We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short CommunicationOpen Accesscc iconby iconnc iconnd icon

The value of the accelerated approval pathway: real-world outcomes associated with reducing the time between innovations

    , , ,
    Achal Patel

    Genentech, Inc., South San Francisco, CA 94080, USA

    ,
    David Veenstra

    Department of Pharmacy, University of Washington, Seattle, WA 98195, USA

    Curta, Inc., Seattle, WA 98116, USA

    ,
    Louis Garrison

    Department of Pharmacy, University of Washington, Seattle, WA 98195, USA

    VeriTech Corporation, Mercer Island, WA 98040, USA

    &
    Meng Li

    Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA

    Published Online:https://doi.org/10.2217/fon-2023-0514

    Abstract

    Aim: We investigated the effect of shortening time between innovations with the accelerated approval (AA) pathway on patient outcomes for three solid tumors. Methods: This real-world analysis evaluated patients receiving sequential AA pathway-approved innovations after initial treatment with existing therapies in three solid tumor case studies. Outcomes attributable to AA were estimated and assumed approval occurred at the time of conversion to approval and extrapolated to the US population. Results: Survival gains from accessing innovative therapies were 2.3–3.8-times higher when using the AA pathway. At the US population level, AA was associated with ∼8000 life-years gained across all three tumor case studies. Conclusion: In areas of rapid clinical development, the value of existing therapies can be enhanced by earlier access to AA pathway innovations and should be considered when evaluating the AA program.

    Plain language summary

    What is this study about?

    The US Food and Drug Administration's accelerated approval pathway provides patients with access to innovative drugs sooner than standard regulatory pathways. Using three case studies in solid tumors, this study measured how many patients on current cancer drugs received future cancer drugs because of the accelerated approval pathway and asked whether quicker access to new drugs resulted in them living longer.

    What were the results?

    In three cancer case studies, the accelerated approval pathway led to more patients receiving future cancer drugs. Patients who received future drugs through the AA pathway lived longer than patients who did not have access to them.

    What do the results of the study mean?

    The accelerated approval pathway is important because it can improve outcomes of current cancer drugs by giving patients additional treatments to choose from in the future and therefore a chance to live longer. Policymakers should consider this when thinking about making changes to the accelerated approval pathway.

    Advances in pharmaceutical innovation are a leading contributor to overall life expectancy gains in the USA [1]. Innovation in the oncology therapeutic arena has been particularly rapid with the number of targeted therapy new molecular entities (NME) approximately doubling during 2018–2022 compared with 2015–2017 [2]. These advances in oncology drug therapy have led to improved survival for patients with cancer [3,4]. Between 2000 and 2016, new cancer medicines were associated with nearly 1.3 million deaths avoided for the 15 most common tumors [5].

    In addition to the extended survival observed in the clinical trial setting, use of new cancer drugs in the real world may increase the chance that a patient remains alive to access future treatments still in clinical development. This concept of extending survival as a bridge to future, not yet approved, innovative therapies is also referred to as ‘real option value’ (ROV) and has been studied across different therapeutic areas. When the opportunity for patients to move from one life-extending treatment to a newly approved treatment is present, information on how this changes a patient's overall treatment trajectory and survival outcomes becomes increasingly important. Specifically, this information can impact how new treatments are valued by healthcare decision-makers and can influence treatment decision-making for oncologists [6–8].

    Prior studies in oncology have found that this increased access to future innovations may provide an additional 5–20% survival to patients beyond the survival gains expected from their current life-extending therapy [9–12]. For example, in advanced melanoma, patients who initiated treatment with first-line ipilimumab and survived to access pembrolizumab via its accelerated approval (AA) gained 35% more survival (3.7 months) than patients in the ipilimumab clinical trial [13]. However, the magnitude of survival benefit may vary depending on the pace of clinical innovation, with a higher likelihood of this additional survival value seen in diseases with rapid clinical innovation where more patients are likely to access future innovations.

    In the US market, one factor contributing to the pace of innovation has been the US FDA's AA program. The AA program is a regulatory pathway that allows the FDA more expeditiously to approve drugs that treat serious conditions with high unmet need based on a surrogate end point [14]. In recent years, the FDA has increasingly utilized the pathway to accelerate access to oncology products [15,16]. However, the AA pathway also has come under closer scrutiny given that not all medicines convert to traditional approval owing to withdrawals (i.e. olaratumab for soft tissue sarcoma), the cost of the medicines, as well as noted delays in completion of confirmatory trials for a handful of AA products [17–19]. Information on the net impact and value of the AA program in oncology is needed to better inform ongoing public discussion of the program's benefits and challenges [20]. Past research suggests that drugs approved through the AA program bring benefits based on modeled comparisons of health outcomes relative to standard of care, as well as analyses of aggregated clinical trial results [21,22]. However, there is no information on real-world outcomes for patients on life-extending therapies who access future drugs approved via the AA program.

    This study sought to examine the role of the AA pathway in enabling ROV. Specifically, we isolate the impact of the AA pathway in enabling patients on life-extending treatments to access newly approved treatments as a next line of therapy. The objectives of this analysis were: (i) to estimate the number of patients receiving future treatments and their survival gain due to the AA pathway using a real-world database, and (ii) to extrapolate the results to the US population to estimate the overall impact of the AA pathway on total life-years (LYs). Given most of the recent AAs have occurred in oncology, we use real-world data for treatments in three different tumors as case studies to study the impact of this pathway on clinical outcomes.

    Methods

    Study design

    In total, 36 previous AA indications were initially reviewed to assess their suitability as case studies for analysis, based on whether the AA drug was converted to a full approval and if the AA drug represented a potential follow-on line of care after a recent approval of a new drug in an earlier line of therapy. Case studies eligible for inclusion were further narrowed down based on the presence of an AA in the electronic health records (EHRs; cancer type had an AA between 2011 [date of data availability] and 2021), potential for ROV to be measured (cancer type had a newly approved ‘initial’ treatment in a previous line of therapy to the AA drug indication), data availability (cancer type available to the research team in the Flatiron Health database, availability of real-world progression-free survival data) and feasibility (sufficient sample size to conduct the analysis [i.e., rare tumors were excluded], sufficient follow-up time to estimate median progression-free and overall survival). Based on the selection criteria described above, metastatic melanoma, advanced bladder cancer and epidermal growth factor receptor positive (EGFR+) advanced non-small-cell lung cancer (aNSCLC) were considered eligible and selected as case studies. To assess real-world outcomes, we used the nationwide Flatiron Health EHR-derived longitudinal database, which is composed of de-identified patient-level structured and unstructured data, curated via technology-enabled abstraction [23,24]. We estimated patient-level outcomes for case studies in tumors with high levels of clinical innovation and for which recently approved products with extended survival could be eligible for future AA products within current survival estimates.

    We estimated outcomes associated with the AA pathway by comparing observed outcomes to a counterfactual (hypothetical) scenario in which approval occurred at a later time through a traditional approval pathway (Figure 1). For our primary analysis, we assumed that the traditional approval would occur at the date of conversion from accelerated approval per the FDA-reported date of traditional approval. Two other counterfactual scenario analyses were conducted to understand how differences in the timing of full approval, including potential earlier off-label use after data presentations or a longer time to complete a confirmatory trial, impacted findings. For these scenarios, the date of approval was either (i) the average time to full approval conversion in oncology overall (i.e., a longer time to approval than the actual dates for our case studies) or (ii) the date of the initial data presentation (i.e., phase III study) at a clinical conference (i.e., a shorter time to approval than the actual dates for our case studies).

    Figure 1. Study concept and design.

    (A) Depicts how the analysis isolates the impact of the AA pathway on patient survival. All patients start with the initial treatment (a new drug with extended survival) and then surviving patients progress to a subsequent line of therapy. As the AA drug represents the best overall survival, it is assumed that more patients who are alive when the drug is approved will use the AA drug, and the remainder of patients are distributed between the no treatment C and SoC B cohorts based on real-world trends in the real-word data. As the timing of the AA drug availability is delayed for the counterfactual scenario, the percentage of patients alive and eligible for the drug reduces. The analysis compares average patient outcomes across these two possible outcomes to isolate how the timing of the AA drug impacts patient outcomes. (B) Depicts how patients flow through initial and subsequent treatments across the three treatment cohorts (AA drug [A], SoC [B], no treatment [C]). Each cohort is followed up from the time of the initiation of the initial drug through subsequent treatments to estimate overall survival at the patient level before extrapolating patient-level outcomes to population estimates based on real-world market use of AA drugs during the AA period based on shares from publicly available epidemiology data.

    *Next-line SoC treatments for surviving patients in cohorts A and B may occur in pre-AA period.

    AA: Accelerated approval; SoC: Standard of care.

    To assess the proportions of patients receiving future AA drug(s) and survival outcomes we followed up patients starting treatment with a newly approved drug (referred to hereafter as the ‘initial drug’) in the three selected tumor types during the pre-AA period, defined as the period after their approval until the date of the AA of the future drug (Table 1). The initial drugs evaluated included first-line ipilimumab (metastatic melanoma), post-platinum cancer immunotherapies (advanced bladder cancer), and first- and second-generation EGFR tyrosine kinase inhibitors (TKIs) (EGFR+ aNSCLC; Table 1). Patient inclusion criteria included treatment within 120 days of their advanced diagnosis, use of the initial drug(s) during the pre AA period, prior platinum therapy (bladder cancer only) and an EGFR+ test result prior to treatment (EGFR+ aNSCLC only; Supplementary Table 1). Future AA drugs studied in the subsequent line of treatment included cancer immunotherapies (metastatic melanoma), enfortumab vedotin-ejfv (advanced bladder cancer) and osimertinib (EGFR+ aNSCLC). The AA dates for each of the above drugs, as well as the assumed counterfactual scenario dates, can be found in Table 1. For the cases where there were multiple drugs, the date for the drug with the earliest approval date was used as the start of the pre-AA period or start of the AA period.

    Table 1. Included drugs and key approval dates.
     Metastatic melanomaAdvanced bladder cancerEGFR+ aNSCLC
      New drug/indication1L ipilimumab2L+ post platinum immunotherapy§EGFR+ erlotinib, gefitinib or afatinib
      Traditional approval date28 March 201118 May 2016 (atezolizumab)14 May 2013 (erlotinib)
      Next innovation (AA product[s])Immunotherapy (pembrolizumab and nivolumab)Enfortumab vedotin-ejfvOsimertinib
      AA date4 September 2014 (pembrolizumab)18 December 201913 November 2015
    Counterfactual AA dates
      Date of data presentation19 April 201512 February 20216 December 2016
      Date of actual full approval18 December 2015 (pembrolizumab)9 July 202130 March 2017
      Average oncology conversion time4 March 201717 June 202213 May 2018
    Assumed delay in access to AA drug, months
      Date of data presentation7.614.113.0
      Date of conversion to full approval15.719.016.8
      Average oncology conversion time30.430.430.4

    †Average time for an oncology product approved through the AA pathway to convert to traditional approval, in months, based on oncology products with completed confirmatory trials per Center for Drug Evaluation and Research data on accelerated approvals; available at: www.fda.gov/drugs/nda-and-bla-approvals/accelerated-approvals.

    ‡For instances where there is more than one drug, we used the drug with the earliest approval dates.

    §Includes atezolizumab, nivolumab, durvalumab, avelumab and pembrolizumab.

    ¶Pembrolizumab data presented at AACR (metastatic melanoma), enforumab vedotin-ejfv data presented at ASCO GU (advanced bladder cancer) and osimertinib data published in The New England Journal of Medicine and presented at World Conference on Lung Cancer.

    1L: First-line; 2L+: Second-line and beyond; AA: Accelerated approval; AACR: American Association for Cancer Research; aNSCLC: Advanced non-small-cell lung cancer; ASCO: American Society of Clinical Oncology; EGFR+: Epidermal growth factor receptor positive; GU: Genitourinary.

    Outcomes

    Patient-level outcomes

    Outcomes measured from the real-world database included the proportion of patients: receiving the initial drug (by year of treatment initiation); surviving to the AA date for the future drug; and receiving either the future AA drug (treatment cohort A), standard of care (SoC) (treatment cohort B) or no subsequent treatment (treatment cohort C). For each of these cohorts, the mean overall survival (OS) was estimated. Given the limited sample size in metastatic melanoma, to ensure more stable survival estimates, we followed the methodology in a previous ROV analysis that expanded the patient pool for the survival analyses to include patients treated up until the date first-line pembrolizumab data were publicly presented [13], which may have initiated a change in SoC. For the treatment cohorts in which the initial drug was followed by the future AA drug or no subsequent treatment (cohorts A and C), mean OS was estimated from the time of initial drug initiation until the date of death. To estimate the OS for patients who received SoC after the initial drug, we constructed a survival curve by adding: the length of time from the start of the initial drug to the future AA drug initiation for each patient from cohort A; with mean time from SoC initiation to death from cohort B. This was done so that the OS differences between those receiving the future AA drug and SoC were not driven by differences in the length of time spent on the initial drug, but rather differences in OS from the future AA drug and SoC.

    As we sought to understand how the timing of future AA availability impacts outcomes for patients on treatments before the AA date, we estimated the average survival gain among all patients on the initial drug to understand the overall impact of the AA pathway. Specifically, this average estimate reflects the probability of remaining alive to receive the future AA drug as well as the additional survival provided by the initial AA drug.

    Statistical analysis

    Baseline patient characteristics for the three cohorts in each tumor were summarized using descriptive statistics and compared using χ2 test for categorical variables and the Kruskal–Wallis rank sum test for continuous variables. We generated Kaplan–Meier curves to estimate restricted mean OS for each cohort (A–C) in each tumor. Survival curves were also generated for patients within cohort B by line of therapy for which SoC therapy was initiated (for use in construction of survival curves for patients receiving SoC as described above). To mitigate potential confounding in comparison of survival estimates across cohorts, we weighted patients across cohorts using stabilized inverse probability of treatment weightings (IPTW). Weightings were constructed for propensity to use the future AA drug (cohort A) in relation to SoC (cohort B; for cohort A vs B comparison) and also for propensity to use the future AA drug (cohort A) in relation to no treatment (cohort C; for cohort A vs C comparison). Variables underlying the propensity calculations were age, sex, race and ethnicity, region, practice type (academic or community), payer type (available for bladder cancer and EGFR+ aNSCLC only), Eastern Cooperative Oncology Group (ECOG) performance status (available for EGFR+ aNSCLC and bladder cancer only), BRAF status (metastatic melanoma only) and type of cancer immunotherapy (CIT) used (pembrolizumab, atezolizumab or nivolumab/avelumab/durvalumab; for bladder cancer only). Payer type and ECOG performance status were not included in constructing IPTW for metastatic melanoma owing to a high level of missingness in the cohort. Statistical analyses and calculation of proportions were performed using R Statistical Software (v4.1.3; R Core Team 2022).

    Population-level outcomes

    Outcomes from the real-world data analysis were extrapolated to the population level by identifying the number of patients starting the initial drug in each year according to real-world observations of US population level estimates of drug utilization (Supplementary Table 2) [25]. The number of patients starting the initial drug at the population level was then multiplied by the proportion surviving to the AA date (estimated from the real-world database as described above) and categorized according to receipt of subsequent therapy to estimate the number and percentage of patients receiving the AA drug at the US population level. Outcomes attributable to the AA pathway were measured by estimating the change in the number and percentage of patients able to use the AA drug based on delays in access per the three counterfactual scenarios (primary analysis and two alternative scenarios) described above. The total LY impact at the population level was estimated by multiplying the additional number of patients by the average survival gain due to AA among all initial drug patients.

    Results

    Patient characteristics

    In total, 353 patients with metastatic melanoma, 888 patients with advanced bladder cancer and 734 patients with EGFR+ aNSCLC were included in the analysis (Table 2 & Supplementary Table 1). Patient characteristics by tumor and treatment received are shown in Supplementary Table 2. Baseline patient characteristics, including age, gender, race and ethnicity and region, were similar between cohorts within individual tumor types. Payer type was primarily Medicare, although fewer Medicare beneficiaries received future AA drugs in advanced bladder cancer and EGFR+ aNSCLC.

    Table 2. Real-world outcomes: proportion of patients receiving AA drugs and mean survival gains for patients.
     Metastatic melanomaAdvanced bladder cancerEGFR+ aNSCLC
    Proportion surviving and receiving AA drug
      Total number of initial drug-treated patients353888734
      Proportion of patients surviving to AA date for future drug, %492964
      Number of patients receiving future AA drug7965158
      Proportion of patients surviving and treated with future AA drug among all initial drug patients, %22722
    Survival associated with AA drug
      Mean survival of initial drug followed by AA drug, months56.237.657.1
      Mean survival of initial drug followed by SoC/no treatment, months24.921.332.8
      Difference in survival, months31.316.324.3
      Average survival gain among all new drug patients, months§7.01.25.2

    †Estimate reflects the weighted average survival between no treatment (cohort C) and SoC (cohort B), based on observed utilization patterns in Flatiron Health.

    ‡Estimate is the net difference in survival between initial drug patients who survived to move onto a sequential therapy, expressed as total survival for patients who received the future AA drug (cohort A) minus the weighted average of patients who did not receive the AA drug (cohorts B and C).

    §Estimate reflects the probability of receipt for the future AA drug based on the survival curve for the initial drug and is therefore weighted (downwards) by the likelihood of surviving to the future AA drug.

    AA: Accelerated approval; aNSCLC: Advanced non-small-cell lung cancer; EGFR+: Epidermal growth factor receptor positive; SoC: Standard of care.

    Receipt of AA drugs in the real-world database

    The proportion of patients on the initial drug surviving to the FDA AA date of the future drug was 49% (n = 173) in metastatic melanoma, 29% (n = 260) in advanced bladder cancer and 64% (n = 469) in EGFR+ aNSCLC (Table 2 & Supplementary Table 3). The proportion of patients who were subsequently treated with a future AA drug following treatment with an initial drug was 20% (70/353) in metastatic melanoma, 7% (65/888) in advanced bladder cancer and 22% (158/734) in EGFR+ aNSCLC. For bladder cancer, the median OS for the initial treatment was lower (37.6 months compared with 57.1 and 56.2 months for EGFR+ aNSCLC and metastatic melanoma, respectively), leading to a reduced likelihood that patients would remain alive to access new drugs for the next line of therapy.

    Mean survival gains of AA drugs in the real-world database

    The difference in mean survival for patients treated with the initial drug and then treated with the AA drug compared with SoC/no treatment was highest for the CITs in second-line and beyond metastatic melanoma (31.3 months), followed by osimertinib in EGFR+ aNSCLC (24.3 months), and third-line and beyond enfortumab vedotin-ejfv in advanced bladder cancer (16.3 months; Table 2 & Supplementary Figure 1). In patients treated with the initial drug, the average additional survival gain as a result of accelerated access to future drugs via the AA pathway was 7.0 months in metastatic melanoma, 5.2 months in EGFR+ aNSCLC and 1.2 months in advanced bladder cancer.

    Receipt of AA drugs in the US population

    After rescaling patient-level estimates from the real-world database to reflect the utilization of drugs within each tumor in the USA, the proportion of patients projected to survive to the AA date of the next innovation was 40% (n = 1743) in metastatic melanoma, 31% (n = 6635) in advanced bladder cancer and 62% (n = 12,646) in EGFR+ aNSCLC (Table 3 & Supplementary Table 3). The proportion and number of initial drug patients at the US population level treated with a subsequent future AA drug was 17% (745/4394) in metastatic melanoma, 8% (1698/21 488) in advanced bladder cancer and 21% (4233/20 317) in EGFR+ aNSCLC.

    Table 3. Population outcomes for AA pathway compared with counterfactual scenarios.
     Metastatic melanomaAdvanced bladder cancerEGFR+ aNSCLC
    Proportion surviving and receiving AA drug
      Total number of initial drug-treated patients439421,48820,317
      Proportion of patients surviving to AA date for future drug, %403162
      Number of patients receiving future AA drug74516984233
      Proportion of patients surviving and treated with future AA drug among all initial drug patients, %17821
      Average survival gain among all new drug patients, months5.31.35.1
    Primary analysis: traditional approval = date of AA conversion
      Number of additional patients receiving future AA drug owing to AA pathway52312472430
      Average survival gain among all initial drug patients due to AA pathway, months3.730.942.89
      Percentage of survival gain among all initial drug patients attributable to AA pathway, %707357
      Ratio of survival gain with AA pathway to traditional pathway3.43.82.3
      Population LYs gained with AA pathway136717024930
    Sensitivity analysis 1: shorter time to traditional approval
      Number of additional patients receiving future AA drug owing to AA pathway35510341942
      Average survival gain among all initial drug patients due to AA pathway, months2.530.782.31
      Percentage of survival gain among all initial drug patients attributable to AA pathway, %486146
      Ratio of survival gain with AA pathway to traditional pathway1.92.61.8
      Population LYs gained with AA pathway92714113934
    Sensitivity analysis 2: longer time to traditional approval
      Number of additional patients receiving future AA drug owing to AA pathway67815713473
      Average survival gain among all initial drug patients due to AA pathway, months4.841.194.15
      Percentage of survival gain among all initial drug patients attributable to AA pathway, %919282
      Ratio of survival gain with AA pathway to traditional pathway11.113.35.6
      Population LYs gained with AA pathway177221477075

    †Estimate reflects the probability of receipt for the future AA drug based on the survival curve for the initial drug, and is therefore weighted (downwards) by the likelihood of surviving to the future AA drug.

    AA: Accelerated approval; aNSCLC: Advanced non-small-cell lung cancer; EGFR+: Epidermal growth factor receptor positive; LY: Life-year.

    Mean survival gains attributable to the AA pathway in the US population

    In the primary counterfactual analysis, where the traditional approval for the AA drug would have occurred at the time of conversion from AA approval to full approval, the number of initial drug patients subsequently receiving the AA drug owing to the AA pathway in the USA was 523/745 in metastatic melanoma, 1247/1698 in advanced bladder cancer and 2430/4233 in EGFR+ aNSCLC (Table 3). The AA pathway was estimated to contribute 3.73 additional months of survival per patient among all patients who initiated ipilimumab in metastatic melanoma (in the pre-AA period), 0.85 months per patient among all patients initiating CITs post platinum therapy in advanced bladder cancer and 2.89 months per patient among all patients initiating first- and second-generation EGFR TKIs in EGFR+ aNSCLC. These survival gains attributable to the AA pathway were 70, 73 and 57% of the total average survival gains across all initial drug patients, respectively, and represent a 3.4-, 3.8- and 2.3-times increase in survival compared with the traditional approval pathway. Extrapolating the attributable survival gains to the US population resulted in an estimated 1367 (metastatic melanoma), 1702 (advanced bladder cancer) and 4930 (EGFR+ aNSCLC) LYs gained owing to the AA pathway.

    In scenario analyses, assuming a shorter time to traditional approval (i.e., date of clinical data presentation) resulted in survival gains attributable to the AA pathway ranging from 46 to 61% and LYs gained ranging from 827 to 38,934. In contrast, assuming a longer time to traditional approval (i.e., average oncology conversion time) increased the proportion of survival gains attributable to the AA pathway ranging from 82 to 92% and the LY gains ranging from 1772 to 7075.

    Outcomes associated with the AA pathway varied by the timing of the initial drug start. The number of patients receiving a future AA drug (as a result of the AA pathway) and the average additional survival gains due to the AA pathway was largest for patients who initiated their initial drug closest to the AA approval date (Figure 2A & B). The number of LYs gained owing to the AA pathway was also largest for those initiating the initial drug closer to the AA approval date (Figure 2C).

    Figure 2. Accelerated approval pathway outcomes.

    (A) Average additional survival (in months) per patient with and without AA pathway by proximity to the AA date; (B) number of patients receiving AA drug with and without AA pathway by proximity to the AA date; and (C) LYs gained for initial drug patients with AA pathway by proximity to the AA date.

    The calendar year prior to AA reflects the year of initial drug start relative to the date of the AA drug.

    AA: Accelerated approval; EGFR+: Epidermal growth factor receptor positive; LYs: Life years; N/A: Not applicable; NSCLC: Non-small-cell lung cancer.

    Discussion

    In this study using real-world data, we found that the AA pathway played a key role in enabling access to future AA drugs for patients who initiated a prior line of therapy in three case studies for solid tumors. Specifically, we found that the majority of the additional survival gains added to the initial drug from use of the future AA drug was attributable to the AA pathway, resulting in substantial LYs gained at a US population level. To our knowledge, this is the first study estimating the ROV of the AA pathway by measuring the real-world survival benefits of earlier access to oncology drugs among patients already treated with existing therapies.

    Our analysis offers a more complete understanding of the benefits and consequences of the AA program via evidence of a potential benefit of the AA pathway previously not considered. Previous research using modeling has suggested that drugs approved via expedited approval pathways have greater health outcome gains than drugs approved via traditional approval pathways [22]. Our study is complementary to these previous findings because our analyses demonstrate that the AA pathway can shorten the time between innovations thereby increasing the potential of a larger proportion of patients to access sequential innovative therapies to provide additional real-world survival gains. An important and differentiating point is that this additional survival value accrues to the initial drug – the life extension due to the initial drug is improved by the AA pathway for a future innovative drug. Given this observation, the ability of the AA pathway to shorten the time between innovations should be considered when deliberating potential modifications to the AA program.

    Considering that only a proportion of patients on the additional drug remained alive to access the future AA drug, the LYs gained estimates are substantial. For metastatic melanoma, 725 patients remained alive to access the future AA drug, leading to 1367 more population LYs gained over only 15.7 months (the conversion time for the primary analysis). Similarly, future AA drug access for 1698 patients led to 1702 LY gains in 19 months for advanced bladder cancer, and future AA drug access for 4233 patients led to 4930 LYs gained over 16.8 months in EFGR+ aNSCLC. As the AA pathway provides a one-time increase in early access, these population estimates of LYs gained translate into meaningful differences for individual patients facing limited survival on current treatment options. The combined effect of an initial treatment with extended survival, as well as the ability to increase access to future AA drug(s), led to OS estimates that were 1.6 (metastatic melanoma) to 2.7 (EGFR+ aNSCLC) times as long as survival for patients who did not have access to future AA drugs.

    While overall we found that the AA pathway contributes meaningfully to survival gains, there was variability in the magnitude of the benefit depending on the proximity in time of patients initiated on the initial drug therapy to the AA date. In general, those patients who initiated their initial drug closer to the AA date of the future drug had the most benefit from the AA pathway. This observation is driven by both the effectiveness of the AA drug as well as the severity of the tumor and suggests that patients with tumors with rapid disease progression may stand to benefit more from shortening the time between innovations via the AA pathway. This observation also highlights the importance of rapid innovation and facilitating access to these therapies in areas of high unmet need (such as limited survival) because delays could result in substantial LYs lost.

    This study has a number of limitations. First, the primary analysis was based on an assumption, namely that approval via a traditional pathway did not occur. We addressed this limitation by varying the date of traditional approval in sensitivity analyses to explore a range of real-world timings for conversion based on potential earlier use when positive data are presented at clinical conferences and potential later use based on average conversion time in oncology. Across these scenarios, we consistently found that the counterfactual scenario still resulted in LYs gained attributable to the AA pathway. Second, these results may not be generalizable to all tumors with AA drugs. The benefit of shortening the time between innovations and increasing the probability of patients benefiting from future therapies requires specific situations where innovations are both rapid and in subsequent lines of therapy, thus the value contribution of AA should be considered in this context. This potential benefit of the AA program is not present for all approvals and is much more likely to be relevant in areas of rapid innovation where a number of drugs are in clinical development for an indication. Further, the three AA case studies examined in this analysis were for drugs that converted to traditional approval, and our sample did not include AA drugs that were withdrawn. We therefore presented the impact of the AA pathway on patient survival and LYs for a subset of AA drugs. However, only about 10% of all products, and 8% of all oncology products, in the AA pathway have been withdrawn to date [26,27]. Additionally, other health outcomes, such as patients' quality of life and the cost impact of AA products on spending, were not measured in this analysis. Future research would be required to understand whether the benefit of bridging between innovations manifests in other health outcome measures. Finally, this analysis sought to isolate the impact of the AA program on ROV, or situations where patients on current life-extending therapies use newly approved medicines as a next line of treatment. This research topic was chosen given the demonstrated increasing importance of this information in drug value assessment and clinical decision-making [6–8]. Given this focus, our analysis and commentary did not comprehensively discuss the current debate on the benefits and challenges of the AA program [28,29].

    Conclusion

    The AA pathway in oncology can increase the likelihood for patients to benefit from future drugs by shortening the time between innovative therapies. While research has examined the impact of AA drugs on patients after AA approval date, our research suggests that survival gains may also be realized for patients who start on therapy in earlier lines of treatment before the AA approval date. Given the finding that a majority of the additional survival gains due to the AA drug was attributable to the AA program and its impact on earlier access, this aspect of value of the AA pathway should be addressed when considering future modifications to the FDA's AA program.

    Summary points
    • The accelerated approval (AA) pathway increased the number of patients already being treated with existing therapies who were able to access future subsequent treatments in three solid tumor case studies.

    • The AA drugs added additional survival to that of the initial drugs, with a majority of this additional survival being attributable to the AA pathway.

    • The additional survival provided by the AA pathway led to meaningful life-year gains at the US population level.

    • The results were driven by both the effectiveness of the AA drug as well as tumor severity and suggest that tumors with rapid disease progression may derive greater benefits from shortening the time between innovations via the AA pathway.

    • Policymakers should address the improved outcomes as a result of the earlier access due to the AA pathway when considering future modifications to the US FDA's AA program.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2023-0514

    Author contributions

    Concept and design: W Wong, S Kowal, D Veenstra, M Li, L Garrison. Acquisition, analysis or interpretation: W Wong, S Kowal, D Veenstra, M Li, L Garrison, T To, A Patel. Drafting or revising manuscript: W Wong, S Kowal, D Veenstra, M Li, L Garrison, T To, A Patel. Final approval of manuscript: W Wong, S Kowal, D Veenstra, M Li, L Garrison, T To, A Patel.

    Financial disclosure

    W Wong, T To, A Patel and S Kowal are employees of Genentech, Inc. and have stock in F Hoffmann-La Roche AG. D Veenstra, M Li and L Garrison received research funding for VeriTech Corporation from Genentech, Inc. This study was funded by Genentech, Inc. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    The authors have no competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    Writing disclosure

    Medical writing assistance was provided by S McKenna and R Hornby of Oxford PharmaGenesis, Oxford, UK, with funding from Genentech, Inc.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Previous presentations

    Presented at: ISPOR 2023, 7–10 May 2023, Boston, MA, USA [Poster HPR6] [30].

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Buxbaum JD, Chernew ME, Fendrick AM, Cutler DM. Contributions of public health, pharmaceuticals, and other medical care to US life expectancy changes, 1990–2015: study examines the conditions most responsible for changing us life expectancy and how public health, pharmaceuticals, other medical care, and other factors may have contributed to the changes. Health Aff. 39(9), 1546–1556 (2020).
    • 2. US Food and Drug Administration (FDA). New drugs at FDA: CDER's new molecular entities and new therapeutic biological products. www.fda.gov/drugs/development-approval-process-drugs/new-drugs-fda-cders-new-molecular-entities-and-new-therapeutic-biological-products (13 January 2023).
    • 3. Howlader N, Forjaz G, Mooradian MJ et al. The effect of advances in lung-cancer treatment on population mortality. N. Engl. J. Med. 383(7), 640–649 (2020).
    • 4. Li M, Goldman DP, Chen AJ. Spending on targeted therapies reduced mortality in patients with advanced-stage breast cancer: study examines spending on targeted therapies for patients with advanced-stage breast cancer and the impact on mortality. Health Aff. 40(5), 763–771 (2021).
    • 5. Macewan JP, Dennen S, Kee R, Ali F, Shafrin J, Batt K. Changes in mortality associated with cancer drug approvals in the United States from 2000 to 2016. J. Med. Econ. 23(12), 1558–1569 (2020).
    • 6. Li M, Garrison L Jr, Lee W, Kowal S, Wong W, Veenstra D. A pragmatic guide to assessing real option value for medical technologies. Value Health doi:10.1016/j.jval.2022.05.014 (2022). • Provides the framework for real option value used in this analysis.
    • 7. Lee W, Li M, Wong WB, To TM, Garrison LP, Veenstra DL. Modeling the ex post real option value in metastatic melanoma using real-world data. Value in Health 24(12), 1746–1753 (2021). • Example of a real option value analysis using real-world data.
    • 8. Li M, Elsisi Z, Wong WB, Kowal S, Veenstra DL, Garrison LP. Do future innovations in development influence oncologists' treatment recommendations today? A survey of U.S. oncologists. J. Clin. Oncol. 41(Suppl. 16), e24004–e24004 (2023).
    • 9. Lee W, Wong WB, Kowal S, Garrison LP Jr, Veenstra DL, Li M. Modeling the Ex Ante clinical real option value in an innovative therapeutic area: ALK-positive non-small-cell lung cancer. Pharmaco Economics 40(6), 623–631 (2022).
    • 10. Li M, Basu A, Bennette C, Veenstra D, Garrison LP Jr. How does option value affect the potential cost-effectiveness of a treatment? The case of ipilimumab for metastatic melanoma. Value Health 22(7), 777–784 (2019).
    • 11. Sanchez Y, Penrod JR, Qiu XL, Romley J, Thornton Snider J, Philipson T. The option value of innovative treatments in the context of chronic myeloid leukemia. Am. J. Manag. Care 18(Suppl. 11), S265–S271 (2012).
    • 12. Snider JT, Batt K, Wu Y, Tebeka MG, Seabury S. The option value of innovative treatments for non-small cell lung cancer and renal cell carcinoma. Am. J. Manag. Care 23, e340–e346 (2017).
    • 13. Wong WB, To TM, Li M, Lee W, Veenstra DL, Garrison LP Jr. Real-world evidence for option value in metastatic melanoma. J. Manag. Care Spec. Pharm. 27(11), 1546–1555 (2021).
    • 14. FDA. Accelerated approval program. www.fda.gov/drugs/nda-and-bla-approvals/accelerated-approval-program (13 January 2023).
    • 15. Beakes-Read G, Neisser M, Frey P, Guarducci M. Analysis of FDA's accelerated approval program performance December 1992-December 2021. Ther. Innov. Regul. Sci. 56(5), 698–703 (2022).
    • 16. Beaver JA, Howie LJ, Pelosof L et al. A 25-year experience of US Food and Drug Administration accelerated approval of malignant hematology and oncology drugs and biologics: a review. JAMA Oncol. 4(6), 849–856 (2018).
    • 17. Beaver JA, Pazdur R. “Dangling” accelerated approvals in oncology. N. Engl. J. Med. 384(18), e68 (2021).
    • 18. Kaltenboeck A, Mehlman A, Pearson SD. Potential policy reforms to strengthen the accelerated approval pathway. J. Comp. Eff. Res. 10(16), 1177–1186 (2021).
    • 19. Vokinger KN, Kesselheim AS, Glaus CEG, Hwang TJ. Therapeutic value of drugs granted accelerated approval or conditional marketing authorization in the US and Europe from 2007 to 2021. JAMA Health Forum. 3(8), e222685–e222685 (2022).
    • 20. Ladd V. The FDA's accelerated approval process is working (2021). https://morningconsult.com/opinions/fdas-accelerated-approval-process-working/ (13 January 2023).
    • 21. Gloy V, Schmitt AM, Düblin P et al. The evidence base of US Food and Drug Administration approvals of novel cancer therapies from 2000 to 2020. Int. J. Cancer 152(12), 2474–2484 (2023). • Previous research examining endpoints of accelerated approvals.
    • 22. Chambers JD, Thorat T, Wilkinson CL, Neumann PJ. Drugs cleared through the FDA's expedited review offer greater gains than drugs approved by conventional process. Health Aff. 36(8), 1408–1415 (2017). • Previous research examining outcomes of accelerated approval drugs.
    • 23. Birnbaum B, Nussbaum N, Seidl-Rathkopf K et al. Model-assisted cohort selection with bias analysis for generating large-scale cohorts from the EHR for oncology research. arXiv preprint arXiv:2001.09765 (2020).
    • 24. Ma X, Long L, Moon S, Adamson BJS, Baxi SS. Comparison of population characteristics in real-world clinical oncology databases in the US: Flatiron Health, SEER, and NPCR. medRxiv doi:10.1101/2020.03.16.20037143 (2020).
    • 25. CancerMPact® Patient metrics, Cerner Enviza. www.cancermpact.com (24 January 2023).
    • 26. Institute for Clinical and Economic Review. Strengthening the accelerated approval pathway: an analysis of potential policy reforms and their impact on uncertainty, access, innovation, and costs (2021). https://icer.org/wp-content/uploads/2021/04/Strengthening-the-Accelerated-Approval-Pathway-_-ICER-White-Paper-_-April-2021.pdf (22 February 2023).
    • 27. FOCR data on file presentation.
    • 28. Benjamin DJ, Lythgoe MP. The FDA's latest draft guidance on accelerated approvals – one step forward, two steps back? Nat. Rev. Clin. Oncol. 20(9), 577–578 (2023). • Review and commentary on the new US FDA guidance on accelerated approvals.
    • 29. Gyawali B, Kesselheim AS, Ross JS. The accelerated approval program for cancer drugs – finding the right balance. N. Engl. J. Med. 389(11), 968–971 (2023).
    • 30. Wong W, Kowal S, To TM et al. HPR6 beyond conventional clinical value: estimating the impact of accelerated approvals on therapeutic bridging between innovative therapies. Value in Health 26, S211–S212 (2023). www.ispor.org/docs/default-source/intl2023/gnea046-z-wong-42-x-72-l-postersolid-tumors-rweispor24apr23-pdf.pdf?sfvrsn=9b10f1b0_0