推动肿瘤剂量优化:行动呼吁。

IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY CPT: Pharmacometrics & Systems Pharmacology Pub Date : 2024-05-22 DOI:10.1002/psp4.13157
Karthik Venkatakrishnan, Priya Jayachandran, Shirley K. Seo, Piet H. van der Graaf, John A. Wagner, Neeraj Gupta
{"title":"推动肿瘤剂量优化:行动呼吁。","authors":"Karthik Venkatakrishnan,&nbsp;Priya Jayachandran,&nbsp;Shirley K. Seo,&nbsp;Piet H. van der Graaf,&nbsp;John A. Wagner,&nbsp;Neeraj Gupta","doi":"10.1002/psp4.13157","DOIUrl":null,"url":null,"abstract":"<p>Project Optimus is a major FDA initiative aimed at ensuring dose optimization in oncology drug development, moving away from the maximum tolerated dose paradigm and prospectively characterizing dose–response for efficacy and safety for patient-focused maximization of benefit versus risk.<span><sup>1-3</sup></span> Mitigating toxicities and enhancing overall benefit versus risk of oncology therapies necessitates dose optimization with commitment to evaluation of innovative dosing paradigms including individualized approaches, where appropriate. This requires the quantitative integration of pharmacological mechanism of action, efficacy, and safety in the context of associated population variability. The problem of dose optimization in the context of mechanism of action, cancer pathophysiology, and associated population variability sits neatly at the intersection of translational/ precision medicine and quantitative clinical pharmacology and is important to approach with a patient-focused mindset.</p><p>Forums convened on the topic of oncology dose optimization largely engage scientific leaders primarily working on oncology research and development, and cancer medicine. These include workshops organized by Friends of Cancer Research (FOCR),<span><sup>4</sup></span> American Society of Clinical Oncology (ASCO),<span><sup>5, 6</sup></span> American Association for Cancer Research (AACR),<span><sup>7, 8</sup></span> and the International Society of Pharmacometrics (ISoP)<span><sup>9</sup></span> in partnership with the US Food and Drugs Administration (FDA). Of note, some of these efforts have yielded seminal publications<span><sup>1, 2, 10-13</sup></span> and White Papers<span><sup>14</sup></span> offering initial recommendations, including availability of a Draft FDA guidance on the topic.<span><sup>15</sup></span> We posited that the American Society for Clinical Pharmacology and Therapeutics (ASCPT) – as a premier scientific and professional organization for clinical pharmacology and translational medicine – is optimally positioned to host a discussion of opportunities for our constituent disciplines (e.g., translational science, clinical pharmacology, pharmacometrics) to synergistically address this problem with a multi-disciplinary approach. To this end, a session was convened at the 2023 ASCPT Annual Meeting bringing together representative scientific leaders from the three scientific journals of the Society – <i>Clinical Pharmacology and Therapeutics</i> (<i>CPT</i>), <i>Clinical and Translational Science</i> (<i>CTS</i>), and <i>CPT: Pharmacometrics and Systems Pharmacology</i> (<i>PSP</i>). These scientific leaders, as at-large representatives of the disciplines of clinical pharmacology and translational medicine, were invited to bring forward their opinions and participate in a fireside chat to identify opportunities for moving the oncology dose optimization needle. This enabled engagement of a broad group of experts without requiring primary scientific or professional affiliation to the oncology therapeutic area, thereby maximizing diversity of opinion, out-of-the-box solutioning, and fresh perspectives that should help advance us beyond the current state. Ahead of the session at the Annual Meeting, a survey was launched to ASCPT members and meeting attendees to get our finger on the pulse of our Society's membership on issues faced in oncology dose optimization and provide substrate for the fireside chat with the expert panel. Herein, we present the findings from this ASCPT survey, and review the insights gained from this Annual Meeting session including recommendations for our scientific communities to join forces and drive progress.</p><p>A focused survey was developed and sent out in February 2023 to meeting attendees and broader ASCPT membership on the topic of the session, which consisted of six questions that were relevant to dose optimization (Data S1). The survey was open for 3 weeks and 65 respondents participated in the survey.</p><p>We were not only interested in understanding the background of survey respondents that may influence their feedback, but also various dose optimization approaches including challenges with various modalities. In response to our question about full time engagement with oncology R&amp;D, 58% of respondents were either not engaged or only had part time engagement with oncology R&amp;D. This suggested that survey feedback was from members with diverse backgrounds, as intended. Similarly, we were interested in understanding if strategies for dose optimization in other therapeutic areas are relevant for oncology therapies. 86% of respondents suggested that strategies from other therapeutic areas are indeed relevant to oncology.</p><p>Three questions focused on approaches applied for dose optimization – one on the utility of pharmacodynamic (PD) biomarkers, another one on quantitative approaches for dose selection and finally a question on study designs for dose optimization with a focus on randomization. 92% of responses suggest that PD biomarkers are at least useful. Clinical Exposure-response modeling (57%) followed by pharmacokinetic (PK)/PD modeling (28%) are most preferred approaches for selecting doses. Of note, 62% of respondents did not consider randomized dose-ranging evaluation as necessary for dose optimization, suggesting the value of application on a case-by-case approach leveraging the totality of evidence to optimize dose (Figure 1).</p><p>Given that oncology is a therapeutic area with a wide range of modalities from small molecules to cell therapies, we sought to understand the level of challenge associated with dose optimization in developing each of these modalities. Respondents noted that dose optimization for next-generation cytotoxic agents, small molecule targeted agents, and monoclonal antibodies is relatively straightforward with many historical examples to guide dose selection. However, dose optimization for antibody-drug conjugates was viewed to be moderately complex while newer modalities such as multi-specific biologics and cell therapies were considered very challenging with very few or no examples of dose optimization (Figure 2).</p><p>From a translational perspective, the focus of dose optimization is to find the <i>right</i> dose for patients as <i>swiftly</i> and <i>safely</i> as possible, buttressed by nonclinical and clinical translational data. Translational dose optimization doesn't always have to be complex. Goldstein et al.<span><sup>16</sup></span> describe a relatively simple concept for translational dose optimization for small molecule targeted oncology agents in the first-in-human setting. These suggestions can be implemented today. The approved doses of 25 targeted therapies were examined and the average free concentration at steady state (Css) was determined to be similar to the in vitro cell potency (half-maximal inhibitory concentration (IC50)). Furthermore, the authors propose a revised first-in-human trial design for next-generation targeted therapy in which dose cohort expansion is initiated at doses less than the maximum tolerated dose when there is evidence of clinical activity and Css exceeds a threshold informed by in vitro cell potency.</p><p>Ji et al.<span><sup>17</sup></span> describe another relatively straightforward approach to translational dose optimization in oncology. In this case, the drug is an inhibitor of Porcupine, a membrane-bound O-acyltransferase required for Wnt secretion. Wnt pathway is expressed in skin tissues; AXIN2 mRNA expression in skin is a robust and sensitive biomarker for the Wnt pathway. A predominant safety issue in this case is dysgeusia. The authors performed integrated population PK and exposure-response analyses of PD biomarker and safety data to determine the recommended dose for expansion, rather than the conventional maximum tolerated approach.</p><p>More complex approaches are also possible and have great utility, particularly for complex therapeutic modalities. Weddell et al.<span><sup>18</sup></span> describe an elegant mechanistic model that characterizes antibody drug conjugate (ADC) pharmacokinetics and tumor penetration by incorporating tumor growth inhibition via ADC binding radially across solid tumors. The model demonstrates that with low target expression, the potency of the payload should be increased. Furthermore, the model mechanistically links clinical response rates and relapse or resistance to ADC therapies, which could facilitate dose optimization. In another recent example, Susilo et al. leveraged a quantitative systems pharmacology (QSP) model of an anti-CD20/CD3 T-cell engaging bispecific antibody, mosunetuzumab, to account for different dosing regimens and inter-patient heterogeneity in the phase I study to identify biological determinants of clinical response and dose/exposure-response relationships using a novel QSP-derived digital twins approach.<span><sup>19</sup></span> Approaches of this nature raise opportunities for multi-dimensional optimization across the dimensions of dose, patient population, and combination partner – a challenge faced routinely in oncology drug development.</p><p>The value of new, innovative biomarkers in translational development is continuing to be realized. Recent examples indicate the emerging value of circulating tumor DNA (ctDNA).<span><sup>20, 21</sup></span> The translational utility of ctDNA, cancer cell DNA found in the bloodstream, is manifold, including detecting and diagnosing cancer, guiding tumor-specific treatment, monitoring treatment and remission. In the context of dose optimization, characterizing the underlying exposure-response relationship for on-treatment ctDNA dynamics to inform definition of a clinically active dose range represents an untapped opportunity. Another important innovation has been in the area of digital health technologies such as a proposed multi-domain, digital model for capturing functional status, and health-related quality of life in oncology,<span><sup>22</sup></span> which can be particularly relevant to realize the promise of Project Optimus aimed at dosage optimization for improved quality of life during long-term therapy.</p><p>ASCPT, clinical pharmacologists, and translational scientists have a key role in collaboration on dose optimization challenges and opportunities across different stakeholders. ASCPT membership straddles a variety of stakeholders including academics, industry, regulators, and others to help drive brainstorming and consensus formation. For example, Ji et al.,<span><sup>23</sup></span> reported on an ASCPT annual scientific meeting symposium. The authors describe a number of challenges observed before Project Optimus, including post-market dose-finding, continued use of traditional 3 + 3 designs, lack of characterization of chronic toxicity, and opportunities for adopting novel designs and testing more than one dose in phase 2/3 clinical trials. Oncology is one of the most innovative fields in science and yet there are only very few examples of value-added use of pharmacodynamic biomarkers and dose optimization. Cross-stakeholder work and Project Optimus are expected to drive the field to increased biomarker-based and model-informed solutions for oncology dose finding and optimization.</p><p>In their paper “The Future of Clinical Trial Design in Oncology,” Spreafico and co-workers from the Toronto Princess Margaret Cancer Centre<span><sup>24</sup></span> describe how therapeutic approaches in cancer drug discovery and development have shifted from traditional cytotoxic chemotherapy focused on histology-based targets to molecularly targeted and immune therapies in patient subsets stratified by biomarkers and other diagnostic precision tools. The authors argue that the classical clinical trial paradigm in oncology urgently needs to be transformed to ensure patients will benefit from this scientific revolution in a timely manner. In a wide-ranging call to action, they present a patient-centric framework for the next-generation oncology clinical trials, which maps out the journey of a trial participant as a dynamic and adaptive one continuously leveraging scientific and technological innovations to develop individualized therapeutic strategies. They conclude that “<i>The success of next-generation clinical trials will be based on the fundamental principles of acting locally to learn globally and treating participants individually to advance the field collectively</i>.” This speaks directly to the opportunity for clinical pharmacology to play a core role in this new paradigm, in particular with regard to dose optimization and individualization based on quantitative, model-informed approaches that integrate the totality knowledge and data of the drug, disease, and patient. An example of such an approach is QSP, which in a recent survey conducted by the ISoP was identified as an emerging key tool utilized by oncology drug developers for dose and dose regimen selection and optimization.<span><sup>25</sup></span> A recent example was presented by Li et al.,<span><sup>26</sup></span> who developed a mechanistic model to determine the recommended phase II dose (R2P2D) for epcoritamab, a CD3×CD20 bispecific antibody (bsAb). The authors justified this novel approach, which integrated preclinical, clinical PK, biomarker, tumor, and response data from the dose-escalation part of the phase I/II trial, on the basis that traditional dose/exposure-response modeling methods may not adequately predict the complex dose/exposure-response relationship for bsAbs. Therefore, trimer formation predicted by the mechanistic model instead of actual clinical measures was used to guide dose prediction.</p><p>Along the same lines, in a paper by Chelliah and representatives from a consortium of pharmaceutical companies,<span><sup>27</sup></span> the case is made that conventional, empirical pharmacometrics approaches do not fully capitalize on all the available biological and disease knowledge and that QSP models provide a more rational and better alternative to guide complex IO combination therapy development. Their proposal that “virtual patients” simulated by the QSP model under conditions that mimic the actual clinical trial should be added to the drug development paradigm is fully aligned with the earlier-mentioned call-to-action by Spreafico et al. outlined in <i>Figure 2</i> of their publication,<span><sup>24</sup></span> suggesting that the future of clinical trial design in oncology may already have arrived.</p><p>Poorly characterized dose and schedule may lead to selection of a dose that provides more toxicity without additional efficacy, severe toxicities that require a high rate of dose reductions or premature discontinuation and may result in missed opportunity for continued benefit from the drug. To optimize benefit versus risk with a patient-focused approach, there remain significant opportunities for model-based analyses to inform dosing regimen design that may sometimes involve non-static posology, with patient response or outcome-based dose adaptation to ensure individualized dosing for maximizing benefit versus risk.<span><sup>28, 29</sup></span></p><p>Project Optimus offers a pivotal opportunity to reform the oncology dosing paradigm using a robust quantitative clinical pharmacology framework.<span><sup>2, 3, 14, 30-33</sup></span> By integrating a model development lifecycle, Bayesian trial designs, and a learning-and-confirming mindset across the development spectrum, this framework may be used to prospectively guide dose optimization.</p><p>One of the main advantages of examining an oncology challenge as a non-oncologist is the ability to translate similar principles and successful examples from other therapeutic areas to oncology. These examples can aid in enriching a holistic approach toward solving longstanding problems. One clear correlate is in HIV drug discovery. In the 1980s, the average life expectancy following an AIDS diagnosis was approximately one year. And by the early 1990s, HIV was the leading cause of death among Americans aged 25 to 44. In many ways, much like with cancer, the urgency to save lives and need for therapeutics to control the epidemic fueled innovation and discovery. The beginning of that discovery phase did lead to some unsophisticated dosing – zidovudine was initially studied and approved at a dosage of 200 mg q4h, which caused severe anemia and neutropenia. However, more fine-tuning of the dose through clinical trials eventually led to its current dosage regimen of 300 mg twice daily. Several advancements along the way led to HIV infection largely being regarded as a chronic condition with near normal life expectancy for patients and a much improved quality of life. Some of these advancements included a deeper and continual understanding of the pharmacological mechanisms of antiretroviral agents, development of enhanced diagnostics, and acceptance of early biomarkers. When these approaches were deployed simultaneously, the result was a highly integrated, advanced methodology to solving an urgent public health problem. One of the biggest challenges that the area of oncology faces now is the issue of how to operationalize. No matter the disease area, proper prospective dose-finding at the outset, focusing on a broad strategy, and early biomarker work can be incredibly beneficial. Several examples, such as blood pressure reduction, lowering of HbA1c, and reduction in LDL cholesterol have been studied extensively and correlated to strongly with outcomes of interest that they are all now considered as surrogate endpoints. Therefore, the exploration of biomarkers at an early stage can be an incredibly critical area of investment with the potential for a high rate of return.</p><p>Oncology is a major therapeutic area in pharmaceutical R&amp;D with diverse therapeutic modalities and explosive advances in precision medicine. Drug development in oncology involves multi-dimensional optimization, where <i>Dose</i> is one of several dimensions (Figure 4), demanding inter-connected and iterative evidence generation with a <i>Totality of Evidence</i> mindset. When approaching the development of tailored precision medicines in cancers with diverse molecular footprints, dose selection cannot be approached as a <i>One Size Fits all</i> approach. Diversity in tumor molecular profile and host immunophenotype are important considerations in the discovery and development of precision oncology therapies at the right dose and dosing schedule for all patients. Advances in biomarker sciences and translational informatics are enabling deep characterization of the diversity of cancer biology and immunology across patient populations, with rapidly emerging applications of machine learning and artificial intelligence to harness such multimodal multidimensional data. These data represent invaluable inputs for the development of next-generation QSP platforms and their seamless integration in clinical drug development to identify the biological determinants of variability in clinical response and dosage requirements. Such integrated approaches have the potential to elevate the efficiency and fidelity of our current approaches to patient selection, combination partner selection, and dosage optimization.</p><p>As evident from the results of our 2023 ASCPT survey, randomized dose-ranging evaluation was not considered as an obligate requirement for dose optimization in all cases by about 60% of survey respondents. Indeed, examples exist where the application of biomarker-based and model-informed integrative approaches with a <i>Totality of Evidence</i> mindset have enabled confidence in the approved dosage of anticancer therapies, with many published success stories.<span><sup>26, 46-48</sup></span> In a <i>Totality of Evidence</i> approach, evidence is substantiated through the confidence gained from consistency across multiple approaches and data sources integrated in a mechanism-informed manner through modeling and simulation.<span><sup>49</sup></span> Such holistic integrative approaches are critically important when approaching the development of novel therapeutic modalities such as multi-specific biologics and cell therapies, where our survey indeed suggested that dose optimization will be most challenging. We are pleased to note steady progress in this area, with several recent publications across all three ASCPT journals highlighting advances in translational, quantitative, and clinical pharmacology applications for these emerging anticancer therapeutics.<span><sup>50-55</sup></span> As we learn from present and future real-life examples and continue to refine best practices in oncology dose optimization, we invite our readership and cross-sector practitioners to submit these advances for timely publication. We trust that the scientific discussion and rigorous debate that will ensue across our communities of practice, further facilitated by ASCPT's Networks and Communities, will go a long way in elevating patient-focused evidence generation for maximizing the benefit/ risk profile of next-generation oncology therapies.</p><p>No funding was received for this work.</p><p>The authors declared no competing interests in this work.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":null,"pages":null},"PeriodicalIF":3.1000,"publicationDate":"2024-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11179700/pdf/","citationCount":"0","resultStr":"{\"title\":\"Moving the needle for oncology dose optimization: A call for action\",\"authors\":\"Karthik Venkatakrishnan,&nbsp;Priya Jayachandran,&nbsp;Shirley K. Seo,&nbsp;Piet H. van der Graaf,&nbsp;John A. Wagner,&nbsp;Neeraj Gupta\",\"doi\":\"10.1002/psp4.13157\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Project Optimus is a major FDA initiative aimed at ensuring dose optimization in oncology drug development, moving away from the maximum tolerated dose paradigm and prospectively characterizing dose–response for efficacy and safety for patient-focused maximization of benefit versus risk.<span><sup>1-3</sup></span> Mitigating toxicities and enhancing overall benefit versus risk of oncology therapies necessitates dose optimization with commitment to evaluation of innovative dosing paradigms including individualized approaches, where appropriate. This requires the quantitative integration of pharmacological mechanism of action, efficacy, and safety in the context of associated population variability. The problem of dose optimization in the context of mechanism of action, cancer pathophysiology, and associated population variability sits neatly at the intersection of translational/ precision medicine and quantitative clinical pharmacology and is important to approach with a patient-focused mindset.</p><p>Forums convened on the topic of oncology dose optimization largely engage scientific leaders primarily working on oncology research and development, and cancer medicine. These include workshops organized by Friends of Cancer Research (FOCR),<span><sup>4</sup></span> American Society of Clinical Oncology (ASCO),<span><sup>5, 6</sup></span> American Association for Cancer Research (AACR),<span><sup>7, 8</sup></span> and the International Society of Pharmacometrics (ISoP)<span><sup>9</sup></span> in partnership with the US Food and Drugs Administration (FDA). Of note, some of these efforts have yielded seminal publications<span><sup>1, 2, 10-13</sup></span> and White Papers<span><sup>14</sup></span> offering initial recommendations, including availability of a Draft FDA guidance on the topic.<span><sup>15</sup></span> We posited that the American Society for Clinical Pharmacology and Therapeutics (ASCPT) – as a premier scientific and professional organization for clinical pharmacology and translational medicine – is optimally positioned to host a discussion of opportunities for our constituent disciplines (e.g., translational science, clinical pharmacology, pharmacometrics) to synergistically address this problem with a multi-disciplinary approach. To this end, a session was convened at the 2023 ASCPT Annual Meeting bringing together representative scientific leaders from the three scientific journals of the Society – <i>Clinical Pharmacology and Therapeutics</i> (<i>CPT</i>), <i>Clinical and Translational Science</i> (<i>CTS</i>), and <i>CPT: Pharmacometrics and Systems Pharmacology</i> (<i>PSP</i>). These scientific leaders, as at-large representatives of the disciplines of clinical pharmacology and translational medicine, were invited to bring forward their opinions and participate in a fireside chat to identify opportunities for moving the oncology dose optimization needle. This enabled engagement of a broad group of experts without requiring primary scientific or professional affiliation to the oncology therapeutic area, thereby maximizing diversity of opinion, out-of-the-box solutioning, and fresh perspectives that should help advance us beyond the current state. Ahead of the session at the Annual Meeting, a survey was launched to ASCPT members and meeting attendees to get our finger on the pulse of our Society's membership on issues faced in oncology dose optimization and provide substrate for the fireside chat with the expert panel. Herein, we present the findings from this ASCPT survey, and review the insights gained from this Annual Meeting session including recommendations for our scientific communities to join forces and drive progress.</p><p>A focused survey was developed and sent out in February 2023 to meeting attendees and broader ASCPT membership on the topic of the session, which consisted of six questions that were relevant to dose optimization (Data S1). The survey was open for 3 weeks and 65 respondents participated in the survey.</p><p>We were not only interested in understanding the background of survey respondents that may influence their feedback, but also various dose optimization approaches including challenges with various modalities. In response to our question about full time engagement with oncology R&amp;D, 58% of respondents were either not engaged or only had part time engagement with oncology R&amp;D. This suggested that survey feedback was from members with diverse backgrounds, as intended. Similarly, we were interested in understanding if strategies for dose optimization in other therapeutic areas are relevant for oncology therapies. 86% of respondents suggested that strategies from other therapeutic areas are indeed relevant to oncology.</p><p>Three questions focused on approaches applied for dose optimization – one on the utility of pharmacodynamic (PD) biomarkers, another one on quantitative approaches for dose selection and finally a question on study designs for dose optimization with a focus on randomization. 92% of responses suggest that PD biomarkers are at least useful. Clinical Exposure-response modeling (57%) followed by pharmacokinetic (PK)/PD modeling (28%) are most preferred approaches for selecting doses. Of note, 62% of respondents did not consider randomized dose-ranging evaluation as necessary for dose optimization, suggesting the value of application on a case-by-case approach leveraging the totality of evidence to optimize dose (Figure 1).</p><p>Given that oncology is a therapeutic area with a wide range of modalities from small molecules to cell therapies, we sought to understand the level of challenge associated with dose optimization in developing each of these modalities. Respondents noted that dose optimization for next-generation cytotoxic agents, small molecule targeted agents, and monoclonal antibodies is relatively straightforward with many historical examples to guide dose selection. However, dose optimization for antibody-drug conjugates was viewed to be moderately complex while newer modalities such as multi-specific biologics and cell therapies were considered very challenging with very few or no examples of dose optimization (Figure 2).</p><p>From a translational perspective, the focus of dose optimization is to find the <i>right</i> dose for patients as <i>swiftly</i> and <i>safely</i> as possible, buttressed by nonclinical and clinical translational data. Translational dose optimization doesn't always have to be complex. Goldstein et al.<span><sup>16</sup></span> describe a relatively simple concept for translational dose optimization for small molecule targeted oncology agents in the first-in-human setting. These suggestions can be implemented today. The approved doses of 25 targeted therapies were examined and the average free concentration at steady state (Css) was determined to be similar to the in vitro cell potency (half-maximal inhibitory concentration (IC50)). Furthermore, the authors propose a revised first-in-human trial design for next-generation targeted therapy in which dose cohort expansion is initiated at doses less than the maximum tolerated dose when there is evidence of clinical activity and Css exceeds a threshold informed by in vitro cell potency.</p><p>Ji et al.<span><sup>17</sup></span> describe another relatively straightforward approach to translational dose optimization in oncology. In this case, the drug is an inhibitor of Porcupine, a membrane-bound O-acyltransferase required for Wnt secretion. Wnt pathway is expressed in skin tissues; AXIN2 mRNA expression in skin is a robust and sensitive biomarker for the Wnt pathway. A predominant safety issue in this case is dysgeusia. The authors performed integrated population PK and exposure-response analyses of PD biomarker and safety data to determine the recommended dose for expansion, rather than the conventional maximum tolerated approach.</p><p>More complex approaches are also possible and have great utility, particularly for complex therapeutic modalities. Weddell et al.<span><sup>18</sup></span> describe an elegant mechanistic model that characterizes antibody drug conjugate (ADC) pharmacokinetics and tumor penetration by incorporating tumor growth inhibition via ADC binding radially across solid tumors. The model demonstrates that with low target expression, the potency of the payload should be increased. Furthermore, the model mechanistically links clinical response rates and relapse or resistance to ADC therapies, which could facilitate dose optimization. In another recent example, Susilo et al. leveraged a quantitative systems pharmacology (QSP) model of an anti-CD20/CD3 T-cell engaging bispecific antibody, mosunetuzumab, to account for different dosing regimens and inter-patient heterogeneity in the phase I study to identify biological determinants of clinical response and dose/exposure-response relationships using a novel QSP-derived digital twins approach.<span><sup>19</sup></span> Approaches of this nature raise opportunities for multi-dimensional optimization across the dimensions of dose, patient population, and combination partner – a challenge faced routinely in oncology drug development.</p><p>The value of new, innovative biomarkers in translational development is continuing to be realized. Recent examples indicate the emerging value of circulating tumor DNA (ctDNA).<span><sup>20, 21</sup></span> The translational utility of ctDNA, cancer cell DNA found in the bloodstream, is manifold, including detecting and diagnosing cancer, guiding tumor-specific treatment, monitoring treatment and remission. In the context of dose optimization, characterizing the underlying exposure-response relationship for on-treatment ctDNA dynamics to inform definition of a clinically active dose range represents an untapped opportunity. Another important innovation has been in the area of digital health technologies such as a proposed multi-domain, digital model for capturing functional status, and health-related quality of life in oncology,<span><sup>22</sup></span> which can be particularly relevant to realize the promise of Project Optimus aimed at dosage optimization for improved quality of life during long-term therapy.</p><p>ASCPT, clinical pharmacologists, and translational scientists have a key role in collaboration on dose optimization challenges and opportunities across different stakeholders. ASCPT membership straddles a variety of stakeholders including academics, industry, regulators, and others to help drive brainstorming and consensus formation. For example, Ji et al.,<span><sup>23</sup></span> reported on an ASCPT annual scientific meeting symposium. The authors describe a number of challenges observed before Project Optimus, including post-market dose-finding, continued use of traditional 3 + 3 designs, lack of characterization of chronic toxicity, and opportunities for adopting novel designs and testing more than one dose in phase 2/3 clinical trials. Oncology is one of the most innovative fields in science and yet there are only very few examples of value-added use of pharmacodynamic biomarkers and dose optimization. Cross-stakeholder work and Project Optimus are expected to drive the field to increased biomarker-based and model-informed solutions for oncology dose finding and optimization.</p><p>In their paper “The Future of Clinical Trial Design in Oncology,” Spreafico and co-workers from the Toronto Princess Margaret Cancer Centre<span><sup>24</sup></span> describe how therapeutic approaches in cancer drug discovery and development have shifted from traditional cytotoxic chemotherapy focused on histology-based targets to molecularly targeted and immune therapies in patient subsets stratified by biomarkers and other diagnostic precision tools. The authors argue that the classical clinical trial paradigm in oncology urgently needs to be transformed to ensure patients will benefit from this scientific revolution in a timely manner. In a wide-ranging call to action, they present a patient-centric framework for the next-generation oncology clinical trials, which maps out the journey of a trial participant as a dynamic and adaptive one continuously leveraging scientific and technological innovations to develop individualized therapeutic strategies. They conclude that “<i>The success of next-generation clinical trials will be based on the fundamental principles of acting locally to learn globally and treating participants individually to advance the field collectively</i>.” This speaks directly to the opportunity for clinical pharmacology to play a core role in this new paradigm, in particular with regard to dose optimization and individualization based on quantitative, model-informed approaches that integrate the totality knowledge and data of the drug, disease, and patient. An example of such an approach is QSP, which in a recent survey conducted by the ISoP was identified as an emerging key tool utilized by oncology drug developers for dose and dose regimen selection and optimization.<span><sup>25</sup></span> A recent example was presented by Li et al.,<span><sup>26</sup></span> who developed a mechanistic model to determine the recommended phase II dose (R2P2D) for epcoritamab, a CD3×CD20 bispecific antibody (bsAb). The authors justified this novel approach, which integrated preclinical, clinical PK, biomarker, tumor, and response data from the dose-escalation part of the phase I/II trial, on the basis that traditional dose/exposure-response modeling methods may not adequately predict the complex dose/exposure-response relationship for bsAbs. Therefore, trimer formation predicted by the mechanistic model instead of actual clinical measures was used to guide dose prediction.</p><p>Along the same lines, in a paper by Chelliah and representatives from a consortium of pharmaceutical companies,<span><sup>27</sup></span> the case is made that conventional, empirical pharmacometrics approaches do not fully capitalize on all the available biological and disease knowledge and that QSP models provide a more rational and better alternative to guide complex IO combination therapy development. Their proposal that “virtual patients” simulated by the QSP model under conditions that mimic the actual clinical trial should be added to the drug development paradigm is fully aligned with the earlier-mentioned call-to-action by Spreafico et al. outlined in <i>Figure 2</i> of their publication,<span><sup>24</sup></span> suggesting that the future of clinical trial design in oncology may already have arrived.</p><p>Poorly characterized dose and schedule may lead to selection of a dose that provides more toxicity without additional efficacy, severe toxicities that require a high rate of dose reductions or premature discontinuation and may result in missed opportunity for continued benefit from the drug. To optimize benefit versus risk with a patient-focused approach, there remain significant opportunities for model-based analyses to inform dosing regimen design that may sometimes involve non-static posology, with patient response or outcome-based dose adaptation to ensure individualized dosing for maximizing benefit versus risk.<span><sup>28, 29</sup></span></p><p>Project Optimus offers a pivotal opportunity to reform the oncology dosing paradigm using a robust quantitative clinical pharmacology framework.<span><sup>2, 3, 14, 30-33</sup></span> By integrating a model development lifecycle, Bayesian trial designs, and a learning-and-confirming mindset across the development spectrum, this framework may be used to prospectively guide dose optimization.</p><p>One of the main advantages of examining an oncology challenge as a non-oncologist is the ability to translate similar principles and successful examples from other therapeutic areas to oncology. These examples can aid in enriching a holistic approach toward solving longstanding problems. One clear correlate is in HIV drug discovery. In the 1980s, the average life expectancy following an AIDS diagnosis was approximately one year. And by the early 1990s, HIV was the leading cause of death among Americans aged 25 to 44. In many ways, much like with cancer, the urgency to save lives and need for therapeutics to control the epidemic fueled innovation and discovery. The beginning of that discovery phase did lead to some unsophisticated dosing – zidovudine was initially studied and approved at a dosage of 200 mg q4h, which caused severe anemia and neutropenia. However, more fine-tuning of the dose through clinical trials eventually led to its current dosage regimen of 300 mg twice daily. Several advancements along the way led to HIV infection largely being regarded as a chronic condition with near normal life expectancy for patients and a much improved quality of life. Some of these advancements included a deeper and continual understanding of the pharmacological mechanisms of antiretroviral agents, development of enhanced diagnostics, and acceptance of early biomarkers. When these approaches were deployed simultaneously, the result was a highly integrated, advanced methodology to solving an urgent public health problem. One of the biggest challenges that the area of oncology faces now is the issue of how to operationalize. No matter the disease area, proper prospective dose-finding at the outset, focusing on a broad strategy, and early biomarker work can be incredibly beneficial. Several examples, such as blood pressure reduction, lowering of HbA1c, and reduction in LDL cholesterol have been studied extensively and correlated to strongly with outcomes of interest that they are all now considered as surrogate endpoints. Therefore, the exploration of biomarkers at an early stage can be an incredibly critical area of investment with the potential for a high rate of return.</p><p>Oncology is a major therapeutic area in pharmaceutical R&amp;D with diverse therapeutic modalities and explosive advances in precision medicine. Drug development in oncology involves multi-dimensional optimization, where <i>Dose</i> is one of several dimensions (Figure 4), demanding inter-connected and iterative evidence generation with a <i>Totality of Evidence</i> mindset. When approaching the development of tailored precision medicines in cancers with diverse molecular footprints, dose selection cannot be approached as a <i>One Size Fits all</i> approach. Diversity in tumor molecular profile and host immunophenotype are important considerations in the discovery and development of precision oncology therapies at the right dose and dosing schedule for all patients. Advances in biomarker sciences and translational informatics are enabling deep characterization of the diversity of cancer biology and immunology across patient populations, with rapidly emerging applications of machine learning and artificial intelligence to harness such multimodal multidimensional data. These data represent invaluable inputs for the development of next-generation QSP platforms and their seamless integration in clinical drug development to identify the biological determinants of variability in clinical response and dosage requirements. Such integrated approaches have the potential to elevate the efficiency and fidelity of our current approaches to patient selection, combination partner selection, and dosage optimization.</p><p>As evident from the results of our 2023 ASCPT survey, randomized dose-ranging evaluation was not considered as an obligate requirement for dose optimization in all cases by about 60% of survey respondents. Indeed, examples exist where the application of biomarker-based and model-informed integrative approaches with a <i>Totality of Evidence</i> mindset have enabled confidence in the approved dosage of anticancer therapies, with many published success stories.<span><sup>26, 46-48</sup></span> In a <i>Totality of Evidence</i> approach, evidence is substantiated through the confidence gained from consistency across multiple approaches and data sources integrated in a mechanism-informed manner through modeling and simulation.<span><sup>49</sup></span> Such holistic integrative approaches are critically important when approaching the development of novel therapeutic modalities such as multi-specific biologics and cell therapies, where our survey indeed suggested that dose optimization will be most challenging. We are pleased to note steady progress in this area, with several recent publications across all three ASCPT journals highlighting advances in translational, quantitative, and clinical pharmacology applications for these emerging anticancer therapeutics.<span><sup>50-55</sup></span> As we learn from present and future real-life examples and continue to refine best practices in oncology dose optimization, we invite our readership and cross-sector practitioners to submit these advances for timely publication. We trust that the scientific discussion and rigorous debate that will ensue across our communities of practice, further facilitated by ASCPT's Networks and Communities, will go a long way in elevating patient-focused evidence generation for maximizing the benefit/ risk profile of next-generation oncology therapies.</p><p>No funding was received for this work.</p><p>The authors declared no competing interests in this work.</p>\",\"PeriodicalId\":10774,\"journal\":{\"name\":\"CPT: Pharmacometrics & Systems Pharmacology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.1000,\"publicationDate\":\"2024-05-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11179700/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"CPT: Pharmacometrics & Systems Pharmacology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/psp4.13157\",\"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/psp4.13157","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

2, 3, 14, 30-33 通过整合模型开发生命周期、贝叶斯试验设计以及整个开发过程中的学习与确认思维,该框架可用于前瞻性地指导剂量优化。作为非肿瘤学家研究肿瘤学难题的主要优势之一是能够将其他治疗领域的类似原则和成功案例转化为肿瘤学。这些例子有助于丰富解决长期问题的整体方法。一个明显的相关例子是艾滋病药物的发现。20 世纪 80 年代,艾滋病确诊后的平均预期寿命约为一年。到 20 世纪 90 年代初,艾滋病已成为 25 至 44 岁美国人的主要死因。在许多方面,与癌症一样,拯救生命的紧迫性和控制疫情的治疗需求推动了创新和发现。发现阶段的开始确实导致了一些不成熟的剂量--齐多夫定最初以 200 毫克 q4h 的剂量进行研究并获得批准,这导致了严重的贫血和中性粒细胞减少症。然而,通过临床试验对剂量进行更多的微调,最终确定了目前每天两次、每次 300 毫克的剂量方案。一路走来,HIV 感染在很大程度上被视为一种慢性疾病,患者的预期寿命接近正常,生活质量也大大提高。其中一些进步包括对抗逆转录病毒药物的药理机制有了更深入和持续的了解,开发出了更先进的诊断方法,早期生物标志物也得到了认可。当这些方法同时使用时,就会产生一种高度集成的先进方法来解决紧迫的公共卫生问题。肿瘤学领域目前面临的最大挑战之一是如何操作的问题。无论在哪个疾病领域,从一开始就进行适当的前瞻性剂量测定、专注于广泛的战略以及早期生物标志物工作都会带来巨大的益处。有几个例子,如降低血压、降低 HbA1c 和降低低密度脂蛋白胆固醇,已经得到了广泛的研究,并与相关结果密切相关,因此它们现在都被视为替代终点。因此,在早期阶段探索生物标志物可能是一个极其重要的投资领域,具有获得高回报率的潜力。肿瘤学是制药研发领域的一个主要治疗领域,其治疗方式多种多样,精准医疗取得了爆炸性的进展。肿瘤学药物开发涉及多维度优化,其中剂量是多个维度之一(图 4),需要以 "证据整体性 "的思维方式生成相互关联和迭代的证据。在针对具有不同分子足迹的癌症开发量身定制的精准药物时,剂量选择不能采用 "一刀切 "的方法。肿瘤分子特征和宿主免疫表型的多样性是发现和开发适合所有患者的精准肿瘤疗法的重要考虑因素。生物标志物科学和转化信息学的进步使得深入分析不同患者群体的癌症生物学和免疫学多样性成为可能,机器学习和人工智能的应用也在迅速兴起,以利用这些多模态多维数据。这些数据是开发下一代 QSP 平台的宝贵输入,可将其无缝集成到临床药物开发中,以确定临床反应和剂量要求差异的生物学决定因素。从我们 2023 年 ASCPT 调查的结果可以看出,约 60% 的调查对象并不认为随机剂量范围评估在所有情况下都是剂量优化的必要条件。26, 46-48 在 "证据的整体性 "方法中,通过建模和模拟,以机制知情的方式整合多种方法和数据源,通过一致性获得信心,从而证实证据。在开发新型治疗模式(如多特异性生物制剂和细胞疗法)时,这种整体综合方法至关重要。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Moving the needle for oncology dose optimization: A call for action

Project Optimus is a major FDA initiative aimed at ensuring dose optimization in oncology drug development, moving away from the maximum tolerated dose paradigm and prospectively characterizing dose–response for efficacy and safety for patient-focused maximization of benefit versus risk.1-3 Mitigating toxicities and enhancing overall benefit versus risk of oncology therapies necessitates dose optimization with commitment to evaluation of innovative dosing paradigms including individualized approaches, where appropriate. This requires the quantitative integration of pharmacological mechanism of action, efficacy, and safety in the context of associated population variability. The problem of dose optimization in the context of mechanism of action, cancer pathophysiology, and associated population variability sits neatly at the intersection of translational/ precision medicine and quantitative clinical pharmacology and is important to approach with a patient-focused mindset.

Forums convened on the topic of oncology dose optimization largely engage scientific leaders primarily working on oncology research and development, and cancer medicine. These include workshops organized by Friends of Cancer Research (FOCR),4 American Society of Clinical Oncology (ASCO),5, 6 American Association for Cancer Research (AACR),7, 8 and the International Society of Pharmacometrics (ISoP)9 in partnership with the US Food and Drugs Administration (FDA). Of note, some of these efforts have yielded seminal publications1, 2, 10-13 and White Papers14 offering initial recommendations, including availability of a Draft FDA guidance on the topic.15 We posited that the American Society for Clinical Pharmacology and Therapeutics (ASCPT) – as a premier scientific and professional organization for clinical pharmacology and translational medicine – is optimally positioned to host a discussion of opportunities for our constituent disciplines (e.g., translational science, clinical pharmacology, pharmacometrics) to synergistically address this problem with a multi-disciplinary approach. To this end, a session was convened at the 2023 ASCPT Annual Meeting bringing together representative scientific leaders from the three scientific journals of the Society – Clinical Pharmacology and Therapeutics (CPT), Clinical and Translational Science (CTS), and CPT: Pharmacometrics and Systems Pharmacology (PSP). These scientific leaders, as at-large representatives of the disciplines of clinical pharmacology and translational medicine, were invited to bring forward their opinions and participate in a fireside chat to identify opportunities for moving the oncology dose optimization needle. This enabled engagement of a broad group of experts without requiring primary scientific or professional affiliation to the oncology therapeutic area, thereby maximizing diversity of opinion, out-of-the-box solutioning, and fresh perspectives that should help advance us beyond the current state. Ahead of the session at the Annual Meeting, a survey was launched to ASCPT members and meeting attendees to get our finger on the pulse of our Society's membership on issues faced in oncology dose optimization and provide substrate for the fireside chat with the expert panel. Herein, we present the findings from this ASCPT survey, and review the insights gained from this Annual Meeting session including recommendations for our scientific communities to join forces and drive progress.

A focused survey was developed and sent out in February 2023 to meeting attendees and broader ASCPT membership on the topic of the session, which consisted of six questions that were relevant to dose optimization (Data S1). The survey was open for 3 weeks and 65 respondents participated in the survey.

We were not only interested in understanding the background of survey respondents that may influence their feedback, but also various dose optimization approaches including challenges with various modalities. In response to our question about full time engagement with oncology R&D, 58% of respondents were either not engaged or only had part time engagement with oncology R&D. This suggested that survey feedback was from members with diverse backgrounds, as intended. Similarly, we were interested in understanding if strategies for dose optimization in other therapeutic areas are relevant for oncology therapies. 86% of respondents suggested that strategies from other therapeutic areas are indeed relevant to oncology.

Three questions focused on approaches applied for dose optimization – one on the utility of pharmacodynamic (PD) biomarkers, another one on quantitative approaches for dose selection and finally a question on study designs for dose optimization with a focus on randomization. 92% of responses suggest that PD biomarkers are at least useful. Clinical Exposure-response modeling (57%) followed by pharmacokinetic (PK)/PD modeling (28%) are most preferred approaches for selecting doses. Of note, 62% of respondents did not consider randomized dose-ranging evaluation as necessary for dose optimization, suggesting the value of application on a case-by-case approach leveraging the totality of evidence to optimize dose (Figure 1).

Given that oncology is a therapeutic area with a wide range of modalities from small molecules to cell therapies, we sought to understand the level of challenge associated with dose optimization in developing each of these modalities. Respondents noted that dose optimization for next-generation cytotoxic agents, small molecule targeted agents, and monoclonal antibodies is relatively straightforward with many historical examples to guide dose selection. However, dose optimization for antibody-drug conjugates was viewed to be moderately complex while newer modalities such as multi-specific biologics and cell therapies were considered very challenging with very few or no examples of dose optimization (Figure 2).

From a translational perspective, the focus of dose optimization is to find the right dose for patients as swiftly and safely as possible, buttressed by nonclinical and clinical translational data. Translational dose optimization doesn't always have to be complex. Goldstein et al.16 describe a relatively simple concept for translational dose optimization for small molecule targeted oncology agents in the first-in-human setting. These suggestions can be implemented today. The approved doses of 25 targeted therapies were examined and the average free concentration at steady state (Css) was determined to be similar to the in vitro cell potency (half-maximal inhibitory concentration (IC50)). Furthermore, the authors propose a revised first-in-human trial design for next-generation targeted therapy in which dose cohort expansion is initiated at doses less than the maximum tolerated dose when there is evidence of clinical activity and Css exceeds a threshold informed by in vitro cell potency.

Ji et al.17 describe another relatively straightforward approach to translational dose optimization in oncology. In this case, the drug is an inhibitor of Porcupine, a membrane-bound O-acyltransferase required for Wnt secretion. Wnt pathway is expressed in skin tissues; AXIN2 mRNA expression in skin is a robust and sensitive biomarker for the Wnt pathway. A predominant safety issue in this case is dysgeusia. The authors performed integrated population PK and exposure-response analyses of PD biomarker and safety data to determine the recommended dose for expansion, rather than the conventional maximum tolerated approach.

More complex approaches are also possible and have great utility, particularly for complex therapeutic modalities. Weddell et al.18 describe an elegant mechanistic model that characterizes antibody drug conjugate (ADC) pharmacokinetics and tumor penetration by incorporating tumor growth inhibition via ADC binding radially across solid tumors. The model demonstrates that with low target expression, the potency of the payload should be increased. Furthermore, the model mechanistically links clinical response rates and relapse or resistance to ADC therapies, which could facilitate dose optimization. In another recent example, Susilo et al. leveraged a quantitative systems pharmacology (QSP) model of an anti-CD20/CD3 T-cell engaging bispecific antibody, mosunetuzumab, to account for different dosing regimens and inter-patient heterogeneity in the phase I study to identify biological determinants of clinical response and dose/exposure-response relationships using a novel QSP-derived digital twins approach.19 Approaches of this nature raise opportunities for multi-dimensional optimization across the dimensions of dose, patient population, and combination partner – a challenge faced routinely in oncology drug development.

The value of new, innovative biomarkers in translational development is continuing to be realized. Recent examples indicate the emerging value of circulating tumor DNA (ctDNA).20, 21 The translational utility of ctDNA, cancer cell DNA found in the bloodstream, is manifold, including detecting and diagnosing cancer, guiding tumor-specific treatment, monitoring treatment and remission. In the context of dose optimization, characterizing the underlying exposure-response relationship for on-treatment ctDNA dynamics to inform definition of a clinically active dose range represents an untapped opportunity. Another important innovation has been in the area of digital health technologies such as a proposed multi-domain, digital model for capturing functional status, and health-related quality of life in oncology,22 which can be particularly relevant to realize the promise of Project Optimus aimed at dosage optimization for improved quality of life during long-term therapy.

ASCPT, clinical pharmacologists, and translational scientists have a key role in collaboration on dose optimization challenges and opportunities across different stakeholders. ASCPT membership straddles a variety of stakeholders including academics, industry, regulators, and others to help drive brainstorming and consensus formation. For example, Ji et al.,23 reported on an ASCPT annual scientific meeting symposium. The authors describe a number of challenges observed before Project Optimus, including post-market dose-finding, continued use of traditional 3 + 3 designs, lack of characterization of chronic toxicity, and opportunities for adopting novel designs and testing more than one dose in phase 2/3 clinical trials. Oncology is one of the most innovative fields in science and yet there are only very few examples of value-added use of pharmacodynamic biomarkers and dose optimization. Cross-stakeholder work and Project Optimus are expected to drive the field to increased biomarker-based and model-informed solutions for oncology dose finding and optimization.

In their paper “The Future of Clinical Trial Design in Oncology,” Spreafico and co-workers from the Toronto Princess Margaret Cancer Centre24 describe how therapeutic approaches in cancer drug discovery and development have shifted from traditional cytotoxic chemotherapy focused on histology-based targets to molecularly targeted and immune therapies in patient subsets stratified by biomarkers and other diagnostic precision tools. The authors argue that the classical clinical trial paradigm in oncology urgently needs to be transformed to ensure patients will benefit from this scientific revolution in a timely manner. In a wide-ranging call to action, they present a patient-centric framework for the next-generation oncology clinical trials, which maps out the journey of a trial participant as a dynamic and adaptive one continuously leveraging scientific and technological innovations to develop individualized therapeutic strategies. They conclude that “The success of next-generation clinical trials will be based on the fundamental principles of acting locally to learn globally and treating participants individually to advance the field collectively.” This speaks directly to the opportunity for clinical pharmacology to play a core role in this new paradigm, in particular with regard to dose optimization and individualization based on quantitative, model-informed approaches that integrate the totality knowledge and data of the drug, disease, and patient. An example of such an approach is QSP, which in a recent survey conducted by the ISoP was identified as an emerging key tool utilized by oncology drug developers for dose and dose regimen selection and optimization.25 A recent example was presented by Li et al.,26 who developed a mechanistic model to determine the recommended phase II dose (R2P2D) for epcoritamab, a CD3×CD20 bispecific antibody (bsAb). The authors justified this novel approach, which integrated preclinical, clinical PK, biomarker, tumor, and response data from the dose-escalation part of the phase I/II trial, on the basis that traditional dose/exposure-response modeling methods may not adequately predict the complex dose/exposure-response relationship for bsAbs. Therefore, trimer formation predicted by the mechanistic model instead of actual clinical measures was used to guide dose prediction.

Along the same lines, in a paper by Chelliah and representatives from a consortium of pharmaceutical companies,27 the case is made that conventional, empirical pharmacometrics approaches do not fully capitalize on all the available biological and disease knowledge and that QSP models provide a more rational and better alternative to guide complex IO combination therapy development. Their proposal that “virtual patients” simulated by the QSP model under conditions that mimic the actual clinical trial should be added to the drug development paradigm is fully aligned with the earlier-mentioned call-to-action by Spreafico et al. outlined in Figure 2 of their publication,24 suggesting that the future of clinical trial design in oncology may already have arrived.

Poorly characterized dose and schedule may lead to selection of a dose that provides more toxicity without additional efficacy, severe toxicities that require a high rate of dose reductions or premature discontinuation and may result in missed opportunity for continued benefit from the drug. To optimize benefit versus risk with a patient-focused approach, there remain significant opportunities for model-based analyses to inform dosing regimen design that may sometimes involve non-static posology, with patient response or outcome-based dose adaptation to ensure individualized dosing for maximizing benefit versus risk.28, 29

Project Optimus offers a pivotal opportunity to reform the oncology dosing paradigm using a robust quantitative clinical pharmacology framework.2, 3, 14, 30-33 By integrating a model development lifecycle, Bayesian trial designs, and a learning-and-confirming mindset across the development spectrum, this framework may be used to prospectively guide dose optimization.

One of the main advantages of examining an oncology challenge as a non-oncologist is the ability to translate similar principles and successful examples from other therapeutic areas to oncology. These examples can aid in enriching a holistic approach toward solving longstanding problems. One clear correlate is in HIV drug discovery. In the 1980s, the average life expectancy following an AIDS diagnosis was approximately one year. And by the early 1990s, HIV was the leading cause of death among Americans aged 25 to 44. In many ways, much like with cancer, the urgency to save lives and need for therapeutics to control the epidemic fueled innovation and discovery. The beginning of that discovery phase did lead to some unsophisticated dosing – zidovudine was initially studied and approved at a dosage of 200 mg q4h, which caused severe anemia and neutropenia. However, more fine-tuning of the dose through clinical trials eventually led to its current dosage regimen of 300 mg twice daily. Several advancements along the way led to HIV infection largely being regarded as a chronic condition with near normal life expectancy for patients and a much improved quality of life. Some of these advancements included a deeper and continual understanding of the pharmacological mechanisms of antiretroviral agents, development of enhanced diagnostics, and acceptance of early biomarkers. When these approaches were deployed simultaneously, the result was a highly integrated, advanced methodology to solving an urgent public health problem. One of the biggest challenges that the area of oncology faces now is the issue of how to operationalize. No matter the disease area, proper prospective dose-finding at the outset, focusing on a broad strategy, and early biomarker work can be incredibly beneficial. Several examples, such as blood pressure reduction, lowering of HbA1c, and reduction in LDL cholesterol have been studied extensively and correlated to strongly with outcomes of interest that they are all now considered as surrogate endpoints. Therefore, the exploration of biomarkers at an early stage can be an incredibly critical area of investment with the potential for a high rate of return.

Oncology is a major therapeutic area in pharmaceutical R&D with diverse therapeutic modalities and explosive advances in precision medicine. Drug development in oncology involves multi-dimensional optimization, where Dose is one of several dimensions (Figure 4), demanding inter-connected and iterative evidence generation with a Totality of Evidence mindset. When approaching the development of tailored precision medicines in cancers with diverse molecular footprints, dose selection cannot be approached as a One Size Fits all approach. Diversity in tumor molecular profile and host immunophenotype are important considerations in the discovery and development of precision oncology therapies at the right dose and dosing schedule for all patients. Advances in biomarker sciences and translational informatics are enabling deep characterization of the diversity of cancer biology and immunology across patient populations, with rapidly emerging applications of machine learning and artificial intelligence to harness such multimodal multidimensional data. These data represent invaluable inputs for the development of next-generation QSP platforms and their seamless integration in clinical drug development to identify the biological determinants of variability in clinical response and dosage requirements. Such integrated approaches have the potential to elevate the efficiency and fidelity of our current approaches to patient selection, combination partner selection, and dosage optimization.

As evident from the results of our 2023 ASCPT survey, randomized dose-ranging evaluation was not considered as an obligate requirement for dose optimization in all cases by about 60% of survey respondents. Indeed, examples exist where the application of biomarker-based and model-informed integrative approaches with a Totality of Evidence mindset have enabled confidence in the approved dosage of anticancer therapies, with many published success stories.26, 46-48 In a Totality of Evidence approach, evidence is substantiated through the confidence gained from consistency across multiple approaches and data sources integrated in a mechanism-informed manner through modeling and simulation.49 Such holistic integrative approaches are critically important when approaching the development of novel therapeutic modalities such as multi-specific biologics and cell therapies, where our survey indeed suggested that dose optimization will be most challenging. We are pleased to note steady progress in this area, with several recent publications across all three ASCPT journals highlighting advances in translational, quantitative, and clinical pharmacology applications for these emerging anticancer therapeutics.50-55 As we learn from present and future real-life examples and continue to refine best practices in oncology dose optimization, we invite our readership and cross-sector practitioners to submit these advances for timely publication. We trust that the scientific discussion and rigorous debate that will ensue across our communities of practice, further facilitated by ASCPT's Networks and Communities, will go a long way in elevating patient-focused evidence generation for maximizing the benefit/ risk profile of next-generation oncology therapies.

No funding was received for this work.

The authors declared no competing interests in this work.

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
期刊最新文献
A model-informed clinical trial simulation tool with a graphical user interface for Duchenne muscular dystrophy. Physiology-based pharmacokinetic model with relative transcriptomics to evaluate tissue distribution and receptor occupancy of anifrolumab. Interim analysis, a tool to enhance efficiency of pharmacokinetic studies: Pharmacokinetics of rifampicin in lactating mother-infant pairs. PBPK modeling: What is the role of CYP3A4 expression in the gastrointestinal tract to accurately predict first-pass metabolism? Immunostimulatory/Immunodynamic model of mRNA-1273 to guide pediatric vaccine dose selection.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1