性的获益很可能转化为 PFS 的获益,并可能随着随访时间的延长而转化为 OS 的获益。尽管如此,由于外周神经病变的增加会对年老体弱者的 HRQoL 产生重要影响,因此需要成熟的数据来了解 PFS/OS 的获益程度,以便在这一人群中共同决策。值得注意的是,IMROZ 是唯一一项将 HRQoL 作为终点的 RCT,但在 EORTC-QLQ-C30 工具的全球健康状况/生活质量领域,Isa-VRd 并未显示出优于 VRd。6 在一项针对 50 岁以上 MM 成人的前瞻性队列研究中,与年轻成人(50-69 岁)相比,老年人(≥70 岁)更倾向于优先考虑 QoL、功能独立性、保持认知能力和无痛生活,而不是更长的 OS。因此,针对这一人群的临床试验应严格测量 HRQoL,临床医生在治疗决策过程中应认识到这一年龄组患者的独特偏好。总之,随着一线治疗中有效药物的引入,新诊断的 TIE MM 患者的预后在未来几年有望得到改善;然而,我们应如何将这些试验结果转化为常规临床治疗仍不清楚。我们相信,IMROZ 试验提供了令人信服的证据,可以将 CD38mAb 添加到 VRd 骨架中,从而改变治疗标准。这对身体健康的患者和 80 岁以下的患者尤为重要。我们热切期待着进一步的试验,研究 CD38mAb 的最佳持续时间,以及常规的四联疗法对于包括 80 岁以上的体弱成人是否可行和有益。然而,在目前将 CD38mAb + Rd 作为标准治疗方法的情况下,是否所有患者都必须加用硼替佐米仍不清楚,因为从 MRD 阴性率中获益的热情会因临床上显著的周围神经病变的增加而减弱,长期随访对于了解 PFS 和潜在 OS 的获益程度至关重要。我们认为,鉴于移植 "不合格 "这一大类的异质性,未来的试验不应再将其作为纳入标准,而应将重点放在体弱指定的亚组--体弱、中等体弱和虚弱。此外,纳入与体弱相适应的减量给药策略和更动态的体质测量方法可能会让我们既能降低潜在毒性,又能提高疗效,从而在常规临床实践中为这一异质性的 TIE 患者群体提供更个性化的治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
15.70
自引率
3.90%
发文量
363
审稿时长
3-6 weeks
期刊介绍: The American Journal of Hematology offers extensive coverage of experimental and clinical aspects of blood diseases in humans and animal models. The journal publishes original contributions in both non-malignant and malignant hematological diseases, encompassing clinical and basic studies in areas such as hemostasis, thrombosis, immunology, blood banking, and stem cell biology. Clinical translational reports highlighting innovative therapeutic approaches for the diagnosis and treatment of hematological diseases are actively encouraged.The American Journal of Hematology features regular original laboratory and clinical research articles, brief research reports, critical reviews, images in hematology, as well as letters and correspondence.
期刊最新文献
The importance of autologous stem cell transplantation in improving outcomes in newly diagnosed patients with multiple myeloma FDA IDE validation of multiple myeloma MRD test by flow cytometry Clearance of pathogenic erythrocytes is maintained despite spleen dysfunction in children with sickle cell disease Comparison of prognostic scores according to WHO classification in 170 patients with advanced mastocytosis and C-finding treated with midostaurin Thrombotic thrombocytopenic purpura masquerading as Evans syndrome
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1