密码破解还是蓝色代码?评估sotorasib在转移性结直肠癌中的生命体征

IF 5.1 2区 医学 Q1 ONCOLOGY
Cancer Pub Date : 2025-08-19 DOI:10.1002/cncr.70056
Samuel X. Stevens MBBS (Hons), B App Sc, M Bioethics, FRACP, Bishal Gyawali MD, PhD, FASCO
{"title":"密码破解还是蓝色代码?评估sotorasib在转移性结直肠癌中的生命体征","authors":"Samuel X. Stevens MBBS (Hons), B App Sc, M Bioethics, FRACP,&nbsp;Bishal Gyawali MD, PhD, FASCO","doi":"10.1002/cncr.70056","DOIUrl":null,"url":null,"abstract":"<p>The ability to therapeutically target Kirsten rat sarcoma viral oncogene homolog (KRAS)–long considered an <i>undruggable</i> oncogenic driver of nonsmall cell lung cancer (NSCLC) and gastrointestinal (GI) cancer—is one of the most important biomedical discoveries of the past decade. Therefore, sotorasib, the first KRAS glycine-to-cysteine substitution at codon 12 (G12C) mutation inhibitor to reach the clinic was heralded as a triumph of precision oncology.<span><sup>1</sup></span> Yet the path of sotorasib from preclinical discovery to US Food and Drug Administration (FDA) approval offers a cautionary tale for a field increasingly immersed in molecularly targeted therapies. Although heralded as a success of precision oncology, precision oncology is not merely the right drug for the right target. It also entails the right dose at the right time for the right duration, to which one could arguably add right reporting. However, after reading the results from the recent CodeBreak 300 trial of sotorasib in colorectal cancer (ClinicalTrials.gov identifier NCT05198934), we believe sotorasib has fallen short in many domains of this definition. The case of sotorasib also highlights how regulators have not encouraged the conditions needed to deliver the real promise of precision oncology to patients.</p><p>CodeBreaK 300 is a phase 3 trial evaluating the efficacy and safety of sotorasib in combination with panitumumab versus treatment of investigator's choice (TIC) in patients with advanced colorectal cancer harboring a KRAS G12C mutation who progressed on chemotherapies.<span><sup>2</sup></span> Although the included patient population and choice of control arm in this trial were appropriate, a striking feature of this trial is the uncertainty about the appropriate dose level for sotorasib. Doses for registration trials are typically established in early phase clinical trials to minimize the number of participants exposed to potentially ineffective or toxic dosages. Thus it is notable that this phase 3 trial appears to conduct a dose-finding exercise, with separate arms for the 240-mg and 960-mg doses, respectively. The justification of <i>nonlinear pharmacokinetics</i>, although not untrue, is perhaps only a part of the story.</p><p>Having shown promise in early phase trials, sotorasib received FDA Accelerated Approval for use in KRAS G12C mutant NSCLC in 2021. However, the FDA noted that preliminary trials were “hampered by a lack of robust dose exploration,” mandating further exploration in postmarketing studies<span><sup>3</sup></span> and highlighting the tension between speed and certainty in drug development. Appropriately, the 960-mg and 240-mg dosages underwent further evaluation, with no statistically significant difference in efficacy but with higher treatment-related adverse events for the 960-mg dose.<span><sup>4</sup></span> Although published in 2024, evidence from patent applications suggests that this was known to the sponsor as early as March 2022, before CodeBreaK 300 commenced enrolling.<span><sup>5</sup></span> Although the trial appears to address these uncertainties, the hierarchical statistical approach—testing 960 mg versus TIC followed by 240 mg versus TIC—prohibits inferential comparisons between both doses of sotorasib, further clouding the dose-selection issue.</p><p>These design decisions have moral, clinical, and economic implications. Study investigators have a moral imperative to minimize research participants' exposure to harm: this is a common tenet of Good Clinical Practice. Although a trial designed to directly compare efficacy across both drug dose levels would need to be much larger—indeed, approximately twice the number of participants would be required—this <i>robust dose exploration</i> appears to be exactly what the FDA had previously asked for.<span><sup>3</sup></span> The choice to spend participants' <i>time, effort, and welfare</i>, as Kimmelman suggests, on a noninformative dose comparison, in our view, is morally questionable—particularly considering suggestions that the sponsor possessed information unsettling equipoise about dosage.<span><sup>6, 7</sup></span> If this information was not yet known, CodeBreak 300 should not have proceeded without adequately resolving uncertainties about optimal dosing.</p><p>Second, the chosen design raises concerns about the intent behind dose selection: by prioritizing the 960-mg dose for hierarchical testing, the trial may have supported regulatory decisions favoring higher cost dosing with no significant clinical benefit, greater potential harms, and higher economic burden for the health system. Finally, inflating enrolment to include a third arm incurred a significant opportunity cost, reducing the statistical power available to answer more clinically meaningful questions regarding overall survival (OS). Consequently, the trial was underpowered to detect differences in patient-centered outcomes despite enrolling a sizeable population of participants with this relatively infrequent mutation.<span><sup>8</sup></span> A more patient-centric approach would have been either to do the hierarchical testing but in the opposite direction-that is, first test 240mg and then test 960mg.</p><p>The primary end point of CodeBreak 300 was met, with sotorasib 960 mg–panitumumab showing a statistically significant progression-free survival (PFS) advantage over the control arm (hazard ratio [HR], 0.48; 95% confidence interval [CI], 0.30–0.78; <i>p</i> = .005), although the 240-mg dose failed to do so (HR, 0.59; 95% CI, 0.37–0.95; <i>p</i> = .036; negative because of alpha splitting).<span><sup>9, 10</sup></span> The higher dose also led to a higher overall response rate (ORR; 26.4% vs. 5.7%). Both of these PFS and ORR outcomes must have been factored into the FDA decision to approve the 960-mg dose. These comparisons contrast with prior dose-finding studies, which demonstrated more equivocal differences between the 240-mg and 960-mg doses.<span><sup>4</sup></span> Notwithstanding these differences in ORR and PFS outcomes between the two doses versus the control arm—the two doses were not compared against each other for any outcome—they do not answer the questions that are most important to patients, clinicians, and regulators who must make challenging therapeutic decisions for a last-line therapy, which is whether sotorasib improves survival and quality of life.</p><p>In this trial, PFS was chosen as the primary end point, with OS and health-related quality of life relegated to secondary end points. CodeBreak 300 was not even powered to detect differences in OS between treatment arms. This is concerning for several reasons. First, OS and quality of life are consistently identified as being of primal importance to patients living with advanced solid tumors.<span><sup>11-16</sup></span> Second, PFS is not a valid surrogate measure of OS in second-line and later line colorectal cancer trials.<span><sup>17, 18</sup></span> We recognize that there are situations in which intermediate end points like PFS may be justifiable, for example, in diseases with long survival or when prior surrogacy has been established. However, these conditions are not met in this last-line trial population.<span><sup>18</sup></span> Usually, the appeal of PFS is shortening the time to results readout. However, for last-line therapy trials, OS results are usually available quickly, as in this case, I which OS results were available with just 6 months of additional follow-up after PFS results were available.<span><sup>2</sup></span> Notably, the control arms of this trial—trifluridine/tipiracil or regorafenib, as well as other recently approved drugs in this space, such as trifluridine/tipiracil plus bevacizumab or fruquitinib—were all approved based on OS improvement.<span><sup>19, 20</sup></span> Indeed, no other drugs have been approved for last-line colorectal cancer based on PFS alone. The allure of the biology of targeting a novel mutation should not compensate for the need to improve OS in this group of patients. It is striking that, in CodeBreak 200, the last-line trial in lung cancer, the initial primary end point of OS was subsequently changed to PFS according to <i>regulatory feedback</i> to limit patients exposed in the control arm.<span><sup>21</sup></span> This shows that even regulators are not protected from the allure of precision oncology.</p><p>Despite the initial promising results of this trial, the fundamental design flaws of CodeBreak 300 leave it fighting for life amidst the competitive landscape of precision oncology trials. However, it is bias in reporting and approval of the drug that ultimately triggers a <i>code blue</i> for sotorasib in advanced colorectal cancer. Despite what could be otherwise described as an exploratory analysis of survival outcomes, the conclusions of this study assert that sotorasib 960 mg and panitumumab constitutes a <i>new standard of care</i> for patients with KRAS G12C mutations. Speculation about potential survival gains from a nonstatistically significant, underpowered analysis should not be misrepresented as clinical insight: <i>even if it is not statistically different, an improvement in OS could be particularly meaningful</i>. Such statements do not carry evidentiary weight and risk misleading clinicians and patients alike.</p><p>We are quite surprised that the FDA accepted the trial with all of these flaws and approved the drug for this indication.<span><sup>22</sup></span> Unlike the NSCLC indication, the colorectal cancer indication was granted regular approval—not even Accelerated Approval with a safety net of the requirement to confirm clinical benefit in the future. The FDA approved the 960-mg dose, although there were no statistical comparisons between the two dose levels—OS outcomes for both doses were similarly unimproved against the control arm, with HRs of 0.70 (95% CI, 0.41–1.18) and 0.83 (95% CI, 0.49–1.39), respectively, and <i>p</i> values of .2 and .5, respectively.<span><sup>2</sup></span> Granting full regular approval based on PFS for a last-line therapy trial in patients with a poor prognosis, despite negative OS, no quality-of-life improvement, and at a potentially higher dose with increased toxicities, is inexplicable. Sadly, after two large trials of this agent, the field—and, more importantly, our patients—are left with fleeting signals of efficacy, no survival benefit, and a cautionary reminder of the need to temper our excitement for innovation with the need for epistemic humility. Poorly designed or underpowered trials like CodeBreak 300 miss important opportunities to answer critical questions, at great human and economic cost. When regulators rubberstamp these trials with approvals, there is even less incentive to do things the right way.</p><p>To realize the full potential of precision oncology, we must move beyond molecular enthusiasm to methodological discipline. Only rigorously designed trials can separate promising ideas from proven benefit. Although there may yet be a place for sotorasib in the treatment of colorectal cancer, CodeBreak 300 does not provide sufficient evidence that its' time has arrived. The new FDA has signaled that they will require OS data for patients in later lines of therapy.<span><sup>23, 24</sup></span> We hope that this stance results in positive change. Rewarding drugs that fail to improve survival and that seek registration at higher doses—despite the exciting biology and allure of precision oncology—will impede innovation that matters to patients.</p><p><b>Samuel X. Stevens:</b> Conceptualization, writing–review and editing, and writing–original draft. <b>Bishal Gyawali:</b> Conceptualization and writing–review and editing.</p><p>Bishal Gyawali reports personal/consulting fees from Vivio Health outside the submitted work. Samuel X. Stevens disclosed no conflicts of interest.</p>","PeriodicalId":138,"journal":{"name":"Cancer","volume":"131 17","pages":""},"PeriodicalIF":5.1000,"publicationDate":"2025-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://acsjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.70056","citationCount":"0","resultStr":"{\"title\":\"CodeBreaK or code blue? Assessing sotorasib's vital signs in metastatic colorectal cancer\",\"authors\":\"Samuel X. Stevens MBBS (Hons), B App Sc, M Bioethics, FRACP,&nbsp;Bishal Gyawali MD, PhD, FASCO\",\"doi\":\"10.1002/cncr.70056\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>The ability to therapeutically target Kirsten rat sarcoma viral oncogene homolog (KRAS)–long considered an <i>undruggable</i> oncogenic driver of nonsmall cell lung cancer (NSCLC) and gastrointestinal (GI) cancer—is one of the most important biomedical discoveries of the past decade. Therefore, sotorasib, the first KRAS glycine-to-cysteine substitution at codon 12 (G12C) mutation inhibitor to reach the clinic was heralded as a triumph of precision oncology.<span><sup>1</sup></span> Yet the path of sotorasib from preclinical discovery to US Food and Drug Administration (FDA) approval offers a cautionary tale for a field increasingly immersed in molecularly targeted therapies. Although heralded as a success of precision oncology, precision oncology is not merely the right drug for the right target. It also entails the right dose at the right time for the right duration, to which one could arguably add right reporting. However, after reading the results from the recent CodeBreak 300 trial of sotorasib in colorectal cancer (ClinicalTrials.gov identifier NCT05198934), we believe sotorasib has fallen short in many domains of this definition. The case of sotorasib also highlights how regulators have not encouraged the conditions needed to deliver the real promise of precision oncology to patients.</p><p>CodeBreaK 300 is a phase 3 trial evaluating the efficacy and safety of sotorasib in combination with panitumumab versus treatment of investigator's choice (TIC) in patients with advanced colorectal cancer harboring a KRAS G12C mutation who progressed on chemotherapies.<span><sup>2</sup></span> Although the included patient population and choice of control arm in this trial were appropriate, a striking feature of this trial is the uncertainty about the appropriate dose level for sotorasib. Doses for registration trials are typically established in early phase clinical trials to minimize the number of participants exposed to potentially ineffective or toxic dosages. Thus it is notable that this phase 3 trial appears to conduct a dose-finding exercise, with separate arms for the 240-mg and 960-mg doses, respectively. The justification of <i>nonlinear pharmacokinetics</i>, although not untrue, is perhaps only a part of the story.</p><p>Having shown promise in early phase trials, sotorasib received FDA Accelerated Approval for use in KRAS G12C mutant NSCLC in 2021. However, the FDA noted that preliminary trials were “hampered by a lack of robust dose exploration,” mandating further exploration in postmarketing studies<span><sup>3</sup></span> and highlighting the tension between speed and certainty in drug development. Appropriately, the 960-mg and 240-mg dosages underwent further evaluation, with no statistically significant difference in efficacy but with higher treatment-related adverse events for the 960-mg dose.<span><sup>4</sup></span> Although published in 2024, evidence from patent applications suggests that this was known to the sponsor as early as March 2022, before CodeBreaK 300 commenced enrolling.<span><sup>5</sup></span> Although the trial appears to address these uncertainties, the hierarchical statistical approach—testing 960 mg versus TIC followed by 240 mg versus TIC—prohibits inferential comparisons between both doses of sotorasib, further clouding the dose-selection issue.</p><p>These design decisions have moral, clinical, and economic implications. Study investigators have a moral imperative to minimize research participants' exposure to harm: this is a common tenet of Good Clinical Practice. Although a trial designed to directly compare efficacy across both drug dose levels would need to be much larger—indeed, approximately twice the number of participants would be required—this <i>robust dose exploration</i> appears to be exactly what the FDA had previously asked for.<span><sup>3</sup></span> The choice to spend participants' <i>time, effort, and welfare</i>, as Kimmelman suggests, on a noninformative dose comparison, in our view, is morally questionable—particularly considering suggestions that the sponsor possessed information unsettling equipoise about dosage.<span><sup>6, 7</sup></span> If this information was not yet known, CodeBreak 300 should not have proceeded without adequately resolving uncertainties about optimal dosing.</p><p>Second, the chosen design raises concerns about the intent behind dose selection: by prioritizing the 960-mg dose for hierarchical testing, the trial may have supported regulatory decisions favoring higher cost dosing with no significant clinical benefit, greater potential harms, and higher economic burden for the health system. Finally, inflating enrolment to include a third arm incurred a significant opportunity cost, reducing the statistical power available to answer more clinically meaningful questions regarding overall survival (OS). Consequently, the trial was underpowered to detect differences in patient-centered outcomes despite enrolling a sizeable population of participants with this relatively infrequent mutation.<span><sup>8</sup></span> A more patient-centric approach would have been either to do the hierarchical testing but in the opposite direction-that is, first test 240mg and then test 960mg.</p><p>The primary end point of CodeBreak 300 was met, with sotorasib 960 mg–panitumumab showing a statistically significant progression-free survival (PFS) advantage over the control arm (hazard ratio [HR], 0.48; 95% confidence interval [CI], 0.30–0.78; <i>p</i> = .005), although the 240-mg dose failed to do so (HR, 0.59; 95% CI, 0.37–0.95; <i>p</i> = .036; negative because of alpha splitting).<span><sup>9, 10</sup></span> The higher dose also led to a higher overall response rate (ORR; 26.4% vs. 5.7%). Both of these PFS and ORR outcomes must have been factored into the FDA decision to approve the 960-mg dose. These comparisons contrast with prior dose-finding studies, which demonstrated more equivocal differences between the 240-mg and 960-mg doses.<span><sup>4</sup></span> Notwithstanding these differences in ORR and PFS outcomes between the two doses versus the control arm—the two doses were not compared against each other for any outcome—they do not answer the questions that are most important to patients, clinicians, and regulators who must make challenging therapeutic decisions for a last-line therapy, which is whether sotorasib improves survival and quality of life.</p><p>In this trial, PFS was chosen as the primary end point, with OS and health-related quality of life relegated to secondary end points. CodeBreak 300 was not even powered to detect differences in OS between treatment arms. This is concerning for several reasons. First, OS and quality of life are consistently identified as being of primal importance to patients living with advanced solid tumors.<span><sup>11-16</sup></span> Second, PFS is not a valid surrogate measure of OS in second-line and later line colorectal cancer trials.<span><sup>17, 18</sup></span> We recognize that there are situations in which intermediate end points like PFS may be justifiable, for example, in diseases with long survival or when prior surrogacy has been established. However, these conditions are not met in this last-line trial population.<span><sup>18</sup></span> Usually, the appeal of PFS is shortening the time to results readout. However, for last-line therapy trials, OS results are usually available quickly, as in this case, I which OS results were available with just 6 months of additional follow-up after PFS results were available.<span><sup>2</sup></span> Notably, the control arms of this trial—trifluridine/tipiracil or regorafenib, as well as other recently approved drugs in this space, such as trifluridine/tipiracil plus bevacizumab or fruquitinib—were all approved based on OS improvement.<span><sup>19, 20</sup></span> Indeed, no other drugs have been approved for last-line colorectal cancer based on PFS alone. The allure of the biology of targeting a novel mutation should not compensate for the need to improve OS in this group of patients. It is striking that, in CodeBreak 200, the last-line trial in lung cancer, the initial primary end point of OS was subsequently changed to PFS according to <i>regulatory feedback</i> to limit patients exposed in the control arm.<span><sup>21</sup></span> This shows that even regulators are not protected from the allure of precision oncology.</p><p>Despite the initial promising results of this trial, the fundamental design flaws of CodeBreak 300 leave it fighting for life amidst the competitive landscape of precision oncology trials. However, it is bias in reporting and approval of the drug that ultimately triggers a <i>code blue</i> for sotorasib in advanced colorectal cancer. Despite what could be otherwise described as an exploratory analysis of survival outcomes, the conclusions of this study assert that sotorasib 960 mg and panitumumab constitutes a <i>new standard of care</i> for patients with KRAS G12C mutations. Speculation about potential survival gains from a nonstatistically significant, underpowered analysis should not be misrepresented as clinical insight: <i>even if it is not statistically different, an improvement in OS could be particularly meaningful</i>. Such statements do not carry evidentiary weight and risk misleading clinicians and patients alike.</p><p>We are quite surprised that the FDA accepted the trial with all of these flaws and approved the drug for this indication.<span><sup>22</sup></span> Unlike the NSCLC indication, the colorectal cancer indication was granted regular approval—not even Accelerated Approval with a safety net of the requirement to confirm clinical benefit in the future. The FDA approved the 960-mg dose, although there were no statistical comparisons between the two dose levels—OS outcomes for both doses were similarly unimproved against the control arm, with HRs of 0.70 (95% CI, 0.41–1.18) and 0.83 (95% CI, 0.49–1.39), respectively, and <i>p</i> values of .2 and .5, respectively.<span><sup>2</sup></span> Granting full regular approval based on PFS for a last-line therapy trial in patients with a poor prognosis, despite negative OS, no quality-of-life improvement, and at a potentially higher dose with increased toxicities, is inexplicable. Sadly, after two large trials of this agent, the field—and, more importantly, our patients—are left with fleeting signals of efficacy, no survival benefit, and a cautionary reminder of the need to temper our excitement for innovation with the need for epistemic humility. Poorly designed or underpowered trials like CodeBreak 300 miss important opportunities to answer critical questions, at great human and economic cost. When regulators rubberstamp these trials with approvals, there is even less incentive to do things the right way.</p><p>To realize the full potential of precision oncology, we must move beyond molecular enthusiasm to methodological discipline. Only rigorously designed trials can separate promising ideas from proven benefit. Although there may yet be a place for sotorasib in the treatment of colorectal cancer, CodeBreak 300 does not provide sufficient evidence that its' time has arrived. The new FDA has signaled that they will require OS data for patients in later lines of therapy.<span><sup>23, 24</sup></span> We hope that this stance results in positive change. Rewarding drugs that fail to improve survival and that seek registration at higher doses—despite the exciting biology and allure of precision oncology—will impede innovation that matters to patients.</p><p><b>Samuel X. Stevens:</b> Conceptualization, writing–review and editing, and writing–original draft. <b>Bishal Gyawali:</b> Conceptualization and writing–review and editing.</p><p>Bishal Gyawali reports personal/consulting fees from Vivio Health outside the submitted work. Samuel X. Stevens disclosed no conflicts of interest.</p>\",\"PeriodicalId\":138,\"journal\":{\"name\":\"Cancer\",\"volume\":\"131 17\",\"pages\":\"\"},\"PeriodicalIF\":5.1000,\"publicationDate\":\"2025-08-19\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://acsjournals.onlinelibrary.wiley.com/doi/epdf/10.1002/cncr.70056\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://acsjournals.onlinelibrary.wiley.com/doi/10.1002/cncr.70056\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer","FirstCategoryId":"3","ListUrlMain":"https://acsjournals.onlinelibrary.wiley.com/doi/10.1002/cncr.70056","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

Kirsten大鼠肉瘤病毒癌基因同源物(KRAS)长期以来被认为是非小细胞肺癌(NSCLC)和胃肠道(GI)癌症的不可药物的致癌驱动因子,其治疗靶向能力是过去十年最重要的生物医学发现之一。因此,第一个进入临床的KRAS甘氨酸-半胱氨酸密码子12 (G12C)突变抑制剂sotorasib被誉为精准肿瘤学的胜利然而,sotorasib从临床前发现到美国食品和药物管理局(FDA)批准的过程,为这个越来越专注于分子靶向治疗的领域提供了一个警示。尽管精准肿瘤学取得了成功,但精准肿瘤学不仅仅是针对正确的目标使用正确的药物。它还要求在适当的时间、适当的时间使用适当的剂量,除此之外,还可以加上适当的报告。然而,在阅读了最近cocodebreak 300对sotorasib在结直肠癌中的试验结果后(ClinicalTrials.gov识别码NCT05198934),我们认为sotorasib在这一定义的许多领域都存在不足。sotorasib的案例也突显出,监管机构没有鼓励为患者提供真正承诺的精准肿瘤学所需的条件。CodeBreaK 300是一项3期临床试验,旨在评估sotorasib联合帕尼单抗(panitumumab)与研究者选择(TIC)治疗在化疗进展的晚期结直肠癌患者中的疗效和安全性,该患者携带KRAS G12C突变尽管该试验中纳入的患者群体和对照组的选择是适当的,但该试验的一个显著特征是sotorasib的适当剂量水平的不确定性。注册试验的剂量通常在早期临床试验中确定,以尽量减少暴露于可能无效或有毒剂量的参与者人数。因此,值得注意的是,这个3期试验似乎进行了剂量测定,分别为240毫克和960毫克剂量进行了单独的研究。非线性药代动力学的理由,虽然不是不真实的,也许只是故事的一部分。由于在早期试验中显示出希望,sotorasib于2021年获得FDA加速批准用于KRAS G12C突变型NSCLC。然而,FDA指出,初步试验“受到缺乏可靠剂量探索的阻碍”,要求在上市后研究中进行进一步探索,并强调了药物开发速度和确定性之间的紧张关系。适当地,960毫克和240毫克剂量进行了进一步的评估,在疗效上没有统计学上的显著差异,但960毫克剂量的治疗相关不良事件更高虽然在2024年发布,但专利申请的证据表明,早在2022年3月,在CodeBreaK 300开始注册之前,赞助商就知道了这一点尽管该试验似乎解决了这些不确定性,但分层统计方法(先测试960 mg与TIC,然后测试240 mg与TIC)禁止两种剂量的sotorasib之间的推论比较,这进一步模糊了剂量选择问题。这些设计决策具有道德、临床和经济意义。研究人员在道德上有责任将研究参与者暴露于伤害的风险降到最低:这是良好临床实践的共同原则。虽然设计用于直接比较两种药物剂量水平的疗效的试验需要更大的规模——实际上,大约需要两倍的参与者人数——但这种强有力的剂量探索似乎正是FDA之前所要求的在我们看来,将参与者的时间、精力和福利花费在非信息性剂量比较上的选择,在道德上是有问题的——特别是考虑到发起人拥有令人不安的剂量平衡信息的建议。6,7如果还不知道这些信息,在没有充分解决最佳剂量的不确定性之前,CodeBreak 300不应该进行。其次,所选择的设计引起了对剂量选择背后意图的关注:通过优先考虑960毫克剂量进行分层测试,该试验可能支持监管决定,倾向于高成本剂量,但没有显著临床益处,潜在危害更大,给卫生系统带来更高的经济负担。最后,扩大入组以纳入第三组会产生显著的机会成本,从而降低了回答有关总生存期(OS)的更有临床意义的问题的统计能力。因此,尽管该试验招募了相当数量的具有这种相对不常见突变的参与者,但该试验在检测以患者为中心的结果差异方面的能力不足。 一种更以患者为中心的方法是进行分层测试,但方向相反,即先测试240mg,然后测试960mg。CodeBreak 300的主要终点达到了,sotorasib 960 mg-panitumumab与对照组相比具有统计学上显著的无进展生存(PFS)优势(风险比[HR], 0.48; 95%可信区间[CI], 0.30-0.78; p = 0.005),尽管240 mg剂量未能做到这一点(HR, 0.59; 95% CI, 0.37-0.95; p = 0.036;由于α分裂而呈阴性)。9,10较高的剂量也导致较高的总有效率(ORR; 26.4% vs. 5.7%)。这些PFS和ORR结果必须考虑到FDA批准960毫克剂量的决定。这些比较与先前的剂量发现研究形成对比,后者显示240毫克和960毫克剂量之间存在更模糊的差异尽管两种剂量与对照组相比,ORR和PFS结果存在差异(两种剂量之间没有任何结果的相互比较),但它们并没有回答对患者、临床医生和监管机构来说最重要的问题,这些问题必须做出具有挑战性的最后一线治疗决策,即sotorasib是否提高了生存率和生活质量。在这项试验中,PFS被选为主要终点,OS和健康相关生活质量被降级为次要终点。CodeBreak 300甚至无法检测治疗组之间的OS差异。出于几个原因,这令人担忧。首先,OS和生活质量一直被认为对晚期实体瘤患者至关重要。11-16其次,在二线和后期结直肠癌试验中,PFS不是OS的有效替代指标。17,18我们认识到,在某些情况下,像PFS这样的中间终点可能是合理的,例如,在生存期较长的疾病中或在已有代孕的情况下。然而,这些条件在最后一线试验人群中没有得到满足通常,PFS的吸引力在于缩短了结果读取的时间。然而,对于最后一线治疗试验,通常可以很快获得OS结果,如在本例中,在获得PFS结果后仅6个月的额外随访即可获得OS结果值得注意的是,该试验的对照组——trifluridine/tipiracil或reorafenib,以及该领域最近批准的其他药物,如trifluridine/tipiracil + bevacizumab或fruquitinib——都是基于OS改善而获得批准的。19,20事实上,目前还没有其他药物被批准用于仅基于PFS的最后一线结直肠癌。靶向新突变的生物学吸引力不应弥补改善这组患者OS的需要。令人惊讶的是,在肺癌的最后一线试验CodeBreak 200中,根据监管反馈,OS的初始主要终点随后更改为PFS,以限制在对照组中暴露的患者这表明,即使是监管机构也无法免受精准肿瘤学的诱惑。尽管这项试验的初步结果很有希望,但CodeBreak 300的基本设计缺陷使其在竞争激烈的精准肿瘤学试验中挣扎求生。然而,该药物的报道和批准存在偏见,最终触发了sotorasib治疗晚期结直肠癌的蓝色代码。尽管可以被描述为对生存结果的探索性分析,但本研究的结论断言,sotorasib 960 mg和panitumumab构成了KRAS G12C突变患者的新护理标准。从非统计显著性、不充分的分析中推测潜在的生存收益不应该被曲解为临床洞察:即使它在统计上没有不同,OS的改善也可能特别有意义。这样的陈述不具有证据的分量,并且有误导临床医生和患者的风险。我们很惊讶FDA居然接受了有这些缺陷的试验,并批准了这种药物用于这种适应症与非小细胞肺癌适应症不同,结直肠癌适应症获得了常规批准,甚至没有加速批准,也没有要求在未来确认临床益处的安全网。FDA批准了960mg剂量,尽管两种剂量水平之间没有统计学比较-两种剂量的os结果与对照组相比同样没有改善,hr分别为0.70 (95% CI, 0.41-1.18)和0.83 (95% CI, 0.49-1.39), p值为。2和。5, respectively.2基于PFS对预后不良患者的最后一线治疗试验给予完全的常规批准是令人费解的,尽管OS为负,生活质量没有改善,并且可能使用更高的剂量增加毒性。 可悲的是,在对这种药物进行了两次大规模试验之后,这个领域——更重要的是,我们的病人——只留下了短暂的疗效信号,没有生存益处,这是一个警告,提醒我们需要用认知上的谦卑来缓和我们对创新的兴奋。像CodeBreak 300这样设计糟糕或动力不足的试验错失了回答关键问题的重要机会,付出了巨大的人力和经济代价。当监管机构批准这些试验时,就更没有动力按正确的方式做事了。为了充分发挥精准肿瘤学的潜力,我们必须超越分子热情,转向方法论学科。只有经过严格设计的试验才能将有希望的想法与已证实的益处区分开来。尽管sotorasib在结肠直肠癌的治疗中可能仍有一席之地,但CodeBreak 300并没有提供足够的证据表明它的“时机已经到来”。新的FDA已经发出信号,他们将要求后期治疗的患者获得OS数据。我们希望这一立场能带来积极的变化。奖励那些不能提高生存率和寻求更高剂量注册的药物——尽管生物学令人兴奋,精准肿瘤学也很诱人——将阻碍对患者至关重要的创新。塞缪尔·史蒂文斯:概念化,写作-审查和编辑,写作-原稿。Bishal Gyawali:概念和写作-审查和编辑。Bishal Gyawali报告了Vivio Health提交的工作之外的个人/咨询费。塞缪尔·x·史蒂文斯没有透露任何利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

CodeBreaK or code blue? Assessing sotorasib's vital signs in metastatic colorectal cancer

CodeBreaK or code blue? Assessing sotorasib's vital signs in metastatic colorectal cancer

CodeBreaK or code blue? Assessing sotorasib's vital signs in metastatic colorectal cancer

CodeBreaK or code blue? Assessing sotorasib's vital signs in metastatic colorectal cancer

The ability to therapeutically target Kirsten rat sarcoma viral oncogene homolog (KRAS)–long considered an undruggable oncogenic driver of nonsmall cell lung cancer (NSCLC) and gastrointestinal (GI) cancer—is one of the most important biomedical discoveries of the past decade. Therefore, sotorasib, the first KRAS glycine-to-cysteine substitution at codon 12 (G12C) mutation inhibitor to reach the clinic was heralded as a triumph of precision oncology.1 Yet the path of sotorasib from preclinical discovery to US Food and Drug Administration (FDA) approval offers a cautionary tale for a field increasingly immersed in molecularly targeted therapies. Although heralded as a success of precision oncology, precision oncology is not merely the right drug for the right target. It also entails the right dose at the right time for the right duration, to which one could arguably add right reporting. However, after reading the results from the recent CodeBreak 300 trial of sotorasib in colorectal cancer (ClinicalTrials.gov identifier NCT05198934), we believe sotorasib has fallen short in many domains of this definition. The case of sotorasib also highlights how regulators have not encouraged the conditions needed to deliver the real promise of precision oncology to patients.

CodeBreaK 300 is a phase 3 trial evaluating the efficacy and safety of sotorasib in combination with panitumumab versus treatment of investigator's choice (TIC) in patients with advanced colorectal cancer harboring a KRAS G12C mutation who progressed on chemotherapies.2 Although the included patient population and choice of control arm in this trial were appropriate, a striking feature of this trial is the uncertainty about the appropriate dose level for sotorasib. Doses for registration trials are typically established in early phase clinical trials to minimize the number of participants exposed to potentially ineffective or toxic dosages. Thus it is notable that this phase 3 trial appears to conduct a dose-finding exercise, with separate arms for the 240-mg and 960-mg doses, respectively. The justification of nonlinear pharmacokinetics, although not untrue, is perhaps only a part of the story.

Having shown promise in early phase trials, sotorasib received FDA Accelerated Approval for use in KRAS G12C mutant NSCLC in 2021. However, the FDA noted that preliminary trials were “hampered by a lack of robust dose exploration,” mandating further exploration in postmarketing studies3 and highlighting the tension between speed and certainty in drug development. Appropriately, the 960-mg and 240-mg dosages underwent further evaluation, with no statistically significant difference in efficacy but with higher treatment-related adverse events for the 960-mg dose.4 Although published in 2024, evidence from patent applications suggests that this was known to the sponsor as early as March 2022, before CodeBreaK 300 commenced enrolling.5 Although the trial appears to address these uncertainties, the hierarchical statistical approach—testing 960 mg versus TIC followed by 240 mg versus TIC—prohibits inferential comparisons between both doses of sotorasib, further clouding the dose-selection issue.

These design decisions have moral, clinical, and economic implications. Study investigators have a moral imperative to minimize research participants' exposure to harm: this is a common tenet of Good Clinical Practice. Although a trial designed to directly compare efficacy across both drug dose levels would need to be much larger—indeed, approximately twice the number of participants would be required—this robust dose exploration appears to be exactly what the FDA had previously asked for.3 The choice to spend participants' time, effort, and welfare, as Kimmelman suggests, on a noninformative dose comparison, in our view, is morally questionable—particularly considering suggestions that the sponsor possessed information unsettling equipoise about dosage.6, 7 If this information was not yet known, CodeBreak 300 should not have proceeded without adequately resolving uncertainties about optimal dosing.

Second, the chosen design raises concerns about the intent behind dose selection: by prioritizing the 960-mg dose for hierarchical testing, the trial may have supported regulatory decisions favoring higher cost dosing with no significant clinical benefit, greater potential harms, and higher economic burden for the health system. Finally, inflating enrolment to include a third arm incurred a significant opportunity cost, reducing the statistical power available to answer more clinically meaningful questions regarding overall survival (OS). Consequently, the trial was underpowered to detect differences in patient-centered outcomes despite enrolling a sizeable population of participants with this relatively infrequent mutation.8 A more patient-centric approach would have been either to do the hierarchical testing but in the opposite direction-that is, first test 240mg and then test 960mg.

The primary end point of CodeBreak 300 was met, with sotorasib 960 mg–panitumumab showing a statistically significant progression-free survival (PFS) advantage over the control arm (hazard ratio [HR], 0.48; 95% confidence interval [CI], 0.30–0.78; p = .005), although the 240-mg dose failed to do so (HR, 0.59; 95% CI, 0.37–0.95; p = .036; negative because of alpha splitting).9, 10 The higher dose also led to a higher overall response rate (ORR; 26.4% vs. 5.7%). Both of these PFS and ORR outcomes must have been factored into the FDA decision to approve the 960-mg dose. These comparisons contrast with prior dose-finding studies, which demonstrated more equivocal differences between the 240-mg and 960-mg doses.4 Notwithstanding these differences in ORR and PFS outcomes between the two doses versus the control arm—the two doses were not compared against each other for any outcome—they do not answer the questions that are most important to patients, clinicians, and regulators who must make challenging therapeutic decisions for a last-line therapy, which is whether sotorasib improves survival and quality of life.

In this trial, PFS was chosen as the primary end point, with OS and health-related quality of life relegated to secondary end points. CodeBreak 300 was not even powered to detect differences in OS between treatment arms. This is concerning for several reasons. First, OS and quality of life are consistently identified as being of primal importance to patients living with advanced solid tumors.11-16 Second, PFS is not a valid surrogate measure of OS in second-line and later line colorectal cancer trials.17, 18 We recognize that there are situations in which intermediate end points like PFS may be justifiable, for example, in diseases with long survival or when prior surrogacy has been established. However, these conditions are not met in this last-line trial population.18 Usually, the appeal of PFS is shortening the time to results readout. However, for last-line therapy trials, OS results are usually available quickly, as in this case, I which OS results were available with just 6 months of additional follow-up after PFS results were available.2 Notably, the control arms of this trial—trifluridine/tipiracil or regorafenib, as well as other recently approved drugs in this space, such as trifluridine/tipiracil plus bevacizumab or fruquitinib—were all approved based on OS improvement.19, 20 Indeed, no other drugs have been approved for last-line colorectal cancer based on PFS alone. The allure of the biology of targeting a novel mutation should not compensate for the need to improve OS in this group of patients. It is striking that, in CodeBreak 200, the last-line trial in lung cancer, the initial primary end point of OS was subsequently changed to PFS according to regulatory feedback to limit patients exposed in the control arm.21 This shows that even regulators are not protected from the allure of precision oncology.

Despite the initial promising results of this trial, the fundamental design flaws of CodeBreak 300 leave it fighting for life amidst the competitive landscape of precision oncology trials. However, it is bias in reporting and approval of the drug that ultimately triggers a code blue for sotorasib in advanced colorectal cancer. Despite what could be otherwise described as an exploratory analysis of survival outcomes, the conclusions of this study assert that sotorasib 960 mg and panitumumab constitutes a new standard of care for patients with KRAS G12C mutations. Speculation about potential survival gains from a nonstatistically significant, underpowered analysis should not be misrepresented as clinical insight: even if it is not statistically different, an improvement in OS could be particularly meaningful. Such statements do not carry evidentiary weight and risk misleading clinicians and patients alike.

We are quite surprised that the FDA accepted the trial with all of these flaws and approved the drug for this indication.22 Unlike the NSCLC indication, the colorectal cancer indication was granted regular approval—not even Accelerated Approval with a safety net of the requirement to confirm clinical benefit in the future. The FDA approved the 960-mg dose, although there were no statistical comparisons between the two dose levels—OS outcomes for both doses were similarly unimproved against the control arm, with HRs of 0.70 (95% CI, 0.41–1.18) and 0.83 (95% CI, 0.49–1.39), respectively, and p values of .2 and .5, respectively.2 Granting full regular approval based on PFS for a last-line therapy trial in patients with a poor prognosis, despite negative OS, no quality-of-life improvement, and at a potentially higher dose with increased toxicities, is inexplicable. Sadly, after two large trials of this agent, the field—and, more importantly, our patients—are left with fleeting signals of efficacy, no survival benefit, and a cautionary reminder of the need to temper our excitement for innovation with the need for epistemic humility. Poorly designed or underpowered trials like CodeBreak 300 miss important opportunities to answer critical questions, at great human and economic cost. When regulators rubberstamp these trials with approvals, there is even less incentive to do things the right way.

To realize the full potential of precision oncology, we must move beyond molecular enthusiasm to methodological discipline. Only rigorously designed trials can separate promising ideas from proven benefit. Although there may yet be a place for sotorasib in the treatment of colorectal cancer, CodeBreak 300 does not provide sufficient evidence that its' time has arrived. The new FDA has signaled that they will require OS data for patients in later lines of therapy.23, 24 We hope that this stance results in positive change. Rewarding drugs that fail to improve survival and that seek registration at higher doses—despite the exciting biology and allure of precision oncology—will impede innovation that matters to patients.

Samuel X. Stevens: Conceptualization, writing–review and editing, and writing–original draft. Bishal Gyawali: Conceptualization and writing–review and editing.

Bishal Gyawali reports personal/consulting fees from Vivio Health outside the submitted work. Samuel X. Stevens disclosed no conflicts of interest.

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
Cancer
Cancer 医学-肿瘤学
CiteScore
13.10
自引率
3.20%
发文量
480
审稿时长
2-3 weeks
期刊介绍: The CANCER site is a full-text, electronic implementation of CANCER, an Interdisciplinary International Journal of the American Cancer Society, and CANCER CYTOPATHOLOGY, a Journal of the American Cancer Society. CANCER publishes interdisciplinary oncologic information according to, but not limited to, the following disease sites and disciplines: blood/bone marrow; breast disease; endocrine disorders; epidemiology; gastrointestinal tract; genitourinary disease; gynecologic oncology; head and neck disease; hepatobiliary tract; integrated medicine; lung disease; medical oncology; neuro-oncology; pathology radiation oncology; translational research
×
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:604180095
Book学术官方微信