Bioequivalence of alternative pembrolizumab dosing regimens: current practice and future perspectives

IF 20.1 1区 医学 Q1 ONCOLOGY
Ruben Malmberg, Bram C. Agema, Maaike M. Hofman, Stefani Oosterveld, Sander Bins, Daphne W. Dumoulin, Arjen Joosse, Joachim G. J. V. Aerts, Reno Debets, Birgit C. P. Koch, Astrid A. M. van der Veldt, Roelof W. F. van Leeuwen, Ron H. J. Mathijssen
{"title":"Bioequivalence of alternative pembrolizumab dosing regimens: current practice and future perspectives","authors":"Ruben Malmberg,&nbsp;Bram C. Agema,&nbsp;Maaike M. Hofman,&nbsp;Stefani Oosterveld,&nbsp;Sander Bins,&nbsp;Daphne W. Dumoulin,&nbsp;Arjen Joosse,&nbsp;Joachim G. J. V. Aerts,&nbsp;Reno Debets,&nbsp;Birgit C. P. Koch,&nbsp;Astrid A. M. van der Veldt,&nbsp;Roelof W. F. van Leeuwen,&nbsp;Ron H. J. Mathijssen","doi":"10.1002/cac2.12661","DOIUrl":null,"url":null,"abstract":"<p>Immune checkpoint inhibitors (ICIs), including humanized anti-programmed cell death protein 1/programmed death-ligand 1 (anti-PD-1/PD-L1) monoclonal antibodies (mAbs), such as pembrolizumab, have transformed cancer treatment. Pembrolizumab was initially approved as a three-weekly (Q3W) 2 mg/kg weight-based dose by the Food and Drug Administration (FDA), which was later replaced by a 200 mg Q3W fixed-dose, mainly based on in silico simulations. Later, a fixed 400 mg six-weekly (Q6W) regimen was approved based on pharmacokinetic simulations [<span>1</span>].</p><p>Due to increasing ICI use and high costs per treatment, increasingly large portions of healthcare budgets are shifted to ICI treatment. Therefore, it is important to handle ICIs as efficiently and cost-effectively as possible. Hence, the Q6W regimen was introduced, lowering the total amount of ICI administrations per patient and the strain on healthcare capacity, and improving patient convenience. The cost-effectiveness of ICI treatment could be optimized further by using alternative dosing strategies (ADS) [<span>1</span>]. Some of these ADS have already been tested for pembrolizumab in some countries [<span>2, 3</span>].</p><p>Recently, the FDA provided bioequivalence guidelines for ADS, specifically for anti-PD-1/PD-L1 mAbs [<span>4</span>]. These guidelines support the use of pharmacokinetic-modeling to simulate steady-state trough concentrations (C<sub>trough,ss</sub>) and area under the curve (AUC; exposure) to establish bioequivalence for anti-PD-1/PD-L1 mAbs. In these guidelines, ADS are considered bioequivalent when both C<sub>trough,ss</sub> and exposure are at most 20% lower compared to the dosing regimen used while establishing efficacy in clinical trials (i.e. the reference dosing regimen). A maximum of 25% increase in the maximum concentration (C<sub>max</sub>) is used as the upper boundary, unless adequate clinical evidence shows that increasing the C<sub>max</sub> does not increase toxicity. For pembrolizumab, incidences of toxicities were generally consistent across a 2-10 mg/kg dose range in multiple trials [<span>5</span>].</p><p>To verify whether various ADS (Figure 1, Supplementary Table S1) are bioequivalent following the current FDA guidelines, we assessed bioequivalence using pharmacokinetic modelling. Using pharmacokinetic data from a real-world cohort, the best performing model was selected with which bioequivalence was assessed in extrapolations from this cohort (“extrapolation”) and also from a virtual (“simulation”) cohort. Details on both cohorts, as well as modelling procedures, are depicted in the Supplementary Material and Methods. As a reference, the Q3W 2 mg/kg dosing regimen was used, as this regimen was used in the original approval. Results of these analyses are shown in Figure 1. The simulated ADS for pembrolizumab given every 3 or 4 weeks (Q3/4W) were bioequivalent according to the FDA guidelines. Interestingly, despite meeting the criteria for exposure, none of the (non-registered) simulated Q6W ADS were bioequivalent, as the criteria for C<sub>trough,ss</sub> were not met.</p><p>Pharmacokinetic bioequivalence is usually utilized to ensure equivalent efficacy. However, the applicability of the thresholds proposed in the FDA guidelines could be questioned, as it disregards clinical pharmacological data regarding exposure-response relationships (except for C<sub>max</sub>). Consequently, the implications of non-adherence to the bioequivalence criteria are unclear. To explore the possible implications of non-adherence, we shall examine arguments in favor of and against the FDA-guidelines for bioequivalence of anti-PD-1/PD-L1 mAbs in the next section.</p><p>Firstly, the thresholds in these guidelines are derived from bioequivalence guidelines for small molecules. However, anti-PD-1/PD-L1 mAbs do not show similar exposure-response relationships to most small molecules. Therefore, similar anti-PD-1/PD-L1 mAb pharmacokinetics are probably not necessary to ensure similar efficacy. For example, no significant differences in efficacy were observed in a 2-10 mg/kg dose range [<span>5</span>]. Besides, no adequately powered prospective study has validated the FDA criteria for exposure (nor C<sub>trough,ss</sub>) [<span>5</span>]. This is exemplified during the registration of the pembrolizumab 400 mg Q6W dosing regimen [<span>5</span>]. The FDA initially rejected this regimen, as a pharmacokinetic model predicted that 400 mg Q6W was not bioequivalent to the 200 mg Q3W regimen [<span>5</span>]. Nonetheless, conditional approval was granted, based on a limited descriptive interim analysis (<i>n</i> = 44) of objective response rates in a non-randomized cohort, in which the response rates were compared with literature values [<span>5</span>].</p><p>Furthermore, clinical pharmacological data suggests that approved pembrolizumab doses are higher than required for maximum efficacy. This may be caused by a focus on the maximum tolerated dose in phase I trials, instead of the lowest maximally effective dose. For instance, it was demonstrated that doses ≥ 0.1 mg/kg result in near-complete (&gt; 95%) receptor occupancy in blood. Early research with nivolumab showed that maximum efficacy is achieved when near-complete receptor occupancy is reached [<span>1, 6</span>]. Nevertheless, only doses ≥ 2 mg/kg were selected for subsequent trials as simulations showed that this would result in near-complete intra-tumoral receptor occupancy after the first administration, which was considered a surrogate marker for maximum efficacy [<span>1</span>]. Two hundred mg Q3W results in a geometric mean of ≥ 10.7 µg/mL after the first dose. Consequently, the approved doses result in a much higher C<sub>trough,ss</sub> than required, which suggests that ADS resulting in C<sub>trough,ss</sub> levels ≥ 10.7 µg/mL (corresponding with a simulated dose of ≥ 0.85 mg/kg Q3W or ≥ 2.58 mg/kg Q6W, Supplementary Material and Methods) would also be maximally effective [<span>7</span>].</p><p>It has been reported that lower pembrolizumab exposure is associated with poorer outcomes, suggesting an exposure-response relationship. The <i>post-hoc</i> analyses of the Keynote-002 and Keynote-010 studies have shown however that worse clinical outcomes are associated with increased pembrolizumab clearance [<span>8</span>]. These findings could explain the absence of an exposure-response relationship, illustrated by similar overall survival, in the 2-10 mg/kg dose range despite substantial variation in C<sub>trough,ss</sub> (2 mg/kg Q3W (C<sub>trough,ss</sub> range: 1.13-127 µg/mL) [<span>1, 5</span>]. This underlines that bioequivalent exposure is not always indicative of outcome and could explain the reports of prolonged responses after treatment cessation during phase I-II trials [<span>1</span>].</p><p>Clinical evidence from various pembrolizumab trials in different tumor types also support non-bioequivalent doses which are lower than the EMA/FDA approved doses [<span>1</span>]. For instance, a retrospective study in patients with non-small cell lung cancer (NSCLC) found that administration of Q3W-Q6W ADS (<i>n</i> = 604) resulted in comparable overall survival as standard dosing (<i>n</i> = 1,362) [<span>2</span>]. Furthermore, three small Canadian studies retrospectively analyzed cohorts of NSCLC patients treated with 2 mg/kg Q3W and 4 mg/kg Q6W (combined <i>n</i> = 139) and concluded that 4 mg/kg Q6W was equally effective [<span>3</span>].</p><p>However, there is uncertainty in measuring receptor occupancy in vivo because this is a complex technical endeavor. Therefore, the intra-tumoral receptor occupancy studies were performed in silico using in vitro data and the developed model has never been validated in vivo. Consequently, its clinical predictive value remains unclear [<span>6</span>]. Moreover, even at near-complete receptor occupancy, attained by the 2 mg/kg dosing regimen under the most optimal conditions, only 15%-50% of patients with solid tumors respond [<span>5, 9</span>]. This indicates that the effectiveness of treatment is not solely dependent on receptor occupancy and pharmacokinetic parameters, suggesting a more complex array of non-pharmacokinetic factors influencing the response, such as tumor type, tumor mutational burden, the ability of T-cells to infiltrate and become activated within tumors, as well as PD-L1 expression and the expression of certain genomic alterations [<span>10</span>].</p><p>Randomized controlled trials (RCTs) are the golden standard for determining efficacy and safety of ADS. Currently, multiple studies are being conducted, illustrating the unmet need for ADS (NCT04913025, NCT04295863, NCT05692999, and NCT04909684) (detailed descriptions are depicted in Supplementary Table S2). However, presently, no adequately powered RCT to assess efficacy of doses &lt; 2 mg/kg has been completed yet, as this is time-consuming and expensive.</p><p>In conclusion, there are indications that the stipulated FDA criteria are not reflective of the true exposure-response relationship of pembrolizumab, thereby underlining the potential of ADS to improve cost-effectiveness of treatment. However, we still do not fully understand the driving mechanisms of ICI efficacy and the exact threshold for maximum efficacy remains elusive. Consequently, the implications of implementing ADS that are not bioequivalent to the reference dosing regimen in daily practice are not yet fully understood. Nevertheless, lower doses that deviate from bioequivalence criteria may result in exposures below the threshold for maximum efficacy, potentially leading to reduced efficacy. More sophisticated methods are needed to determine the minimum effectivity threshold in a faster way, and provide adequate guidance to support ADS for pembrolizumab. For future dose-optimization studies Tannock <i>et al.</i> [<span>11</span>] suggest moving away from the requirement for non-inferiority trials. Additionally, we recommend initiating ADS studies earlier, preferably prior to market reimbursement, to facilitate faster validation and regulatory acceptance of ADS. Until then, all available pembrolizumab dosing data should be carefully discussed, balanced, and reviewed.</p><p><b>Ruben Malmberg</b>: Methodology; investigation; validation; formal analysis; writing—original draft; writing—review &amp; editing, visualization. <b>Bram C. Agema</b>: Methodology; investigation; validation; formal analysis; writing—original draft; writing—review &amp; editing; visualization. <b>Maaike M. Hofman</b>: Methodology; investigation; validation; formal analysis; writing—original draft; writing—review &amp; editing; visualization. <b>Stefani Oosterveld</b>: Investigation; writing—review &amp; editing. <b>Sander Bins</b>: Methodology; writing—review &amp; editing. <b>Daphne W. Dumoulin</b>: Methodology; writing—review &amp; editing. <b>Arjen Joosse</b>: Methodology; writing—review &amp; editing. <b>Joachim G.J.V Aerts</b>: Methodology; writing—review &amp; editing. <b>Reno Debets</b>: Methodology; writing—review &amp; editing. <b>Birgit C.P. Koch</b>: Methodology; writing—review &amp; editing. <b>Astrid A.M. van der Veldt</b>: Methodology; writing—review &amp; editing. <b>Roelof W.F. van Leeuwen</b>: Conceptualization; methodology; investigation; writing—review &amp; editing. <b>Ron H.J. Mathijssen</b>: Conceptualization; methodology; resources; investigation; writing—review &amp; editing.</p><p>None of the co-authors have a conflict of interest in relation to the current manuscript. Ruben Malmberg reports speaker fees from Bristol Myers Squibb. Daphne W. Dumoulin reports speaker fees and consultancy fees from Roche, Bristol Myers Squibb, Merck Sharp &amp; Dohme, Astra Zeneca, Pfizer, and Amgen. Joachim G.J.V. Aerts reports grants from Boehringer-Ingelheim, Merck Sharp &amp; Dohme, Bristol Myers Squibb, Astra-Zeneca, Eli-Lilly, Verastem, Nutricia, Amphera, and CureVac (payments to institution and personnel). Fees for lectures: Merck Sharp &amp;Dohme, Astra-Zeneca, and Eli-Lilly. Travel fees: Merck Sharp &amp; Dohme and Astra Zeneca. Patent issued: allogeneic tumor cell lysate, combination immunotherapy. Board of directors’ member for International Association for the Study of Lung Cancer and International Mesothelioma Interest Group. Stock: Amphera. Astrid A.M. van der Veldt reports consultancy fees from Bristol Myers Squibb, Eisai, Ipsen, Merck Sharp &amp; Dohme, Sanofi, Pierre Fabre, Novartis, Pfizer, and Roche (payments to institution). Roelof W.F. van Leeuwen reports research grants from Bristol Myers Squibb and Pfizer. Consulting fees from Bristol Myers Squibb, Pfizer, Pierre-Fabre and Merck Sharp &amp; Dohme. Ron H.J. Mathijssen reports research grants (payments to institution) from Astellas, Bayer, Boehringer-Ingelheim, Cristal Therapeutics, Deuter Oncology, Echo Pharmaceuticals, Nordic Pharma, Novartis, Pamgene, Pfizer, Roche, Sanofi and Servier.</p><p>This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.</p><p>This study was conducted in accordance with the Declaration of Helsinki and was approved by the local ethics committee of the Erasmus Medical Center (METC 16-011). All participants provided written informed consent prior to their inclusion in the study.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"45 4","pages":"471-475"},"PeriodicalIF":20.1000,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12661","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12661","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Immune checkpoint inhibitors (ICIs), including humanized anti-programmed cell death protein 1/programmed death-ligand 1 (anti-PD-1/PD-L1) monoclonal antibodies (mAbs), such as pembrolizumab, have transformed cancer treatment. Pembrolizumab was initially approved as a three-weekly (Q3W) 2 mg/kg weight-based dose by the Food and Drug Administration (FDA), which was later replaced by a 200 mg Q3W fixed-dose, mainly based on in silico simulations. Later, a fixed 400 mg six-weekly (Q6W) regimen was approved based on pharmacokinetic simulations [1].

Due to increasing ICI use and high costs per treatment, increasingly large portions of healthcare budgets are shifted to ICI treatment. Therefore, it is important to handle ICIs as efficiently and cost-effectively as possible. Hence, the Q6W regimen was introduced, lowering the total amount of ICI administrations per patient and the strain on healthcare capacity, and improving patient convenience. The cost-effectiveness of ICI treatment could be optimized further by using alternative dosing strategies (ADS) [1]. Some of these ADS have already been tested for pembrolizumab in some countries [2, 3].

Recently, the FDA provided bioequivalence guidelines for ADS, specifically for anti-PD-1/PD-L1 mAbs [4]. These guidelines support the use of pharmacokinetic-modeling to simulate steady-state trough concentrations (Ctrough,ss) and area under the curve (AUC; exposure) to establish bioequivalence for anti-PD-1/PD-L1 mAbs. In these guidelines, ADS are considered bioequivalent when both Ctrough,ss and exposure are at most 20% lower compared to the dosing regimen used while establishing efficacy in clinical trials (i.e. the reference dosing regimen). A maximum of 25% increase in the maximum concentration (Cmax) is used as the upper boundary, unless adequate clinical evidence shows that increasing the Cmax does not increase toxicity. For pembrolizumab, incidences of toxicities were generally consistent across a 2-10 mg/kg dose range in multiple trials [5].

To verify whether various ADS (Figure 1, Supplementary Table S1) are bioequivalent following the current FDA guidelines, we assessed bioequivalence using pharmacokinetic modelling. Using pharmacokinetic data from a real-world cohort, the best performing model was selected with which bioequivalence was assessed in extrapolations from this cohort (“extrapolation”) and also from a virtual (“simulation”) cohort. Details on both cohorts, as well as modelling procedures, are depicted in the Supplementary Material and Methods. As a reference, the Q3W 2 mg/kg dosing regimen was used, as this regimen was used in the original approval. Results of these analyses are shown in Figure 1. The simulated ADS for pembrolizumab given every 3 or 4 weeks (Q3/4W) were bioequivalent according to the FDA guidelines. Interestingly, despite meeting the criteria for exposure, none of the (non-registered) simulated Q6W ADS were bioequivalent, as the criteria for Ctrough,ss were not met.

Pharmacokinetic bioequivalence is usually utilized to ensure equivalent efficacy. However, the applicability of the thresholds proposed in the FDA guidelines could be questioned, as it disregards clinical pharmacological data regarding exposure-response relationships (except for Cmax). Consequently, the implications of non-adherence to the bioequivalence criteria are unclear. To explore the possible implications of non-adherence, we shall examine arguments in favor of and against the FDA-guidelines for bioequivalence of anti-PD-1/PD-L1 mAbs in the next section.

Firstly, the thresholds in these guidelines are derived from bioequivalence guidelines for small molecules. However, anti-PD-1/PD-L1 mAbs do not show similar exposure-response relationships to most small molecules. Therefore, similar anti-PD-1/PD-L1 mAb pharmacokinetics are probably not necessary to ensure similar efficacy. For example, no significant differences in efficacy were observed in a 2-10 mg/kg dose range [5]. Besides, no adequately powered prospective study has validated the FDA criteria for exposure (nor Ctrough,ss) [5]. This is exemplified during the registration of the pembrolizumab 400 mg Q6W dosing regimen [5]. The FDA initially rejected this regimen, as a pharmacokinetic model predicted that 400 mg Q6W was not bioequivalent to the 200 mg Q3W regimen [5]. Nonetheless, conditional approval was granted, based on a limited descriptive interim analysis (n = 44) of objective response rates in a non-randomized cohort, in which the response rates were compared with literature values [5].

Furthermore, clinical pharmacological data suggests that approved pembrolizumab doses are higher than required for maximum efficacy. This may be caused by a focus on the maximum tolerated dose in phase I trials, instead of the lowest maximally effective dose. For instance, it was demonstrated that doses ≥ 0.1 mg/kg result in near-complete (> 95%) receptor occupancy in blood. Early research with nivolumab showed that maximum efficacy is achieved when near-complete receptor occupancy is reached [1, 6]. Nevertheless, only doses ≥ 2 mg/kg were selected for subsequent trials as simulations showed that this would result in near-complete intra-tumoral receptor occupancy after the first administration, which was considered a surrogate marker for maximum efficacy [1]. Two hundred mg Q3W results in a geometric mean of ≥ 10.7 µg/mL after the first dose. Consequently, the approved doses result in a much higher Ctrough,ss than required, which suggests that ADS resulting in Ctrough,ss levels ≥ 10.7 µg/mL (corresponding with a simulated dose of ≥ 0.85 mg/kg Q3W or ≥ 2.58 mg/kg Q6W, Supplementary Material and Methods) would also be maximally effective [7].

It has been reported that lower pembrolizumab exposure is associated with poorer outcomes, suggesting an exposure-response relationship. The post-hoc analyses of the Keynote-002 and Keynote-010 studies have shown however that worse clinical outcomes are associated with increased pembrolizumab clearance [8]. These findings could explain the absence of an exposure-response relationship, illustrated by similar overall survival, in the 2-10 mg/kg dose range despite substantial variation in Ctrough,ss (2 mg/kg Q3W (Ctrough,ss range: 1.13-127 µg/mL) [1, 5]. This underlines that bioequivalent exposure is not always indicative of outcome and could explain the reports of prolonged responses after treatment cessation during phase I-II trials [1].

Clinical evidence from various pembrolizumab trials in different tumor types also support non-bioequivalent doses which are lower than the EMA/FDA approved doses [1]. For instance, a retrospective study in patients with non-small cell lung cancer (NSCLC) found that administration of Q3W-Q6W ADS (n = 604) resulted in comparable overall survival as standard dosing (n = 1,362) [2]. Furthermore, three small Canadian studies retrospectively analyzed cohorts of NSCLC patients treated with 2 mg/kg Q3W and 4 mg/kg Q6W (combined n = 139) and concluded that 4 mg/kg Q6W was equally effective [3].

However, there is uncertainty in measuring receptor occupancy in vivo because this is a complex technical endeavor. Therefore, the intra-tumoral receptor occupancy studies were performed in silico using in vitro data and the developed model has never been validated in vivo. Consequently, its clinical predictive value remains unclear [6]. Moreover, even at near-complete receptor occupancy, attained by the 2 mg/kg dosing regimen under the most optimal conditions, only 15%-50% of patients with solid tumors respond [5, 9]. This indicates that the effectiveness of treatment is not solely dependent on receptor occupancy and pharmacokinetic parameters, suggesting a more complex array of non-pharmacokinetic factors influencing the response, such as tumor type, tumor mutational burden, the ability of T-cells to infiltrate and become activated within tumors, as well as PD-L1 expression and the expression of certain genomic alterations [10].

Randomized controlled trials (RCTs) are the golden standard for determining efficacy and safety of ADS. Currently, multiple studies are being conducted, illustrating the unmet need for ADS (NCT04913025, NCT04295863, NCT05692999, and NCT04909684) (detailed descriptions are depicted in Supplementary Table S2). However, presently, no adequately powered RCT to assess efficacy of doses < 2 mg/kg has been completed yet, as this is time-consuming and expensive.

In conclusion, there are indications that the stipulated FDA criteria are not reflective of the true exposure-response relationship of pembrolizumab, thereby underlining the potential of ADS to improve cost-effectiveness of treatment. However, we still do not fully understand the driving mechanisms of ICI efficacy and the exact threshold for maximum efficacy remains elusive. Consequently, the implications of implementing ADS that are not bioequivalent to the reference dosing regimen in daily practice are not yet fully understood. Nevertheless, lower doses that deviate from bioequivalence criteria may result in exposures below the threshold for maximum efficacy, potentially leading to reduced efficacy. More sophisticated methods are needed to determine the minimum effectivity threshold in a faster way, and provide adequate guidance to support ADS for pembrolizumab. For future dose-optimization studies Tannock et al. [11] suggest moving away from the requirement for non-inferiority trials. Additionally, we recommend initiating ADS studies earlier, preferably prior to market reimbursement, to facilitate faster validation and regulatory acceptance of ADS. Until then, all available pembrolizumab dosing data should be carefully discussed, balanced, and reviewed.

Ruben Malmberg: Methodology; investigation; validation; formal analysis; writing—original draft; writing—review & editing, visualization. Bram C. Agema: Methodology; investigation; validation; formal analysis; writing—original draft; writing—review & editing; visualization. Maaike M. Hofman: Methodology; investigation; validation; formal analysis; writing—original draft; writing—review & editing; visualization. Stefani Oosterveld: Investigation; writing—review & editing. Sander Bins: Methodology; writing—review & editing. Daphne W. Dumoulin: Methodology; writing—review & editing. Arjen Joosse: Methodology; writing—review & editing. Joachim G.J.V Aerts: Methodology; writing—review & editing. Reno Debets: Methodology; writing—review & editing. Birgit C.P. Koch: Methodology; writing—review & editing. Astrid A.M. van der Veldt: Methodology; writing—review & editing. Roelof W.F. van Leeuwen: Conceptualization; methodology; investigation; writing—review & editing. Ron H.J. Mathijssen: Conceptualization; methodology; resources; investigation; writing—review & editing.

None of the co-authors have a conflict of interest in relation to the current manuscript. Ruben Malmberg reports speaker fees from Bristol Myers Squibb. Daphne W. Dumoulin reports speaker fees and consultancy fees from Roche, Bristol Myers Squibb, Merck Sharp & Dohme, Astra Zeneca, Pfizer, and Amgen. Joachim G.J.V. Aerts reports grants from Boehringer-Ingelheim, Merck Sharp & Dohme, Bristol Myers Squibb, Astra-Zeneca, Eli-Lilly, Verastem, Nutricia, Amphera, and CureVac (payments to institution and personnel). Fees for lectures: Merck Sharp &Dohme, Astra-Zeneca, and Eli-Lilly. Travel fees: Merck Sharp & Dohme and Astra Zeneca. Patent issued: allogeneic tumor cell lysate, combination immunotherapy. Board of directors’ member for International Association for the Study of Lung Cancer and International Mesothelioma Interest Group. Stock: Amphera. Astrid A.M. van der Veldt reports consultancy fees from Bristol Myers Squibb, Eisai, Ipsen, Merck Sharp & Dohme, Sanofi, Pierre Fabre, Novartis, Pfizer, and Roche (payments to institution). Roelof W.F. van Leeuwen reports research grants from Bristol Myers Squibb and Pfizer. Consulting fees from Bristol Myers Squibb, Pfizer, Pierre-Fabre and Merck Sharp & Dohme. Ron H.J. Mathijssen reports research grants (payments to institution) from Astellas, Bayer, Boehringer-Ingelheim, Cristal Therapeutics, Deuter Oncology, Echo Pharmaceuticals, Nordic Pharma, Novartis, Pamgene, Pfizer, Roche, Sanofi and Servier.

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

This study was conducted in accordance with the Declaration of Helsinki and was approved by the local ethics committee of the Erasmus Medical Center (METC 16-011). All participants provided written informed consent prior to their inclusion in the study.

Abstract Image

替代性派姆单抗给药方案的生物等效性:当前实践和未来展望
免疫检查点抑制剂(ICIs),包括人源化抗程序性细胞死亡蛋白1/程序性死亡配体1(抗pd -1/PD-L1)单克隆抗体(mab),如派姆单抗,已经改变了癌症治疗。Pembrolizumab最初被美国食品和药物管理局(FDA)批准为3周(Q3W) 2 mg/kg基于体重的剂量,后来被200 mg Q3W固定剂量所取代,主要基于计算机模拟。随后,基于药代动力学模拟[1],一个固定的400mg 6周(Q6W)方案被批准。由于ICI使用的增加和每次治疗的高成本,越来越多的医疗预算转移到ICI治疗上。因此,尽可能高效和经济地处理ici非常重要。因此,引入了Q6W方案,降低了每位患者的ICI管理总量和医疗保健能力的压力,并改善了患者的便利性。采用替代给药策略(ADS)可以进一步优化ICI治疗的成本效益。其中一些ADS已经在一些国家进行了派姆单抗测试[2,3]。最近,FDA提供了ADS的生物等效性指南,特别是抗pd -1/PD-L1单克隆抗体[4]。这些指南支持使用药代动力学模型来模拟稳态波谷浓度(Ctrough,ss)和曲线下面积(AUC;暴露)以建立抗pd -1/PD-L1单克隆抗体的生物等效性。在这些指南中,与临床试验中确定疗效时所使用的给药方案(即参考给药方案)相比,当Ctrough、ss和暴露量最多降低20%时,ADS被认为具有生物等效性。最大浓度(Cmax)最大增加25%作为上限,除非有足够的临床证据表明增加Cmax不会增加毒性。对于派姆单抗,在多个试验中,2- 10mg /kg剂量范围内的毒性发生率通常是一致的。为了验证不同的ADS(图1,补充表S1)是否遵循FDA现行指南具有生物等效性,我们使用药代动力学模型评估了生物等效性。使用来自真实世界队列的药代动力学数据,选择了表现最佳的模型,并在该队列(“外推”)和虚拟(“模拟”)队列的外推中评估生物等效性。关于这两个队列的详细信息,以及建模程序,在补充材料和方法中描述。作为参考,使用了Q3W 2mg /kg给药方案,因为该方案在最初的批准中使用。分析结果如图1所示。根据FDA指南,每3或4周(Q3/4W)给予派姆单抗的模拟ADS具有生物等效性。有趣的是,尽管符合暴露标准,但没有(未注册的)模拟Q6W ADS具有生物等效性,因为不符合cough,ss的标准。通常采用药代动力学生物等效性来保证药效等效。然而,FDA指南中提出的阈值的适用性可能会受到质疑,因为它忽略了有关暴露-反应关系的临床药理学数据(Cmax除外)。因此,不遵守生物等效性标准的含义尚不清楚。为了探讨不依从性的可能影响,我们将在下一节中研究支持和反对fda抗pd -1/PD-L1单克隆抗体生物等效性指南的论点。首先,这些指南中的阈值来源于小分子生物等效性指南。然而,抗pd -1/PD-L1单克隆抗体对大多数小分子并不表现出类似的暴露-反应关系。因此,相似的抗pd -1/PD-L1单抗药代动力学可能不是保证相似疗效的必要条件。例如,在2-10 mg/kg剂量范围内,没有观察到疗效的显著差异。此外,没有足够有力的前瞻性研究证实了FDA的暴露标准(也不是cough,ss)。这在pembrolizumab 400mg Q6W给药方案[5]的注册过程中得到了例证。FDA最初拒绝了该方案,因为药代动力学模型预测400mg Q6W与200mg Q3W方案不具有生物等效性。尽管如此,基于一项有限的描述性中期分析(n = 44),在一个非随机队列中,客观反应率获得了有条件的批准,其中反应率与文献值[5]进行了比较。此外,临床药理学数据表明,批准的派姆单抗剂量高于达到最大疗效所需的剂量。这可能是由于在I期试验中关注的是最大耐受剂量,而不是最低的最大有效剂量。例如,已证明剂量≥0.1 mg/kg可导致近乎完全的(&gt;95%)受体在血液中的占用。 早期对纳武单抗的研究表明,当受体几乎完全占据时,达到最大疗效[1,6]。然而,后续试验只选择剂量≥2mg /kg,因为模拟表明,这将导致首次给药后几乎完全占据肿瘤内受体,这被认为是最大疗效[1]的替代标志。第一次给药后,200 mg Q3W的几何平均值≥10.7µg/mL。因此,批准剂量导致的穿透率ss远高于要求,这表明ADS导致的穿透率ss水平≥10.7µg/mL(对应于模拟剂量≥0.85 mg/kg Q3W或≥2.58 mg/kg Q6W,补充材料和方法)也将是最大有效的[7]。据报道,较低的派姆单抗暴露与较差的预后相关,表明暴露-反应关系。然而,Keynote-002和Keynote-010研究的事后分析表明,更差的临床结果与派姆单抗清除率增加有关。这些发现可以解释在2-10 mg/kg剂量范围内,尽管Ctrough,ss (2 mg/kg Q3W (Ctrough,ss范围:1.13-127µg/mL)有很大变化,但总体生存率相似,没有暴露-反应关系[1,5]。这强调了生物等效暴露并不总是表明结果,并且可以解释在I-II期试验中停止治疗后反应延长的报告[10]。来自不同肿瘤类型的各种派姆单抗试验的临床证据也支持非生物等效剂量,其剂量低于EMA/FDA批准的剂量。例如,一项针对非小细胞肺癌(NSCLC)患者的回顾性研究发现,给予Q3W-Q6W ADS (n = 604)的总生存率与标准剂量(n = 1362)相当。此外,加拿大的三项小型研究回顾性分析了2mg /kg Q3W和4mg /kg Q6W治疗的NSCLC患者队列(合并n = 139),得出4mg /kg Q6W同样有效的结论。然而,在体内测量受体占用存在不确定性,因为这是一项复杂的技术努力。因此,肿瘤内受体占用研究是使用体外数据在计算机上进行的,所开发的模型从未在体内得到验证。因此,其临床预测价值尚不清楚。此外,即使在最优条件下,通过2mg /kg给药方案获得接近完全的受体占用,也只有15%-50%的实体瘤患者有反应[5,9]。这表明治疗的有效性并不仅仅取决于受体占用和药代动力学参数,表明影响疗效的非药代动力学因素更为复杂,如肿瘤类型、肿瘤突变负担、t细胞浸润和肿瘤内活化的能力,以及PD-L1表达和某些基因组改变[10]的表达。随机对照试验(RCTs)是确定ADS疗效和安全性的黄金标准。目前,正在进行的多项研究表明,ADS (NCT04913025、NCT04295863、NCT05692999和NCT04909684)的需求尚未得到满足(详细描述见补充表S2)。然而,目前还没有足够有力的随机对照试验来评估剂量的疗效。目前还没有完成2毫克/公斤,因为这既耗时又昂贵。总之,有迹象表明,FDA规定的标准并不能反映派姆单抗的真实暴露-反应关系,从而强调了ADS提高治疗成本效益的潜力。然而,我们仍然不完全了解ICI疗效的驱动机制,最大疗效的确切阈值仍然难以捉摸。因此,在日常实践中实施与参考给药方案不具有生物等效性的ADS的影响尚未完全了解。然而,偏离生物等效性标准的较低剂量可能导致暴露低于最大效力阈值,从而可能导致效力降低。需要更复杂的方法以更快的方式确定最小有效阈值,并提供足够的指导来支持派姆单抗的ADS。对于未来的剂量优化研究,Tannock等人建议放弃对非劣效性试验的要求。此外,我们建议更早地启动ADS研究,最好是在市场报销之前,以促进ADS的更快验证和监管接受。在此之前,所有可用的派姆单抗剂量数据应仔细讨论、平衡和审查。Ruben Malmberg:方法论;调查;验证;正式的分析;原创作品草案;writing-review,编辑、可视化。布拉姆C。 Agema:方法论;调查;验证;正式的分析;原创作品草案;writing-review,编辑;可视化。马克·霍夫曼:方法论;调查;验证;正式的分析;原创作品草案;writing-review,编辑;可视化。Stefani Oosterveld:调查;writing-review,编辑。Sander Bins:方法论;writing-review,编辑。Daphne W. Dumoulin:方法论;writing-review,编辑。Arjen Joosse:方法论;writing-review,编辑。Joachim g.j.v. Aerts:方法论;writing-review,编辑。雷诺债务:方法论;writing-review,编辑。Birgit C.P. Koch:方法论;writing-review,编辑。Astrid A.M. van der Veldt:方法论;writing-review,编辑。Roelof W.F. van Leeuwen:概念化;方法;调查;writing-review,编辑。Ron H.J. Mathijssen:概念化;方法;资源;调查;writing-review,编辑。所有共同作者都没有与当前稿件相关的利益冲突。Ruben Malmberg报道Bristol Myers Squibb的演讲费用。Daphne W. Dumoulin报告了罗氏(Roche)、百时美施贵宝(Bristol Myers Squibb)、默克夏普(Merck Sharp &amp)的演讲费和咨询费。杜邦、阿斯利康、辉瑞和安进。Joachim G.J.V. Aerts报道勃林格-殷格翰、默克夏普和安培;Dohme、Bristol Myers Squibb、Astra-Zeneca、Eli-Lilly、Verastem、Nutricia、Amphera和CureVac(支付给机构和人员)。讲座费用:默沙东、阿斯利康和礼来。差旅费:Merck Sharp &amp;多芬和阿斯利康。专利:异体肿瘤细胞裂解液,联合免疫治疗。国际肺癌研究协会和国际间皮瘤兴趣小组董事会成员。股票:Amphera。Astrid A.M. van der Veldt报告了Bristol Myers Squibb、卫材、Ipsen、Merck Sharp &amp;多美、赛诺菲、皮尔法伯、诺华、辉瑞和罗氏(支付给机构)。Roelof W.F. van Leeuwen报道了Bristol Myers Squibb和Pfizer的研究资助。Bristol Myers Squibb、Pfizer、Pierre-Fabre和Merck Sharp &amp的咨询费;Dohme。Ron H.J. Mathijssen报告了来自安斯泰来、拜耳、勃林格殷格翰、crystal Therapeutics、Deuter Oncology、Echo Pharmaceuticals、Nordic Pharma、诺华、Pamgene、辉瑞、罗氏、赛诺菲和施维雅的研究资助(支付给机构)。这项研究没有从公共、商业或非营利部门的资助机构获得任何具体的资助。本研究是根据赫尔辛基宣言进行的,并得到了伊拉斯谟医学中心当地伦理委员会(METC 16-011)的批准。所有参与者在纳入研究之前都提供了书面知情同意书。
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 求助全文
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信