PBPK 是否有助于为产品标签提供信息,以管理由细胞因子释放综合征介导的临床重大药物相互作用?

IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY
Xinyuan Zhang, Ping Zhao
{"title":"PBPK 是否有助于为产品标签提供信息,以管理由细胞因子释放综合征介导的临床重大药物相互作用?","authors":"Xinyuan Zhang,&nbsp;Ping Zhao","doi":"10.1002/psp4.13185","DOIUrl":null,"url":null,"abstract":"<p>Evaluating drug interactions caused by cytokine release syndrome (CRS) with PBPK (Physiologically Based Pharmacokinetic) modeling has been reported in some bispecific antibody regulatory submissions for 10 years. However, the published regulatory reviews and sponsors' analyses seem to disagree on the roles of PBPK modeling in regulatory decision-making. In this editorial, we reviewed and provided our opinions on the FDA's current practice and sponsors' position in evaluating CRS-mediated drug interactions. We discussed what has been done and what is lacking in the current PBPK approach assessing the CRS-mediated drug interactions and proposed areas to bridge the gaps. And finally, we call to actions to improve the current practice toward a patient-centric clinical pharmacology approach with more quantitative assessment and management of CRS-mediated drug interactions.</p><p>The manuscript by Willemin et al.<span><sup>1</sup></span> described the use of a PBPK approach to evaluate the effect of elevated IL-6 following the treatment of teclistamab on the PK of CYP enzyme (1A2, 2C9, 2C19, 3A4, 3A5) substrates. This marks the 4th PBPK publication by CPT-PSP of the effect of CRS as a result of biologics-treatment on co-medications that are CYP substrates, after blinatumomab,<span><sup>2</sup></span> mosunetuzumab,<span><sup>3</sup></span> and glofitamab.<span><sup>4</sup></span> The scientific community and drug developers are using the PBPK modeling tool to study the effect of CRS on the PK and safety of co-administered CYP substrate drugs. However, there seems to be a gap between the peer-reviewed papers<span><sup>1-4</sup></span> and the regulatory evaluations<span><sup>5-8</sup></span> in terms of concluding the impact of PBPK predictions. In this editorial, we examine the gap and share our opinions on the value, expectation, and future of PBPK modeling in this specific area with the aim of increasing awareness, calling for enhanced predictive performance, and ultimately, achieving patient-centric clinical pharmacology.</p><p>Cytokine release syndrome is characterized by the rapid release of pro-inflammatory cytokines and immune cell activation. T cell-engaging bispecific antibodies can cause transient release of cytokines that may potentially suppress CYP450 enzymes. Utilizing the PBPK modeling approach to evaluate the CRS-mediated drug interactions in a regulatory submission can be traced back to the first FDA-approved T-cell-engaging bispecific antibody, blinatumomab, in 2014.<span><sup>5</sup></span> Over the past 10 years, a few additional T-cell-engaging bispecific antibodies were approved by FDA (mosunetuzumab, tebentafusp, teclistamab, epcoritamab, glofitamab, and talquetamab). We examined the FDA's biologics license application assessment packages, USPIs (United States Prescribing Information), and relevant PBPK publications to see how drug interactions mediated by CRS were evaluated and reported to healthcare professionals.</p><p>Among the seven programs (blinatumomab, mosunetuzumab, tebentafusp, teclistamab, epcoritamab, glofitamab, and talquetamab), no dedicated drug–drug interaction (DDI) study for CRS was conducted. Except for tebentafusp, six programs include “cautious periods” when the CRS-mediated drug interactions may occur and ask for monitoring for toxicity of concomitant CYP substrates where minimal changes in concentration may lead to serious adverse reactions in Section 7 (Drug Interactions) of USPI. PBPK analyses were submitted for five programs (blinatumomab, mosunetuzumab, teclistamab, glofitamab, and talquetamab) to evaluate the duration and magnitude of CRS-mediated drug interactions (Table 1). None of the PBPK evaluations was deemed adequate by the FDA. Therefore, it appears that the recommendation in Section 7 of USPI is based on the observed CRS period. We also noted that the delayed onset and offset effect of IL-6 on CYP enzymes, and the enzyme turnover rate were considered by the FDA when recommending the “cautious period” for talquetamab.<span><sup>9</sup></span></p><p>On the contrary, the sponsors appear to have high confidence in their PBPK model predictions of CRS-mediated drug interactions.<span><sup>1-4</sup></span> The quantitative prediction results are also published on the product website to inform the healthcare providers regarding the exposure changes of concomitantly administered CYP substrates.<span><sup>10, 11</sup></span></p><p>In our opinion, the current practice of informing Section 7 of USPI is not optimal and not a patient-centric clinical pharmacology approach. In the following sections, we evaluate what has been done and what is lacking in the current PBPK analyses, and propose approaches to improve the confidence in the PBPK modeling and simulation, and eventually to better inform the USPI, healthcare providers, and patients on the risk of CRS-mediated drug interactions.</p><p>Because detailed FDA assessments were often redacted, we have to assume that the same analyses published by the sponsors were submitted in BLA (Table 1). We found that sponsors' analyses were generally rigorous and risk-based. All sponsors developed a fit-for-purpose PK model of IL-6 to capture transient elevation of the cytokine after dosing of the biologic product, combined cytokine profiles with its CYP suppression mechanism and turnover of the CYP enzyme to predict the magnitude and duration of DDI. For all cases, worst-case scenarios were explored using IL-6 profile that represented patient(s) with the highest observed elevation following treatment of the biologics. In some cases, the effects of CRS on CYP3A in the gut,<span><sup>1, 3</sup></span> co-medications that may suppress IL-6,<span><sup>1</sup></span> and underlying disease (using virtual cancer population)<span><sup>3</sup></span> were evaluated.</p><p>The rationales behind FDA's inadequate conclusions include the lack of an established exposure–response relationship between IL-6 and CYP suppression as well as the time course of the interaction, the focus on evaluating the effect of IL-6 on CYP substrates but no other cytokines (such as IL-2, IL-6, IL-10, TNF-α, IFN-γ, etc.), and the use of data in patients with chronic autoimmune and inflammatory diseases to validate PBPK model for IL-6.<span><sup>5-8</sup></span> The IL-6 levels in those diseases are generally much lower compared with the IL-6 levels in CRS (a few hundred vs. several thousand pg/mL reported by the sponsors<span><sup>2-7</sup></span>), while the reported in vitro EC<sub>50</sub> of IL-6 against CYP3A4 activity was ~200 pg/mL in the absence of dexamethasone.<span><sup>12</sup></span> The limitation that the time course of the CRS-mediated inhibition effect on CYP enzymes has not been studied (clinically) in the non-rheumatoid arthritis patients and the PBPK modeling may not capture the time course of recovery from the suppression effect, is a valid point. However, the transient nature of CRS and the number of CYP enzymes likely affected by elevated cytokines indeed make it difficult to design and conduct dedicated clinical studies to address CRS-mediated drug interactions.</p><p>Additional in vivo data might be needed to address the knowledge gaps. A recent PBPK publication evaluated the impact of elevated IL-6 on CYP3A substrates in patients with COVID-19 predicted DDI liability under the highest observed IL-6 concentration of 4462 pg/mL, which might provide an additional dataset for model validation.<span><sup>13</sup></span> Ideally, a dedicated drug interaction study could be conducted with appropriate design and data collection for relevant model validation. This can be considered as an one-time investment to evaluate future (and confirm the past) CRS-mediated drug interactions. Recognizing the challenge of conducting dedicated clinical studies to fully address transient nature of CRS and the number of CYPs affected, one can establish real-world evidence detecting potential adverse effects related to elevated exposure of co-medications as a result of much higher cytokine levels during CRS . Considering the number of T-cell-engaging bispecific antibodies in development, and the common pathway of the postulated drug interaction through CRS, it might be worthwhile for both the sponsors and regulators to collaborate on addressing these knowledge gaps and enhancing the predictability of PBPK.</p><p>To summarize, we commented on the current practice of how CRS-mediated drug interaction risk is being communicated by both the Agency and the sponsors. We expressed our opinion that the current practice is not optimal, not patient-centric, and lacks quantitative evaluation. We reviewed what has been done and what is lacking in the current PBPK modeling to evaluate the CRS-mediated drug interactions. We identified a few gaps and discussed the approaches to achieve the goal of using PBPK to better inform the clinical use of concomitant medications in CRS events. We hope that soon, there will be a breakthrough in utilizing the quantitative approach to inform patients and healthcare providers about CRS-mediated drug interaction risks.</p><p>No funding was received for this work.</p><p>The authors declared no competing interests for this work.</p><p>The views expressed in this editorial are those of the authors and do not represent the opinions of their employers.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.1000,"publicationDate":"2024-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13185","citationCount":"0","resultStr":"{\"title\":\"Is PBPK useful to inform product label on managing clinically significant drug interactions mediated by cytokine release syndrome?\",\"authors\":\"Xinyuan Zhang,&nbsp;Ping Zhao\",\"doi\":\"10.1002/psp4.13185\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Evaluating drug interactions caused by cytokine release syndrome (CRS) with PBPK (Physiologically Based Pharmacokinetic) modeling has been reported in some bispecific antibody regulatory submissions for 10 years. However, the published regulatory reviews and sponsors' analyses seem to disagree on the roles of PBPK modeling in regulatory decision-making. In this editorial, we reviewed and provided our opinions on the FDA's current practice and sponsors' position in evaluating CRS-mediated drug interactions. We discussed what has been done and what is lacking in the current PBPK approach assessing the CRS-mediated drug interactions and proposed areas to bridge the gaps. And finally, we call to actions to improve the current practice toward a patient-centric clinical pharmacology approach with more quantitative assessment and management of CRS-mediated drug interactions.</p><p>The manuscript by Willemin et al.<span><sup>1</sup></span> described the use of a PBPK approach to evaluate the effect of elevated IL-6 following the treatment of teclistamab on the PK of CYP enzyme (1A2, 2C9, 2C19, 3A4, 3A5) substrates. This marks the 4th PBPK publication by CPT-PSP of the effect of CRS as a result of biologics-treatment on co-medications that are CYP substrates, after blinatumomab,<span><sup>2</sup></span> mosunetuzumab,<span><sup>3</sup></span> and glofitamab.<span><sup>4</sup></span> The scientific community and drug developers are using the PBPK modeling tool to study the effect of CRS on the PK and safety of co-administered CYP substrate drugs. However, there seems to be a gap between the peer-reviewed papers<span><sup>1-4</sup></span> and the regulatory evaluations<span><sup>5-8</sup></span> in terms of concluding the impact of PBPK predictions. In this editorial, we examine the gap and share our opinions on the value, expectation, and future of PBPK modeling in this specific area with the aim of increasing awareness, calling for enhanced predictive performance, and ultimately, achieving patient-centric clinical pharmacology.</p><p>Cytokine release syndrome is characterized by the rapid release of pro-inflammatory cytokines and immune cell activation. T cell-engaging bispecific antibodies can cause transient release of cytokines that may potentially suppress CYP450 enzymes. Utilizing the PBPK modeling approach to evaluate the CRS-mediated drug interactions in a regulatory submission can be traced back to the first FDA-approved T-cell-engaging bispecific antibody, blinatumomab, in 2014.<span><sup>5</sup></span> Over the past 10 years, a few additional T-cell-engaging bispecific antibodies were approved by FDA (mosunetuzumab, tebentafusp, teclistamab, epcoritamab, glofitamab, and talquetamab). We examined the FDA's biologics license application assessment packages, USPIs (United States Prescribing Information), and relevant PBPK publications to see how drug interactions mediated by CRS were evaluated and reported to healthcare professionals.</p><p>Among the seven programs (blinatumomab, mosunetuzumab, tebentafusp, teclistamab, epcoritamab, glofitamab, and talquetamab), no dedicated drug–drug interaction (DDI) study for CRS was conducted. Except for tebentafusp, six programs include “cautious periods” when the CRS-mediated drug interactions may occur and ask for monitoring for toxicity of concomitant CYP substrates where minimal changes in concentration may lead to serious adverse reactions in Section 7 (Drug Interactions) of USPI. PBPK analyses were submitted for five programs (blinatumomab, mosunetuzumab, teclistamab, glofitamab, and talquetamab) to evaluate the duration and magnitude of CRS-mediated drug interactions (Table 1). None of the PBPK evaluations was deemed adequate by the FDA. Therefore, it appears that the recommendation in Section 7 of USPI is based on the observed CRS period. We also noted that the delayed onset and offset effect of IL-6 on CYP enzymes, and the enzyme turnover rate were considered by the FDA when recommending the “cautious period” for talquetamab.<span><sup>9</sup></span></p><p>On the contrary, the sponsors appear to have high confidence in their PBPK model predictions of CRS-mediated drug interactions.<span><sup>1-4</sup></span> The quantitative prediction results are also published on the product website to inform the healthcare providers regarding the exposure changes of concomitantly administered CYP substrates.<span><sup>10, 11</sup></span></p><p>In our opinion, the current practice of informing Section 7 of USPI is not optimal and not a patient-centric clinical pharmacology approach. In the following sections, we evaluate what has been done and what is lacking in the current PBPK analyses, and propose approaches to improve the confidence in the PBPK modeling and simulation, and eventually to better inform the USPI, healthcare providers, and patients on the risk of CRS-mediated drug interactions.</p><p>Because detailed FDA assessments were often redacted, we have to assume that the same analyses published by the sponsors were submitted in BLA (Table 1). We found that sponsors' analyses were generally rigorous and risk-based. All sponsors developed a fit-for-purpose PK model of IL-6 to capture transient elevation of the cytokine after dosing of the biologic product, combined cytokine profiles with its CYP suppression mechanism and turnover of the CYP enzyme to predict the magnitude and duration of DDI. For all cases, worst-case scenarios were explored using IL-6 profile that represented patient(s) with the highest observed elevation following treatment of the biologics. In some cases, the effects of CRS on CYP3A in the gut,<span><sup>1, 3</sup></span> co-medications that may suppress IL-6,<span><sup>1</sup></span> and underlying disease (using virtual cancer population)<span><sup>3</sup></span> were evaluated.</p><p>The rationales behind FDA's inadequate conclusions include the lack of an established exposure–response relationship between IL-6 and CYP suppression as well as the time course of the interaction, the focus on evaluating the effect of IL-6 on CYP substrates but no other cytokines (such as IL-2, IL-6, IL-10, TNF-α, IFN-γ, etc.), and the use of data in patients with chronic autoimmune and inflammatory diseases to validate PBPK model for IL-6.<span><sup>5-8</sup></span> The IL-6 levels in those diseases are generally much lower compared with the IL-6 levels in CRS (a few hundred vs. several thousand pg/mL reported by the sponsors<span><sup>2-7</sup></span>), while the reported in vitro EC<sub>50</sub> of IL-6 against CYP3A4 activity was ~200 pg/mL in the absence of dexamethasone.<span><sup>12</sup></span> The limitation that the time course of the CRS-mediated inhibition effect on CYP enzymes has not been studied (clinically) in the non-rheumatoid arthritis patients and the PBPK modeling may not capture the time course of recovery from the suppression effect, is a valid point. However, the transient nature of CRS and the number of CYP enzymes likely affected by elevated cytokines indeed make it difficult to design and conduct dedicated clinical studies to address CRS-mediated drug interactions.</p><p>Additional in vivo data might be needed to address the knowledge gaps. A recent PBPK publication evaluated the impact of elevated IL-6 on CYP3A substrates in patients with COVID-19 predicted DDI liability under the highest observed IL-6 concentration of 4462 pg/mL, which might provide an additional dataset for model validation.<span><sup>13</sup></span> Ideally, a dedicated drug interaction study could be conducted with appropriate design and data collection for relevant model validation. This can be considered as an one-time investment to evaluate future (and confirm the past) CRS-mediated drug interactions. Recognizing the challenge of conducting dedicated clinical studies to fully address transient nature of CRS and the number of CYPs affected, one can establish real-world evidence detecting potential adverse effects related to elevated exposure of co-medications as a result of much higher cytokine levels during CRS . Considering the number of T-cell-engaging bispecific antibodies in development, and the common pathway of the postulated drug interaction through CRS, it might be worthwhile for both the sponsors and regulators to collaborate on addressing these knowledge gaps and enhancing the predictability of PBPK.</p><p>To summarize, we commented on the current practice of how CRS-mediated drug interaction risk is being communicated by both the Agency and the sponsors. We expressed our opinion that the current practice is not optimal, not patient-centric, and lacks quantitative evaluation. We reviewed what has been done and what is lacking in the current PBPK modeling to evaluate the CRS-mediated drug interactions. We identified a few gaps and discussed the approaches to achieve the goal of using PBPK to better inform the clinical use of concomitant medications in CRS events. We hope that soon, there will be a breakthrough in utilizing the quantitative approach to inform patients and healthcare providers about CRS-mediated drug interaction risks.</p><p>No funding was received for this work.</p><p>The authors declared no competing interests for this work.</p><p>The views expressed in this editorial are those of the authors and do not represent the opinions of their employers.</p>\",\"PeriodicalId\":10774,\"journal\":{\"name\":\"CPT: Pharmacometrics & Systems Pharmacology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.1000,\"publicationDate\":\"2024-06-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13185\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"CPT: Pharmacometrics & Systems Pharmacology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/psp4.13185\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"CPT: Pharmacometrics & Systems Pharmacology","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/psp4.13185","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

所有申办者都开发了一个适合 IL-6 目的的 PK 模型,以捕捉生物制品用药后该细胞因子的瞬时升高,并将细胞因子图谱与其 CYP 抑制机制和 CYP 酶的周转率相结合,以预测 DDI 的程度和持续时间。在所有病例中,最坏的情况是使用代表生物制剂治疗后观察到最高升高的患者的 IL-6 图谱。在某些情况下,还评估了 CRS 对肠道中 CYP3A 的影响、1、3 可能抑制 IL-6 的联合用药1 以及潜在疾病(使用虚拟癌症人群)3。FDA 得出不充分结论的理由包括:IL-6 与 CYP 抑制之间缺乏既定的暴露-反应关系以及相互作用的时间过程;只重点评估了 IL-6 对 CYP 底物的影响,而未评估其他细胞因子(如 IL-2、IL-6、IL-10、TNF-α、IFN-γ 等)。5-8与 CRS 中的 IL-6 水平相比,这些疾病中的 IL-6 水平通常要低得多(几百 pg/mL 对几千 pg/mL),而据报道,在没有地塞米松的情况下,IL-6 对 CYP3A4 活性的体外 EC50 为 ~200 pg/mL。在非类风湿性关节炎患者中,尚未对 CRS 介导的 CYP 酶抑制作用的时间过程进行(临床)研究,因此 PBPK 模型可能无法捕捉到抑制作用恢复的时间过程。然而,CRS 的短暂性和可能受细胞因子升高影响的 CYP 酶的数量确实使得设计和开展专门的临床研究来解决 CRS 介导的药物相互作用变得困难。最近发表的一篇 PBPK 论文评估了 IL-6 升高对 COVID-19 患者 CYP3A 底物的影响,并预测在观察到的最高 IL-6 浓度(4462 pg/mL)下的 DDI 责任,这可能为模型验证提供了额外的数据集。这可视为一次性投资,用于评估未来(并确认过去)CRS 介导的药物相互作用。认识到开展专门的临床研究以全面解决 CRS 的瞬时性和受影响的 CYPs 数量所面临的挑战,我们可以建立真实世界的证据,以检测由于 CRS 期间细胞因子水平大幅升高而导致的与联合用药暴露升高有关的潜在不良反应。考虑到正在开发中的T细胞参与性双特异性抗体的数量,以及通过CRS推测的药物相互作用的共同途径,可能值得申办者和监管机构合作解决这些知识差距,并提高PBPK的可预测性。总之,我们对目前机构和申办者如何沟通CRS介导的药物相互作用风险的做法发表了意见。我们认为目前的做法并不理想,不是以患者为中心,而且缺乏定量评估。我们回顾了当前 PBPK 建模在评估 CRS 介导的药物相互作用方面所做的工作和存在的不足。我们找出了一些不足之处,并讨论了实现使用 PBPK 的目标的方法,以便在 CRS 事件中更好地为临床使用伴随药物提供信息。我们希望很快就能在利用定量方法告知患者和医疗服务提供者有关 CRS 媒介的药物相互作用风险方面取得突破。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Is PBPK useful to inform product label on managing clinically significant drug interactions mediated by cytokine release syndrome?

Evaluating drug interactions caused by cytokine release syndrome (CRS) with PBPK (Physiologically Based Pharmacokinetic) modeling has been reported in some bispecific antibody regulatory submissions for 10 years. However, the published regulatory reviews and sponsors' analyses seem to disagree on the roles of PBPK modeling in regulatory decision-making. In this editorial, we reviewed and provided our opinions on the FDA's current practice and sponsors' position in evaluating CRS-mediated drug interactions. We discussed what has been done and what is lacking in the current PBPK approach assessing the CRS-mediated drug interactions and proposed areas to bridge the gaps. And finally, we call to actions to improve the current practice toward a patient-centric clinical pharmacology approach with more quantitative assessment and management of CRS-mediated drug interactions.

The manuscript by Willemin et al.1 described the use of a PBPK approach to evaluate the effect of elevated IL-6 following the treatment of teclistamab on the PK of CYP enzyme (1A2, 2C9, 2C19, 3A4, 3A5) substrates. This marks the 4th PBPK publication by CPT-PSP of the effect of CRS as a result of biologics-treatment on co-medications that are CYP substrates, after blinatumomab,2 mosunetuzumab,3 and glofitamab.4 The scientific community and drug developers are using the PBPK modeling tool to study the effect of CRS on the PK and safety of co-administered CYP substrate drugs. However, there seems to be a gap between the peer-reviewed papers1-4 and the regulatory evaluations5-8 in terms of concluding the impact of PBPK predictions. In this editorial, we examine the gap and share our opinions on the value, expectation, and future of PBPK modeling in this specific area with the aim of increasing awareness, calling for enhanced predictive performance, and ultimately, achieving patient-centric clinical pharmacology.

Cytokine release syndrome is characterized by the rapid release of pro-inflammatory cytokines and immune cell activation. T cell-engaging bispecific antibodies can cause transient release of cytokines that may potentially suppress CYP450 enzymes. Utilizing the PBPK modeling approach to evaluate the CRS-mediated drug interactions in a regulatory submission can be traced back to the first FDA-approved T-cell-engaging bispecific antibody, blinatumomab, in 2014.5 Over the past 10 years, a few additional T-cell-engaging bispecific antibodies were approved by FDA (mosunetuzumab, tebentafusp, teclistamab, epcoritamab, glofitamab, and talquetamab). We examined the FDA's biologics license application assessment packages, USPIs (United States Prescribing Information), and relevant PBPK publications to see how drug interactions mediated by CRS were evaluated and reported to healthcare professionals.

Among the seven programs (blinatumomab, mosunetuzumab, tebentafusp, teclistamab, epcoritamab, glofitamab, and talquetamab), no dedicated drug–drug interaction (DDI) study for CRS was conducted. Except for tebentafusp, six programs include “cautious periods” when the CRS-mediated drug interactions may occur and ask for monitoring for toxicity of concomitant CYP substrates where minimal changes in concentration may lead to serious adverse reactions in Section 7 (Drug Interactions) of USPI. PBPK analyses were submitted for five programs (blinatumomab, mosunetuzumab, teclistamab, glofitamab, and talquetamab) to evaluate the duration and magnitude of CRS-mediated drug interactions (Table 1). None of the PBPK evaluations was deemed adequate by the FDA. Therefore, it appears that the recommendation in Section 7 of USPI is based on the observed CRS period. We also noted that the delayed onset and offset effect of IL-6 on CYP enzymes, and the enzyme turnover rate were considered by the FDA when recommending the “cautious period” for talquetamab.9

On the contrary, the sponsors appear to have high confidence in their PBPK model predictions of CRS-mediated drug interactions.1-4 The quantitative prediction results are also published on the product website to inform the healthcare providers regarding the exposure changes of concomitantly administered CYP substrates.10, 11

In our opinion, the current practice of informing Section 7 of USPI is not optimal and not a patient-centric clinical pharmacology approach. In the following sections, we evaluate what has been done and what is lacking in the current PBPK analyses, and propose approaches to improve the confidence in the PBPK modeling and simulation, and eventually to better inform the USPI, healthcare providers, and patients on the risk of CRS-mediated drug interactions.

Because detailed FDA assessments were often redacted, we have to assume that the same analyses published by the sponsors were submitted in BLA (Table 1). We found that sponsors' analyses were generally rigorous and risk-based. All sponsors developed a fit-for-purpose PK model of IL-6 to capture transient elevation of the cytokine after dosing of the biologic product, combined cytokine profiles with its CYP suppression mechanism and turnover of the CYP enzyme to predict the magnitude and duration of DDI. For all cases, worst-case scenarios were explored using IL-6 profile that represented patient(s) with the highest observed elevation following treatment of the biologics. In some cases, the effects of CRS on CYP3A in the gut,1, 3 co-medications that may suppress IL-6,1 and underlying disease (using virtual cancer population)3 were evaluated.

The rationales behind FDA's inadequate conclusions include the lack of an established exposure–response relationship between IL-6 and CYP suppression as well as the time course of the interaction, the focus on evaluating the effect of IL-6 on CYP substrates but no other cytokines (such as IL-2, IL-6, IL-10, TNF-α, IFN-γ, etc.), and the use of data in patients with chronic autoimmune and inflammatory diseases to validate PBPK model for IL-6.5-8 The IL-6 levels in those diseases are generally much lower compared with the IL-6 levels in CRS (a few hundred vs. several thousand pg/mL reported by the sponsors2-7), while the reported in vitro EC50 of IL-6 against CYP3A4 activity was ~200 pg/mL in the absence of dexamethasone.12 The limitation that the time course of the CRS-mediated inhibition effect on CYP enzymes has not been studied (clinically) in the non-rheumatoid arthritis patients and the PBPK modeling may not capture the time course of recovery from the suppression effect, is a valid point. However, the transient nature of CRS and the number of CYP enzymes likely affected by elevated cytokines indeed make it difficult to design and conduct dedicated clinical studies to address CRS-mediated drug interactions.

Additional in vivo data might be needed to address the knowledge gaps. A recent PBPK publication evaluated the impact of elevated IL-6 on CYP3A substrates in patients with COVID-19 predicted DDI liability under the highest observed IL-6 concentration of 4462 pg/mL, which might provide an additional dataset for model validation.13 Ideally, a dedicated drug interaction study could be conducted with appropriate design and data collection for relevant model validation. This can be considered as an one-time investment to evaluate future (and confirm the past) CRS-mediated drug interactions. Recognizing the challenge of conducting dedicated clinical studies to fully address transient nature of CRS and the number of CYPs affected, one can establish real-world evidence detecting potential adverse effects related to elevated exposure of co-medications as a result of much higher cytokine levels during CRS . Considering the number of T-cell-engaging bispecific antibodies in development, and the common pathway of the postulated drug interaction through CRS, it might be worthwhile for both the sponsors and regulators to collaborate on addressing these knowledge gaps and enhancing the predictability of PBPK.

To summarize, we commented on the current practice of how CRS-mediated drug interaction risk is being communicated by both the Agency and the sponsors. We expressed our opinion that the current practice is not optimal, not patient-centric, and lacks quantitative evaluation. We reviewed what has been done and what is lacking in the current PBPK modeling to evaluate the CRS-mediated drug interactions. We identified a few gaps and discussed the approaches to achieve the goal of using PBPK to better inform the clinical use of concomitant medications in CRS events. We hope that soon, there will be a breakthrough in utilizing the quantitative approach to inform patients and healthcare providers about CRS-mediated drug interaction risks.

No funding was received for this work.

The authors declared no competing interests for this work.

The views expressed in this editorial are those of the authors and do not represent the opinions of their employers.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
5.00
自引率
11.40%
发文量
146
审稿时长
8 weeks
×
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