Abstract
Purpose of review:
While the traditional gold standard for demonstrating clinical benefit of a therapy has been to show prolongation of overall survival (OS), there are multiple factors which can hinder the use of OS as a primary endpoint in randomized clinical trials (RCTs). Here, we analyze recent myeloma RCTs and evaluate the issues relevant to current and future myeloma RCT design.
Recent findings:
A review of recent phase III RCTs that led to approval of new agents/combinations reveals that none were designed with OS as the primary endpoint, but instead utilized time to progression (TTP) or progression free survival (PFS). These studies illuminate the inherent difficulties of designing trials with the primary endpoint of OS/PFS in a disease characterized by increasingly prolonged survival times, availability of effective salvage therapies and competing events such as co-morbid conditions.
Summary:
Alternative primary endpoints other than OS or PFS need to be developed for future myeloma RCTs. Validated surrogate endpoints with novel clinical trial designs will help improve the feasibility of conducting comparative clinical trials in a timely manner.
Keywords: multiple myeloma, overall survival, primary endpoint, clinical trial
Introduction:
With the introduction of the immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and more recently monoclonal antibodies, the survival of patients with multiple myeloma has steadily improved over the past several decades [1]. Traditionally, the gold standard for demonstrating true clinical benefit of a therapy or intervention has been to show prolongation of overall survival (OS) with a favorable risk-to-benefit ratio and impact on health-related quality of life (HRQoL). However, there are multiple practical factors which hinder the routine use of OS as a primary endpoint in clinical trials, including the need for large numbers of patients, prolonged follow-up and the confounding effects of crossover/post-progression therapies. For some malignancies such as myeloma, the steady approval of new agents and combinations over the past decade has ensured the availability of multiple lines of effective salvage therapies. There is an increasingly important need to define surrogate endpoints that are associated with clinical benefit yet can be reached in a timely manner to allow for rapid translation into the clinic. Here we provide an overview of the use of different endpoints in oncology and then focus on lessons learned from phase III studies performed in myeloma. We review the recent literature focused on developing novel endpoints in myeloma and discuss the implications for the design of future studies.
Overview of clinical trial endpoints in oncology:
In the early 1980’s, the Food and Drug Administration (FDA) primarily approved drugs based on objective overall response rate (ORR). However, following discussions with the Oncologic Drugs Advisory Committee, the FDA indicated that oncology drug approval should be based on more direct evidence of clinical benefit, including improvements in OS, HRQoL, tumor-related symptoms and/or physical functioning [2]. It was noted that ORR may not predict these types of clinical benefits. Since that time, several endpoints have become accepted as surrogates for clinical benefit, including disease-free survival (DFS) and relief of tumor-specific symptoms. In 1992, Subpart H was added to the new drug application (NDA) regulations in order to create an accelerated approval process. This process was created to allow patients with life-threatening diseases earlier access to therapies that have evidence of benefit but do not yet meet the standard for regular approval. In this context, the accelerated approval is often based on surrogate endpoints such as DFS, time to progression (TTP) or ORR [3].
Historically, in phase III randomized clinical trials (RCTs), OS has been the gold standard endpoint for demonstrating clinical benefit. OS time does not rely upon surveillance schedules, methods of monitoring, number/location of lesions or interpretation of patient symptoms, scans or blood work to draw conclusions concerning disease response. However, a RCT utilizing OS as its primary endpoint usually requires several years of follow-up and large sample sizes. Interpretation of the trial results may become difficult as patients who did not receive the investigational agent in the RCT may receive it later in their disease course or as part of the crossover portion of the RCT. Other potential confounders include variations in supportive care and post-progression therapies over the course of the trial and competing causes of death. Analysis of survival data fails to consider whether the survival period consists of patients receiving multiple lines of therapy in an effort to prolong survival or whether the patient is suffering from a co-morbid condition that will be the eventual cause of death.
TTP and progression-free survival (PFS) are commonly used as primary endpoints in RCTs. Although these studies generally utilize consensus guidelines for determining response (e.g., RECIST criteria for solid tumors, International Myeloma Working Group criteria for myeloma [4]), investigator subjectivity can still be a confounding factor and it is preferable to utilize a blinded external review committee to adjudicate responses. While a study might demonstrate a statistically significant treatment benefit in TTP/PFS, it may not translate to a clinically meaningful OS benefit. TTP and PFS have been proposed as surrogate endpoints for OS. A surrogate endpoint is an outcome measure where changes induced by a treatment intervention reflect changes in the clinical endpoint (such as OS) and reliably predicts whether clinically meaningful changes in the clinical endpoint will occur [5]. Fleming and Powers provide insight into why a putative surrogate endpoint may fail to reliably reflect changes in the clinical endpoint [5]. The use of TTP and PFS as surrogate endpoints for OS should be limited to situations where studies have been conducted to validate the association between prolongation in TTP/PFS and a clinically meaningful prolongation in OS. Cartier et al., performed a meta-analysis of 21 myeloma RCTs (14 first-line, 4 maintenance and 3 relapsed/refractory) using trial level data [6]. They found a moderate to strong positive correlation between hazard ratios for treatment effects for PFS and OS and advocate that patient level data be used to validate these findings.
PFS has also been suggested as a possible surrogate for health-related quality of life (HRQoL). However, there has been little work done in this area for myeloma. In a recent study by Kovic et al., a systematic review was performed evaluating 38 randomized oncology studies to determine whether PFS is associated with HRQoL [7]. This study included only one myeloma RCT and the authors ended up removing this study as an outlier in their analysis. As noted by Freidlin et al., progression in hematological malignancies is often determined by changes in laboratory measures that may not be accompanied by deleterious effects on HRQoL [8].
Phase III RCTs in multiple myeloma:
A summary of key phase III myeloma studies involving PI’s, IMiDs or other novel agents is provided in Table 1. The majority of these studies changed the standard of care and/or led to the subsequent approval of new agents or combinations. Notably, none of these RCTs were designed with OS as its primary endpoint, rather the majority used TTP or PFS as the primary endpoint. Whether prolongation of PFS was associated with prolongation of OS in these studies was variable. The interpretation of the impact of the experimental arm/combination on the prolongation of OS is confounded by the large percentage of patients randomized to the standard treatment arm who switch to the experimental arm at disease progression or when PFS findings are released. As OS is a secondary endpoint in many trials, there is often no a priori plan for the number of deaths needed to have sufficient power to detect a pre-specified survival advantage with the experimental regimen. OS findings are often reported when PFS findings are first reported and the early OS findings may not hold or will change with longer follow-up.
Table 1.
Summary of recent phase III RCTs in myeloma
Study | Patient Population | n | Randomization | Primary Endpoint1 | OS outcomes |
---|---|---|---|---|---|
SWOG S0777 [37] | ND | 525 | RVD vs RD induction | PFS 43 vs 30 mos; p=0.0018 |
Median: 75 vs 64 months; p=0.025 |
GIMEMA-MMY- 3006 [16, 38] |
ND TE | 480 | VTD vs TD (induction, transplant, consolidation) | CR/nCR rate after induction: 31% vs 11%, p<0.001 | Median: NR vs 110 mos; p=0.007 |
IFM2009 [23] | ND TE | 700 | Consolidation with single ASCT vs RVD | PFS 50 vs 36 mos; p<0.01 |
4-yr: 81 vs 82% |
StaMINA [24] | ND TE | 758 | Single ASCT vs single ASCT + RVD consolidation vs double ASCT |
PFS 38-mos: 52.2 vs 56.7 vs 56.5% |
38-mos: 83.4 vs 85.7 vs 82.0% |
VISTA [39, 40] | ND TI | 682 | VMP vs MP | TTP 24.0 vs 16.6 mos; p<0.001 |
Median: 56.4 vs 43.1 mos, p<0.001 |
FIRST [41, 42] | ND TI | 1623 | RD continuous vs RD18 vs MPT |
PFS 25.5 vs 20.7 vs 21.2 mos |
Median: 59.1 vs 62.3 vs 49.1 mos |
HOVON87/NMSG18 [43] |
ND TI | 668 | MPT-T vs MPR- R |
PFS 20 vs 23 mos; p=0.12 |
4 yr: 52% vs 56%; p=0.13 |
ECOG E1A06 [44] | ND TI | 306 | MPT-T vs MPR- R |
PFS 21.0 vs 18.7 mos; p=0.186 | Median: 52.6 vs 47.7 mos; p=0.476 |
GIMEMA RV-2009 [45] |
ND TE | 524 | 1st: ASCT vs MPR 2nd: R maintenance vs observation |
PFS from R1 43.0 (ASCT) vs 22.4 (MPR) mos; p<0.001 | 4 yr: 81.6 vs 65.3%; p=0.02 |
RV-MM-EMN-441 [46] |
ND TE | 389 | 1st: ASCT vs CRD 2nd: maintenance with R vs RP |
PFS from R1 43.3 (ASCT) vs 28.6 (CRD) mos; p<0.001 |
4 yr: 86% vs 73%; p=0.04 |
EMN02/HO95 [47] | ND TE | 1266 | 1st: ASCT vs VMP 2nd: VRD consolidation vs none |
PFS from R1 3 yr: 66% (ASCT) vs 57.5% (VMP) |
Not reported |
CALGB 100104 [15, 48] | Post-ASCT maintenance | 460 | R vs placebo maintenance | TTP 57.3 vs 28.9 mos; p<0.0001 |
Median: 113.8 vs 84.1 mos; p=0.0004 |
IFM 2005–02 [49] | Post-ASCT maintenance | 614 | R vs placebo maintenance | PFS 41 vs 23 mos; p<0.001 |
4-yr: 73% vs 75%; p=0.7 |
MM-009 and MM- 010 [9, 50, 51] |
R/R | 704 | RD vs placeboD | TTP 13.4 vs 4.6 mos; p<0.001 |
Median: 38.0 vs 31.6 mos; p=0.045 |
APEX [10, 11] | R/R | 669 | V vs D | TTP 6.2 vs 3.5 mos, p<0.001 |
Median: 29.8 vs 23.7 mos; p=0.027 |
A.R.R.O.W. [52] | R/R | 478 | Once- vs twiceweekly KD | PFS 11.2 vs 7.6 mos; p=0.0029 |
Median: NR vs NR; p=0.214 |
ENDEAVOR [17, 18] | R/R | 929 | KD vs VD | PFS 18.7 vs 9.4 mos; p<0.0001 |
Median: 47.6 vs 40 mos; p=0.01 |
POLLUX [12] | R/R | 569 | Dara-RD vs RD | PFS NR vs 18.4 mos; p<0.001 | Median: NR vs 20.3 mos; p=0.0534 |
CASTOR [19] | R/R | 498 | Dara-VD vs VD | PFS NR vs 7.2 mos; p<0.001 |
Median: NR vs NR; HR 0.77, p=0.30 |
ASPIRE [13, 14] | R/R | 729 | KRD vs RD | PFS 26.3 vs 17.6 mos; p=0.0001 |
Median: 48.3 vs 40.4 mos; p=0.0045 |
TOURMALINE-MM1 [20] |
R/R | 722 | IRD vs RD | PFS 20.6 vs 14.7 mos; p=0.01 |
Median: NR vs NR |
ELOQUENT-2 [21] | R/R | 646 | Elo-RD vs RD | PFS 19.4 vs 14.9 mos; p<0.001 |
Median: NR vs NR |
PANORMA-1 [53, 54] |
R/R | 768 | Pano-VD vs VD | PFS 12.0 vs 8.1 mos; p<0.0001 |
Median: 40.3 vs 35.8 mos; p=0.51 |
OPTIMISMM [55] | R/R | 559 | PVD vs VD | PFS 11.2 vs 7.1 mos; p<0.0001 |
Not reported |
Abbreviations: ASCT, autologous stem cell transplant; CRD, cyclophosphamide/lenalidomide/dexamethasone; D, dexamethasone; Dara-VD, daratumumab/bortezomib/dexamethasone; Dara-VMP, daratumumab/bortezomib/melphalan/prednisone; Dara-RD, daratumumab/lenalidomide/dexamethasone; Elo-RD, elotuzumab/lenalidomide/dexamethasone; IRD, ixazomib/lenalidomide/dexamethasone; KD, carfilzomib/dexamethasone; MP, melphalan/prednisone; MPR, melphalan/prednisone/lenalidomide; MPT, melphalan/thalidomide/prednisone; ND, newly diagnosed; NR, not reached; Pano-VD, panobinostat/bortezomib/dexamethasone; PVD, pomalidomide/bortezomib/dexamethasone; R, lenalidomide; R1, 1st randomization; RD, lenalidomide/dexamethasone; RP, lenalidomide/prednisone; RVD, lenalidomide/bortezomib/dexamethasone; T, thalidomide; TE, transplant eligible; TI, transplant ineligible; VD, bortezomib/dexamethasone; VMP, bortezomib/melphalan/prednisone;
In the pooled analysis of two phase III RTCs comparing lenalidomide/dexamethasone to placebo/dexamethasone in relapsed/refractory patients, 48% of patients on the placebo arm received lenalidomide-based therapy after unblinding of the studies (following findings of the first interim analyses of each respective study) or after disease progression [9]. With a median follow-up of 48 months for surviving patients, the addition of lenalidomide to dexamethasone significantly improved PFS (p<0.001) and overall survival (p=0.045). In APEX, a phase III RCT comparing bortezomib to dexamethasone in relapsed/refractory patients, 62% of the patients randomized to dexamethasone crossed over to receive bortezomib at time of disease progression [10, 11]. The study was halted at the first interim analysis when the superiority of bortezomib was demonstrated in terms of TTP and OS. Although an OS advantage (HR =0.57, p=0.001) was seen, the median length of follow-up for those patients still alive was only 8.3 months. This type of post-progression treatment information is not available from some of the more recent studies, but presumably a significant number of patients randomized to doublet therapies instead of triplet therapies (e.g., those individuals in the lenalidomide/dexamethasone arms of the POLLUX (evaluating the addition of daratumumab [12]) or ASPIRE (evaluating the addition of carfilzomib [13, 14]) trials) later received the novel agent in question with a subsequent line of therapy.
Within the last several years, a number of RCTs in both the newly diagnosed or post-transplant settings have had sufficient follow-up to report median OS data in one or both treatment arms. These findings have provided the field with new benchmarks to judge clinical benefit, necessitating the next generation of RCTs to be longer in duration and/or larger in sample size. For example, CALGB 100104, which randomized patients to either lenalidomide or placebo maintenance following autologous stem cell transplant (ASCT) reported a median OS of 9.5 years and 7.0 years for the lenalidomide arm and placebo arm, respectively [15]. In this trial, OS time began at the start of ASCT, which was typically 6–12 months after initiation of induction therapy. The GIMEMA-MMY-3006 study, which randomized patients to triple therapy with VTD (bortezomib, thalidomide, dexamethasone) vs doublet therapy with TD (thalidomide, dexamethasone) as induction/consolidation around tandem ASCT, reported a median OS of 9.9 years for the TD arm, but the median has not yet been reached for the VTD arm [16]. In the relapsed/refractory setting, a number of recent phase III RCTs have demonstrated the superiority of triplet over doublet combinations, where the median OS durations of the triplet arms have either been in the 4-year range [13, 14, 17, 18] or have not yet been reached [12, 19–21].
In aggregate these studies highlight the difficulty of planning future prospective phase III RCTs in myeloma with OS as the primary endpoint. When planning such trials there is often a trade-off between the number of patients that can be enrolled per year and the minimum length of follow-up once enrollment is terminated to ensure there is sufficient number of events to assess the primary endpoint with adequate power. The question often arises whether the duration of the study is too long such that by the time the data are mature, the primary aim may no longer be clinically relevant. The planned SWOG S1803 study is an example of a study which may be hindered by many of these issues. This study is a phase III RCT comparing lenalidomide to daratumumab + lenalidomide maintenance therapy post-ASCT. MRD status is evaluated after two years of maintenance. Maintenance therapy is continued for patients who are MRD positive. For patients who are MRD negative, there is a randomization to either continuing or discontinuing maintenance therapy. This study has been designed with OS as the primary objective and powered to compare a median OS of 10 years in the lenalidomide arm to 15.7 years in the combination arm. To achieve this endpoint, 950 patients will need to be accrued over an estimated 6 years, and accounting for drop-out, 1100 patients will need to be enrolled. The study has been designed to include a number of interim analyses to evaluate for futility and there are plans to report earlier endpoints such as the two-year MRD negativity rate one year after all patients are randomized as well as PFS. However, given the very large numbers of patients required, the prolonged enrollment period, the expected median PFS of the control arm (4.8 years based on CALGB 100104 [15]), even the earlier endpoints are predicted to be reported at a minimum of 8 years after the start of the study. Given the rapid rate at which the myeloma therapeutic landscape is evolving and other studies such as the randomized phase II study GRIFFIN study which has already completed enrollment and evaluates lenalidomide vs daratumumab + lenalidomide maintenance post-transplant [22], it is possible that by the time the S1803 study is completed, the primary question will no longer be clinically relevant.
As noted in Table 1, nearly all recent phase III studies have used TTP or PFS as the primary endpoint. The IFM 2009 study has provided a new benchmark for PFS in the newly diagnosed, transplant-eligible setting. In this study, 700 newly diagnosed patients were enrolled between 2010–2012 and received RVD (lenalidomide, bortezomib, dexamethasone) induction followed by randomization to consolidation with ASCT or RVD. The estimated median PFS was 4.2 years and 3.0 years for the ASCT and RVD arms, respectively [23]. Median OS has not yet been reached for either arm in this study. The StaMINA study is another example that highlights the numbers of patients and duration of follow-up required to evaluate PFS as a primary endpoint in the newly diagnosed setting. This study enrolled 758 patients between 2010–2013 and randomized them in equal numbers to single ASCT, tandem ASCT or single ASCT plus RVD consolidation [24]. This study has required a prolonged follow-up and median PFS numbers have not yet been reported. Thus the issues of sample size and study duration associated with planning a RTC where OS is prolonged are also present when planning a RTC in a disease setting with a prolonged PFS time. Another issue that arises in planning the analytic approach to be taken when PFS is the primary endpoint is the extent to which competing events prior to disease progression (such as death due to co-morbid conditions, development of second primary malignancies or initiation of alternative therapies) may be present in order to avoid biased estimates of effect.
Phase II studies in myeloma:
Historically, the most common primary endpoint for phase II studies in the relapsed/refractory setting has been the ORR, typically defined as partial response (PR) or better. With the increasing number of salvage therapy options and identification of molecular subtypes with varying prognoses, it is becoming more difficult to determine appropriate benchmarks for determining whether an agent or combination is promising in a given patient population. For example, Vogl et al., conducted a phase II study assessing whether the ORR with the combination of selinexor and dexamethasone in patients with either quad-refractory (defined as refractory to bortezomib, carfilzomib, lenalidomide, pomalidomide) or penta-refractory (quad-refractory plus refractory to anti-CD38 monoclonal antibody) disease was at most 15% against the alternative hypothesis that it was 30% [25]. The observed ORR in this study was 21%. The authors noted that in retrospect, given the lack of suitable comparators with penta-refractory patients, they should have set their threshold at 10% [25].
A number of studies have reported that the achievement of deeper responses (e.g., very good partial response (VGPR) or complete response (CR)) is associated with improved survival outcomes (reviewed in [26]). Several recent phase II studies have as their primary endpoint VGPR + CR rate [27–29] or CR or stringent CR (sCR) rates (NCT02874742, NCT03012880, NCT03012880). However, it is evident that not all patients who achieve CR or sCR have equivalent outcomes. This heterogeneity is in part due to the presence of minimal residual disease (MRD). A recent meta-analysis of studies which evaluated MRD status in myeloma patients who had achieved a CR revealed that MRD-negativity was associated with increased PFS and OS compared to MRD-positivity [30].
MRD as an endpoint in myeloma studies:
MRD assessment by either flow cytometry or next generation sequencing (NGS), was incorporated into the International Myeloma Working Group (IMWG) response criteria in 2016 [4]. Currently these response criteria specify that a minimum sensitivity of 1 × 10−5 be utilized, however, increasingly, a sensitivity of 1 × 10−6 is becoming a standard of care. The prognostic significance of MRD negativity has been confirmed across the spectrum of myeloma, including newly diagnosed transplant-eligible patients undergoing ASCT, newly diagnosed transplant-ineligible patients, patients receiving maintenance therapy, and relapsed/refractory patients (reviewed in [31]). These findings have raised the question as to whether MRD may be a surrogate endpoint for OS. Currently, MRD is considered a prognostic biomarker, defined as a biomarker that can help identify patients at higher risk of adverse disease-related outcomes. In contrast, response biomarkers are defined as being dynamic assessments that show a biological response has occurred in a patient receiving a therapeutic intervention [32].
Validation studies are needed to demonstrate that a change in MRD status reliably predicts a meaningful change in OS. An assay that is reliable, reproducible and validated for detection of MRD and a cut-point for defining negativity are important pieces in the validation process. The clonoSEQ assay, which evaluates MRD by NGS, was recently given De Novo designation by the FDA for the detection and monitoring of MRD in myeloma patients (https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm622004.htm). Next steps encompass both individual and trial-level validation. In planning these validation studies, Kemp et al., point out that care must be given to minimizing measurement error, evaluation bias, attribution bias or informative censoring which may weaken the association between the potential surrogate and the clinical endpoint [33].
Innovative clinical trial design strategies:
Clinical trial methodology has had to adapt to the changing landscape of limited patient numbers, lower and lower event rates with standard treatments, multiple investigational agents/treatment strategies to be evaluated, costs of trial conduct and the assessment of multiple clinical and correlative hypotheses. Seamless phase II/III clinical trial designs have been proposed where a number of investigational agents are evaluated and the most promising is then brought forward to be compared to the standard treatment [34]. Multi-arm, multi-stage (MAMS) clinical trial designs were proposed when the objective is to compare a number of investigational treatments to the standard treatment [35]. Pre-planned interim analyses are conducted to determine which experimental treatments should continue until their pre-specified maximum sample size is attained. Treatments that lack sufficient activity can be replaced with new investigational agents.
Insights into the molecular genetic aberrations that drive cancer growth have led to the identification of molecular subtypes within a given primary cancer that have heterogeneous clinical outcomes. Novel agents/treatment strategies are being developed and tested on a limited number of individuals with a given molecular cancer subtype. The umbrella trial design typically involves a set of enrichment trials for patients with a single type of cancer but with different genomic alterations [36]. In contrast, basket clinical trials have been proposed to assess the impact of an investigational agent on cancers with a specific mutation rather than conducting a clinical trial in each specific primary tumor site [36]. Thus, the validation of surrogate endpoints, coupled with the incorporation of novel clinical trial designs, will enable future comparative clinical trials to be conducted in a timely manner.
Conclusions and Future Directions
The efficacy of the current standard-of-care combinations as well as those containing investigational agents, coupled with the availability of multiple lines of effective salvage regimens has translated to prolonged OS times for myeloma patients. Moving forward, the detection of clinically significant OS differences will therefore not only require increasingly larger numbers of patients and prolonged follow-up periods but also understanding of the post-progression treatment patterns and competing risks. Thus overall, the conduct of large RCTs with OS, or even PFS as the primary endpoint, is becoming increasingly less feasible in myeloma. While MRD status may prove to be a suitable surrogate endpoint for survival, with the advantage of providing an earlier read-out for studies, the identification of molecular subsets of myeloma and the development of therapies specific for those subsets, will require novel trial designs that extend beyond the traditional RCT approach.
Conflict of Interest:
Sarah Holstein reports serving as an advisory board member for Celgene, Takeda and Adaptive Biotechnologies and as a consultant for Celgene and GlaxoSmithKline, outside the submitted work. Vera Suman declares no potential conflicts of interest. Philip McCarthy reports receiving honoraria from Bristol-Myers Squibb, Celgene, Sanofi-Aventis, Takeda and Binding Site, research funding from Celgene, and has served on advisory committees/review panels/board membership for Bristol-Myers Squibb, Celgene, Sanofi-Aventis, Takeda, Binding Site and Karyopharm outside the submitted work.
Footnotes
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
*Of importance:
--Reference #15: Provides a new benchmark for OS in the post-transplant setting
--Reference #16: Provides a new benchmark for OS in the newly diagnosed setting
--Reference #30: Meta-analysis demonstrating the superior survival outcomes of patients achieving MRD-negativity
**Of major importance
--Reference #6: Systematic review of trial-level data assessing PFS as a surrogate for OS in myeloma
- 1.Costa LJ, Brill IK, Omel J, Godby K, Kumar SK, Brown EE. Recent trends in multiple myeloma incidence and survival by age, race, and ethnicity in the United States. Blood Adv 2017; 1: 282–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Johnson JR, Temple R. Food and Drug Administration requirements for approval of new anticancer drugs. Cancer Treat Rep 1985; 69: 1155–9. [PubMed] [Google Scholar]
- 3.Johnson JR, Williams G, Pazdur R. End points and United States Food and Drug Administration approval of oncology drugs. J Clin Oncol 2003; 21: 1404–11. [DOI] [PubMed] [Google Scholar]
- 4.Kumar S, Paiva B, Anderson KC, Durie B, Landgren O, Moreau P, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol 2016; 17: e328–46. [DOI] [PubMed] [Google Scholar]
- 5.Fleming TR, Powers JH. Biomarkers and surrogate endpoints in clinical trials. Stat Med 2012; 31: 2973–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cartier S, Zhang B, Rosen VM, Zarotsky V, Bartlett JB, Mukhopadhyay P, et al. Relationship between treatment effects on progression-free survival and overall survival in multiple myeloma: a systematic review and meta-analysis of published clinical trial data. Oncol Res Treat 2015; 38: 88–94. [DOI] [PubMed] [Google Scholar]
- 7.Kovic B, Jin X, Kennedy SA, Hylands M, Pedziwiatr M, Kuriyama A, et al. Evaluating ProgressionFree Survival as a Surrogate Outcome for Health-Related Quality of Life in Oncology: A Systematic Review and Quantitative Analysis. JAMA Intern Med 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Freidlin B, Little RF, Korn EL. Design Issues in Randomized Clinical Trials of Maintenance Therapies. J Natl Cancer Inst 2015; 107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dimopoulos MA, Chen C, Spencer A, Niesvizky R, Attal M, Stadtmauer EA, et al. Long-term follow-up on overall survival from the MM-009 and MM-010 phase III trials of lenalidomide plus dexamethasone in patients with relapsed or refractory multiple myeloma. Leukemia 2009; 23: 2147–52. [DOI] [PubMed] [Google Scholar]
- 10.Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med 2005; 352: 2487–98. [DOI] [PubMed] [Google Scholar]
- 11.Richardson PG, Sonneveld P, Schuster M, Irwin D, Stadtmauer E, Facon T, et al. Extended follow-up of a phase 3 trial in relapsed multiple myeloma: final time-to-event results of the APEX trial. Blood 2007; 110: 3557–60. [DOI] [PubMed] [Google Scholar]
- 12.Dimopoulos MA, Oriol A, Nahi H, San-Miguel J, Bahlis NJ, Usmani SZ, et al. Daratumumab, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med 2016; 375: 1319–31. [DOI] [PubMed] [Google Scholar]
- 13.Siegel DS, Dimopoulos MA, Ludwig H, Facon T, Goldschmidt H, Jakubowiak A, et al. Improvement in Overall Survival With Carfilzomib, Lenalidomide, and Dexamethasone in Patients With Relapsed or Refractory Multiple Myeloma. J Clin Oncol 2018; 36: 728–34. [DOI] [PubMed] [Google Scholar]
- 14.Stewart AK, Rajkumar SV, Dimopoulos MA, Masszi T, Spicka I, Oriol A, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med 2015; 372: 142–52. [DOI] [PubMed] [Google Scholar]
- 15.Holstein SA, Jung SH, Richardson PG, Hofmeister CC, Hurd DD, Hassoun H, et al. Updated analysis of CALGB (Alliance) 100104 assessing lenalidomide versus placebo maintenance after single autologous stem-cell transplantation for multiple myeloma: a randomised, double-blind, phase 3 trial. Lancet Haematol 2017; 4: e431–e42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tacchetti P, Patriarca F, Petrucci M, Galli M, Pantani L, Dozza L, et al. , editors. A triplet bortezomib- and immunomodulator-based therapy before and after double ASCT improves overall survival of newly diagnosed MM patients: final analysis of phase 3 GIMEMA-MMY-3006 Study. EHA 2018; 2018. [Google Scholar]
- 17.Dimopoulos MA, Goldschmidt H, Niesvizky R, Joshua D, Chng WJ, Oriol A, et al. Carfilzomib or bortezomib in relapsed or refractory multiple myeloma (ENDEAVOR): an interim overall survival analysis of an open-label, randomised, phase 3 trial. Lancet Oncol 2017; 18: 1327–37. [DOI] [PubMed] [Google Scholar]
- 18.Dimopoulos MA, Moreau P, Palumbo A, Joshua D, Pour L, Hajek R, et al. Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study. Lancet Oncol 2016; 17: 27–38. [DOI] [PubMed] [Google Scholar]
- 19.Palumbo A, Chanan-Khan A, Weisel K, Nooka AK, Masszi T, Beksac M, et al. Daratumumab, Bortezomib, and Dexamethasone for Multiple Myeloma. N Engl J Med 2016; 375: 754–66. [DOI] [PubMed] [Google Scholar]
- 20.Moreau P, Masszi T, Grzasko N, Bahlis NJ, Hansson M, Pour L, et al. Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med 2016; 374: 1621–34. [DOI] [PubMed] [Google Scholar]
- 21.Lonial S, Dimopoulos M, Palumbo A, White D, Grosicki S, Spicka I, et al. Elotuzumab Therapy for Relapsed or Refractory Multiple Myeloma. N Engl J Med 2015; 373: 621–31. [DOI] [PubMed] [Google Scholar]
- 22.Voorhees PM, Rodriguez C, Reeves B, Nathwani N, Costa LJ, Lutska Y, 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: 151-. [Google Scholar]
- 23.Attal M, Lauwers-Cances V, Hulin C, Leleu X, Caillot D, Escoffre M, et al. Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma. N Engl J Med 2017; 376: 1311–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Stadtmauer EA, Pasquini MC, Blackwell B, Knust K, Bashey A, Devine SM, et al. Comparison of autologous hematopoietic cell transplant (autoHCT), bortezomib, lenalidomide (Len) and dexamethasone (RVD) consolidation with Len maintenance (ACM), tandem autoHCT with Len maintenance (TAM) and autoHCT with Len maintenance (AM) for up-front treatment of patients with multiple myeloma (MM): primary results from the randomized phase III trial of the Blood and Marrow Transplant Clinical Trials Network (BMT CTN 0702 - StaMINA trial). Blood (ASH Abstracts) 2016; 128: LBA-1. [Google Scholar]
- 25.Vogl DT, Dingli D, Cornell RF, Huff CA, Jagannath S, Bhutani D, et al. Selective Inhibition of Nuclear Export With Oral Selinexor for Treatment of Relapsed or Refractory Multiple Myeloma. Journal of Clinical Oncology 2018; 36: 859–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Landgren O, Iskander K. Modern multiple myeloma therapy: deep, sustained treatment response and good clinical outcomes. J Intern Med 2017; 281: 365–82. [DOI] [PubMed] [Google Scholar]
- 27.Kumar SK, Buadi FK, LaPlant B, Halvorson A, Leung N, Kapoor P, et al. Phase 1/2 trial of ixazomib, cyclophosphamide and dexamethasone in patients with previously untreated symptomatic multiple myeloma. Blood Cancer J 2018; 8: 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Richardson PG, Hofmeister CC, Rosenbaum CA, Htut M, Vesole DH, Berdeja JG, et al. Twice-weekly ixazomib in combination with lenalidomide-dexamethasone in patients with newly diagnosed multiple myeloma. Br J Haematol 2018; 182: 231–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wester R, van der Holt B, Asselbergs E, Zweegman S, Kersten MJ, Vellenga E, et al. Carfilzomib Combined with Thalidomide and Low-Dose Dexamethasone for Remission Induction and Consolidation in Newly Diagnosed, Transplant Eligible Patients with Multiple Myeloma: The Carthadex Trial. Blood 2017; 130: 3141-. [Google Scholar]
- 30.Munshi NC, Avet-Loiseau H, Rawstron AC, Owen RG, Child JA, Thakurta A, et al. Association of Minimal Residual Disease With Superior Survival Outcomes in Patients With Multiple Myeloma: A Meta-analysis. JAMA Oncol 2017; 3: 28–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Davies FE. Is molecular remission the goal of multiple myeloma therapy? Hematology Am Soc Hematol Educ Program 2017; 2017: 205–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Amur S, LaVange L, Zineh I, Buckman-Garner S, Woodcock J. Biomarker Qualification: Toward a Multiple Stakeholder Framework for Biomarker Development, Regulatory Acceptance, and Utilization. Clin Pharmacol Ther 2015; 98: 34–46. [DOI] [PubMed] [Google Scholar]
- 33.Kemp R, Prasad V. Surrogate endpoints in oncology: when are they acceptable for regulatory and clinical decisions, and are they currently overused? BMC Med 2017; 15: 134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chen C, Anderson K, Mehrotra DV, Rubin EH, Tse A. A 2-in-1 adaptive phase 2/3 design for expedited oncology drug development. Contemp Clin Trials 2018; 64: 238–42. [DOI] [PubMed] [Google Scholar]
- 35.Lin J, Bunn V. Comparison of multi-arm multi-stage design and adaptive randomization in platform clinical trials. Contemp Clin Trials 2017; 54: 48–59. [DOI] [PubMed] [Google Scholar]
- 36.Simon R Critical Review of Umbrella, Basket, and Platform Designs for Oncology Clinical Trials. Clin Pharmacol Ther 2017; 102: 934–41. [DOI] [PubMed] [Google Scholar]
- 37.Durie BG, Hoering A, Abidi MH, Rajkumar SV, Epstein J, Kahanic SP, 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: 519–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cavo M, Tacchetti P, Patriarca F, Petrucci MT, Pantani L, Galli M, et al. Bortezomib with thalidomide plus dexamethasone compared with thalidomide plus dexamethasone as induction therapy before, and consolidation therapy after, double autologous stem-cell transplantation in newly diagnosed multiple myeloma: a randomised phase 3 study. Lancet 2010; 376: 2075–85. [DOI] [PubMed] [Google Scholar]
- 39.San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med 2008; 359: 906–17. [DOI] [PubMed] [Google Scholar]
- 40.San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, et al. Persistent overall survival benefit and no increased risk of second malignancies with bortezomib-melphalan-prednisone versus melphalan-prednisone in patients with previously untreated multiple myeloma. J Clin Oncol 2013; 31: 448–55. [DOI] [PubMed] [Google Scholar]
- 41.Facon T, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, Cavo M. Final Survival Analysis From the FIRST Trial: Lenalidomide Plus Low-Dose Dexamethasone Until Progression (Rd Cont) v Melphalan, Prednisone and Thalidomide (MPT), and Rd for 18 Cycles (Rd18) for Transplant-Ineligible (TNE) Patients (Pts) With Newly Diagnosed Multiple Myeloma. Clinical Lymphoma, Myeloma and Leukemia; 17: e63–e4. [Google Scholar]
- 42.Benboubker L, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, Cavo M, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med 2014; 371: 906–17. [DOI] [PubMed] [Google Scholar]
- 43.Zweegman S, van der Holt B, Mellqvist UH, Salomo M, Bos GM, Levin MD, et al. Melphalan, prednisone, and lenalidomide versus melphalan, prednisone, and thalidomide in untreated multiple myeloma. Blood 2016; 127: 1109–16. [DOI] [PubMed] [Google Scholar]
- 44.Stewart AK, Jacobus S, Fonseca R, Weiss M, Callander NS, Chanan-Khan AA, et al. Melphalan, prednisone, and thalidomide vs melphalan, prednisone, and lenalidomide (ECOG E1A06) in untreated multiple myeloma. Blood 2015; 126: 1294–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Palumbo A, Cavallo F, Gay F, Di Raimondo F, Ben Yehuda D, Petrucci MT, et al. Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med 2014; 371: 895–905. [DOI] [PubMed] [Google Scholar]
- 46.Gay F, Oliva S, Petrucci MT, Conticello C, Catalano L, Corradini P, et al. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet Oncol 2015; 16: 1617–29. [DOI] [PubMed] [Google Scholar]
- 47.Cavo M, Palumbo A, Zweegman S, Dimopoulos M, Hajek R, Pantani L, et al. Upfront autologous stem cell transplantation (ASCT) versus novel agent-based therapy for multiple myeloma (MM): a randomized phase 3 study of the European Myeloma Network (EMN02/HO95 MM trial). J Clin Oncol 2016; 34: abstr 8000. [Google Scholar]
- 48.McCarthy PL, Owzar K, Hofmeister CC, Hurd DD, Hassoun H, Richardson PG, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med 2012; 366: 1770–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Attal M, Lauwers-Cances V, Marit G, Caillot D, Moreau P, Facon T, et al. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. N Engl J Med 2012; 366: 1782–91. [DOI] [PubMed] [Google Scholar]
- 50.Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau JL, Dmoszynska A, et al. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 2007; 357: 2123–32. [DOI] [PubMed] [Google Scholar]
- 51.Weber DM, Chen C, Niesvizky R, Wang M, Belch A, Stadtmauer EA, et al. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 2007; 357: 2133–42. [DOI] [PubMed] [Google Scholar]
- 52.Moreau P, Mateos MV, Berenson JR, Weisel K, Lazzaro A, Song K, et al. Once weekly versus twice weekly carfilzomib dosing in patients with relapsed and refractory multiple myeloma (A.R.R.O.W.): interim analysis results of a randomised, phase 3 study. Lancet Oncol 2018; 19: 953–64. [DOI] [PubMed] [Google Scholar]
- 53.San-Miguel JF, Hungria VT, Yoon SS, Beksac M, Dimopoulos MA, Elghandour A, et al. Overall survival of patients with relapsed multiple myeloma treated with panobinostat or placebo plus bortezomib and dexamethasone (the PANORAMA 1 trial): a randomised, placebo-controlled, phase 3 trial. Lancet Haematol 2016; 3: e506–e15. [DOI] [PubMed] [Google Scholar]
- 54.San-Miguel JF, Hungria VT, Yoon SS, Beksac M, Dimopoulos MA, Elghandour A, et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet Oncol 2014; 15: 1195–206. [DOI] [PubMed] [Google Scholar]
- 55.Richardson P, Rocafiguera AO, Beksac M, Liberati AM, Galli M, Schjesvold F, et al. , editors. OPTIMISMM: Phase 3 trial of pomalidomide, bortezomib and low-dose dexamethasone vs bortezomib and low-dose dexamethasone in lenalidomide-exposed patients with relapsed/refractory multiple myeloma. European Hematology Association Annual Meeting; 2018. [Google Scholar]