考虑肿瘤药物开发过程中候选药物流失的根本原因。

IF 1.5 4区 医学 Q3 PHARMACOLOGY & PHARMACY Clinical Pharmacology in Drug Development Pub Date : 2024-08-20 DOI:10.1002/cpdd.1464
Yin-Ming Kuo, Jeffrey S. Barrett
{"title":"考虑肿瘤药物开发过程中候选药物流失的根本原因。","authors":"Yin-Ming Kuo,&nbsp;Jeffrey S. Barrett","doi":"10.1002/cpdd.1464","DOIUrl":null,"url":null,"abstract":"<p>Cancer remained the second-leading cause of death in the United States in 2020, based on the data from the US Centers for Disease Control and Prevention. While there have been lots of money and time devoted to this therapeutic area, the needs from these patients with cancer were still substantial. The fundamental issue is high attrition rate for oncology drugs, which contributes to the higher cost for oncology drug developers. The study for the success rate from first-in-human trials to registration for 10 big pharmaceutical companies in the United States and Europe indicated that the average success rate in all therapeutic fields was about 11% from 1991 to 2000.<span><sup>1</sup></span> The success rates varied between different therapeutic areas, whereas oncology drugs had a relatively low success rate, approximately 5%. In other words, only 1 in 20 new chemical entities passed through clinical trials and received an approval from the European and/or the US regulatory authorities. Kola and Landis also studied the reasons for drug attrition during drug development from 1991 to 2000. They discovered that the primary reason for drug attrition changed from inappropriate pharmacokinetics (PK) and low bioavailability (approximately 40%) in 1991 to a lack of efficacy and safety (approximately 60%) in 2000.<span><sup>1</sup></span> Kola and Landis concluded 2 strategies that may reduce the rate of attrition. First, in some therapeutic areas with lower success rates (eg, oncology and central nervous system), appropriate animal models and biomarkers have to be carefully chosen during early drug discovery and development stages.<span><sup>1</sup></span> For example, a transgenic animal model is more suitable than a xenograft animal model for preclinical studies of oncology drugs. Second, Kola and Landis observed that biologics had a higher success rate to launch from the first-in-human studies, especially in the areas of immunology and cancer, implying that biologics are safer than conventional chemical drugs.<span><sup>1</sup></span></p><p>Antibody drugs, 1 group of biologics, generally have fewer safety concerns and fewer PK issues.<span><sup>2, 3</sup></span> In general, antibodies possess a few pharmacological characteristics, including high potency, limited off-target toxicity, and a low risk of biotransformation to toxic metabolites.<span><sup>4</sup></span> Thus, the possibility of drug-drug interactions or renal and hepatic impairment on drug excretion is relatively low, which could significantly eliminate a few matters that could potentially result in drug attrition.</p><p>On the other hand, Walker and Newell analyzed the data for small molecular cancer drugs on the attrition from 1995 to 2007, indicating that the attrition rate within the oncology field was 82%; however, the attrition rate of kinase inhibitors was 53%.<span><sup>5</sup></span> It is worth noticing that kinase inhibitors were more successful in the high-risk transition from Phase 2 to Phase 3.<span><sup>5</sup></span> In addition, Hutchinson and Kirk concluded that the estimated glomerular filtration rate and vascular endothelial growth factor targeted agents and/or other kinase inhibitors had relative high success rates, especially adjunctly treating with antiangiogenic drugs.<span><sup>6</sup></span> Overall, for small molecular cancer drugs, molecularly targeted drugs demonstrated the potential to reduce attrition rates.</p><p>Moreover, Waring et al found that safety and toxicology were the largest sources of drug failure from 4 major pharmaceutical companies from 2000 to 2010, suggesting a lack of safety was the main factor to contribute drug attrition.<span><sup>7</sup></span> The links between physicochemical properties and frequent causes of attrition (eg, preclinical toxicology, clinical safety, and human PK) were also assessed. Waring et al concluded that none of the physicochemical properties correlated with the attrition of the drugs. The work was the first study to investigate the relationship between hydrophobicity and clinical failure, implying the stringent control of physicochemical attributes may not be a key to mitigating attrition in small molecular drug development.<span><sup>7</sup></span></p><p>In this study, to understand the root causes of discontinued oncology drugs from 2005 to 2013, the correlated factors were analyzed. Further, a questionnaire was created and disseminated to group leaders in the pharmaceutical industry, the Food and Drug Administration (FDA), and oncology clinicians for first-hand feedback. A few strategies, such as investment on the paradigm-shifting drugs and investigation of biomarkers, were concluded to mitigate attrition. Furthermore, using biomarkers could guide adaptive clinical trials to improve the efficiency of drug discovery and development.</p><p>The study sought to assess factors associated with a relatively high attrition rate for oncology drug candidates, which is centric to the problem of how drugs could be adequately designed and how the clinical studies could be employed effectively and efficiently. Recent paradigm shifts in early-stage development and clinical development plans suggest that candidate selection is less influenced by toxicity reduction with more emphasis placed on biologic activity aligned with efficacy expectations. It may suggest that even when the oncology drug candidates had low safety concerns, a lack of efficacy may still be present and impactful. Further, both the attrition cases of antibodies and small molecules increased from 2005 to 2013, which is similar to the fact that the success rate of biologics had leveled out from 2011 to 2013.<span><sup>26</sup></span> Since efficacy can only be studied by clinical trials, the clinical trials should be unbiasedly designed, and it could be designed in the adaptive ways. For example, with fewer toxicity concerns, the efficacy of biologics could be examined at the early stage of clinical trial to make the process of drug development more efficient and more cost effective. A few options may be considered, such as having Phase 0 to study PK/pharmacodynamics<span><sup>33</sup></span> and/or moving the proof-of-concept study to Phase 1.<span><sup>19</sup></span> Regardless of what adaptions are chosen, prior to implementing the adaptions, effective communications with regulatory agencies are needed to ensure that the regulatory infrastructure is flexible enough and ready to review these adaptions. Otherwise, these adjustments may incur even more issues during panel review.</p><p>In addition, there are unmet needs for a few cancer types in the cancer community. The current advancing technologies can help us identify genomics and/or biomarker alterations, and yet could guide us to design effective clinical trials and further facilitate targeting the right patients. A recent study revealed that the tests from whole genome sequencing for a patient with metastatic colorectal cancer identified more than 2000 genomic alterations.<span><sup>34</sup></span> Along with the results from transcriptome sequencing, the most differentially expressed genes were the members of 2 proto-oncogene families, FOS and JUN. These results strongly suggested that blocking the reninangiotensin system could render therapeutic benefit.<span><sup>34</sup></span> Thus, the antihypertensive angiotensin II receptor antagonist irbesartan was considered for drug repurposing to an oncology treatment, resulting in the patient experiencing a dramatic and persistent response. Although this was the successful application of precision medication, it did demonstrate that these molecular-level technologies can accurately aim at the right patients/targets. Similarly, the application of these tools could lead a correct design in the clinical trials, especially for targeting correct subgroup patients.</p><p>Considering cancer as a group of complicated and highly heterogeneous diseases, the majority of tumors should provide multiple targets. Therefore, the combination of drugs for oncology treatment is likely required. Given the successful aforementioned example of precision medicine, the genomic-based or biomarker-driven stratification for targeting correct subpopulation of patients can significantly facilitate the outcomes of treatment and prognosis. Additionally, the other key to this successful example was drug repurposing. Based on the fact that to date drugs are skyrocketed in nature, not to mention the failed drugs, the possibility of drug repurposing for oncology treatment should be scrutinized. Drug repurposing can save lots of money and time on drug discovery and development and maximize the capability of a drug. To cope with high demands of combination treatments for the “targeted cancer therapies,” new drug discovery including drug repurposing, companion diagnostics, and adaptive clinical designs with flexible regulatory review should all be comprehensively considered.</p><p>The authors declared no competing interests for this work.</p><p>No funding was received for this work.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":null,"pages":null},"PeriodicalIF":1.5000,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpdd.1464","citationCount":"0","resultStr":"{\"title\":\"Consideration of the Root Causes in Candidate Attrition During Oncology Drug Development\",\"authors\":\"Yin-Ming Kuo,&nbsp;Jeffrey S. Barrett\",\"doi\":\"10.1002/cpdd.1464\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Cancer remained the second-leading cause of death in the United States in 2020, based on the data from the US Centers for Disease Control and Prevention. While there have been lots of money and time devoted to this therapeutic area, the needs from these patients with cancer were still substantial. The fundamental issue is high attrition rate for oncology drugs, which contributes to the higher cost for oncology drug developers. The study for the success rate from first-in-human trials to registration for 10 big pharmaceutical companies in the United States and Europe indicated that the average success rate in all therapeutic fields was about 11% from 1991 to 2000.<span><sup>1</sup></span> The success rates varied between different therapeutic areas, whereas oncology drugs had a relatively low success rate, approximately 5%. In other words, only 1 in 20 new chemical entities passed through clinical trials and received an approval from the European and/or the US regulatory authorities. Kola and Landis also studied the reasons for drug attrition during drug development from 1991 to 2000. They discovered that the primary reason for drug attrition changed from inappropriate pharmacokinetics (PK) and low bioavailability (approximately 40%) in 1991 to a lack of efficacy and safety (approximately 60%) in 2000.<span><sup>1</sup></span> Kola and Landis concluded 2 strategies that may reduce the rate of attrition. First, in some therapeutic areas with lower success rates (eg, oncology and central nervous system), appropriate animal models and biomarkers have to be carefully chosen during early drug discovery and development stages.<span><sup>1</sup></span> For example, a transgenic animal model is more suitable than a xenograft animal model for preclinical studies of oncology drugs. Second, Kola and Landis observed that biologics had a higher success rate to launch from the first-in-human studies, especially in the areas of immunology and cancer, implying that biologics are safer than conventional chemical drugs.<span><sup>1</sup></span></p><p>Antibody drugs, 1 group of biologics, generally have fewer safety concerns and fewer PK issues.<span><sup>2, 3</sup></span> In general, antibodies possess a few pharmacological characteristics, including high potency, limited off-target toxicity, and a low risk of biotransformation to toxic metabolites.<span><sup>4</sup></span> Thus, the possibility of drug-drug interactions or renal and hepatic impairment on drug excretion is relatively low, which could significantly eliminate a few matters that could potentially result in drug attrition.</p><p>On the other hand, Walker and Newell analyzed the data for small molecular cancer drugs on the attrition from 1995 to 2007, indicating that the attrition rate within the oncology field was 82%; however, the attrition rate of kinase inhibitors was 53%.<span><sup>5</sup></span> It is worth noticing that kinase inhibitors were more successful in the high-risk transition from Phase 2 to Phase 3.<span><sup>5</sup></span> In addition, Hutchinson and Kirk concluded that the estimated glomerular filtration rate and vascular endothelial growth factor targeted agents and/or other kinase inhibitors had relative high success rates, especially adjunctly treating with antiangiogenic drugs.<span><sup>6</sup></span> Overall, for small molecular cancer drugs, molecularly targeted drugs demonstrated the potential to reduce attrition rates.</p><p>Moreover, Waring et al found that safety and toxicology were the largest sources of drug failure from 4 major pharmaceutical companies from 2000 to 2010, suggesting a lack of safety was the main factor to contribute drug attrition.<span><sup>7</sup></span> The links between physicochemical properties and frequent causes of attrition (eg, preclinical toxicology, clinical safety, and human PK) were also assessed. Waring et al concluded that none of the physicochemical properties correlated with the attrition of the drugs. The work was the first study to investigate the relationship between hydrophobicity and clinical failure, implying the stringent control of physicochemical attributes may not be a key to mitigating attrition in small molecular drug development.<span><sup>7</sup></span></p><p>In this study, to understand the root causes of discontinued oncology drugs from 2005 to 2013, the correlated factors were analyzed. Further, a questionnaire was created and disseminated to group leaders in the pharmaceutical industry, the Food and Drug Administration (FDA), and oncology clinicians for first-hand feedback. A few strategies, such as investment on the paradigm-shifting drugs and investigation of biomarkers, were concluded to mitigate attrition. Furthermore, using biomarkers could guide adaptive clinical trials to improve the efficiency of drug discovery and development.</p><p>The study sought to assess factors associated with a relatively high attrition rate for oncology drug candidates, which is centric to the problem of how drugs could be adequately designed and how the clinical studies could be employed effectively and efficiently. Recent paradigm shifts in early-stage development and clinical development plans suggest that candidate selection is less influenced by toxicity reduction with more emphasis placed on biologic activity aligned with efficacy expectations. It may suggest that even when the oncology drug candidates had low safety concerns, a lack of efficacy may still be present and impactful. Further, both the attrition cases of antibodies and small molecules increased from 2005 to 2013, which is similar to the fact that the success rate of biologics had leveled out from 2011 to 2013.<span><sup>26</sup></span> Since efficacy can only be studied by clinical trials, the clinical trials should be unbiasedly designed, and it could be designed in the adaptive ways. For example, with fewer toxicity concerns, the efficacy of biologics could be examined at the early stage of clinical trial to make the process of drug development more efficient and more cost effective. A few options may be considered, such as having Phase 0 to study PK/pharmacodynamics<span><sup>33</sup></span> and/or moving the proof-of-concept study to Phase 1.<span><sup>19</sup></span> Regardless of what adaptions are chosen, prior to implementing the adaptions, effective communications with regulatory agencies are needed to ensure that the regulatory infrastructure is flexible enough and ready to review these adaptions. Otherwise, these adjustments may incur even more issues during panel review.</p><p>In addition, there are unmet needs for a few cancer types in the cancer community. The current advancing technologies can help us identify genomics and/or biomarker alterations, and yet could guide us to design effective clinical trials and further facilitate targeting the right patients. A recent study revealed that the tests from whole genome sequencing for a patient with metastatic colorectal cancer identified more than 2000 genomic alterations.<span><sup>34</sup></span> Along with the results from transcriptome sequencing, the most differentially expressed genes were the members of 2 proto-oncogene families, FOS and JUN. These results strongly suggested that blocking the reninangiotensin system could render therapeutic benefit.<span><sup>34</sup></span> Thus, the antihypertensive angiotensin II receptor antagonist irbesartan was considered for drug repurposing to an oncology treatment, resulting in the patient experiencing a dramatic and persistent response. Although this was the successful application of precision medication, it did demonstrate that these molecular-level technologies can accurately aim at the right patients/targets. Similarly, the application of these tools could lead a correct design in the clinical trials, especially for targeting correct subgroup patients.</p><p>Considering cancer as a group of complicated and highly heterogeneous diseases, the majority of tumors should provide multiple targets. Therefore, the combination of drugs for oncology treatment is likely required. Given the successful aforementioned example of precision medicine, the genomic-based or biomarker-driven stratification for targeting correct subpopulation of patients can significantly facilitate the outcomes of treatment and prognosis. Additionally, the other key to this successful example was drug repurposing. Based on the fact that to date drugs are skyrocketed in nature, not to mention the failed drugs, the possibility of drug repurposing for oncology treatment should be scrutinized. Drug repurposing can save lots of money and time on drug discovery and development and maximize the capability of a drug. To cope with high demands of combination treatments for the “targeted cancer therapies,” new drug discovery including drug repurposing, companion diagnostics, and adaptive clinical designs with flexible regulatory review should all be comprehensively considered.</p><p>The authors declared no competing interests for this work.</p><p>No funding was received for this work.</p>\",\"PeriodicalId\":10495,\"journal\":{\"name\":\"Clinical Pharmacology in Drug Development\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":1.5000,\"publicationDate\":\"2024-08-20\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpdd.1464\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical Pharmacology in Drug Development\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cpdd.1464\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology in Drug Development","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpdd.1464","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

近期早期开发和临床开发计划的范式转变表明,候选药物的选择已不再受降低毒性的影响,而是更加重视与疗效预期相一致的生物活性。这可能表明,即使肿瘤候选药物的安全性较低,但疗效不足仍可能存在并产生影响。此外,从 2005 年到 2013 年,抗体和小分子药物的损耗率都有所上升,这与 2011 年到 2013 年生物制剂的成功率趋于平稳的事实相似26。例如,由于毒性问题较少,可在临床试验的早期阶段对生物制剂的疗效进行研究,以提高药物开发过程的效率和成本效益。有几种方案可以考虑,如在 0 期研究 PK/药效学33 和/或将概念验证研究移至 1 期19。无论选择哪种调整方式,在实施调整之前,都需要与监管机构进行有效沟通,以确保监管基础设施足够灵活,并做好审查这些调整的准备。否则,这些调整可能会在专家小组审查期间引发更多问题。此外,癌症界还有一些癌症类型的需求未得到满足。目前不断进步的技术可以帮助我们确定基因组学和/或生物标志物的改变,还可以指导我们设计有效的临床试验,并进一步帮助我们锁定合适的患者。最近的一项研究显示,对一名转移性结直肠癌患者进行的全基因组测序检测发现了 2000 多处基因组改变。34 根据转录组测序结果,差异表达最大的基因是 FOS 和 JUN 这两个原癌基因家族的成员。34 因此,抗高血压血管紧张素 II 受体拮抗剂厄贝沙坦(irbesartan)被考虑重新用于肿瘤治疗,结果患者出现了显著而持久的反应。虽然这是精准医疗的成功应用,但它确实证明了这些分子水平的技术能够准确地瞄准正确的患者/靶点。同样,这些工具的应用也能为临床试验带来正确的设计,尤其是针对正确的亚组患者。考虑到癌症是一组复杂且高度异质性的疾病,大多数肿瘤都应提供多个靶点。因此,肿瘤治疗可能需要联合用药。鉴于前述精准医疗的成功范例,基于基因组学或生物标志物驱动的分层疗法可针对正确的患者亚群,极大地促进治疗效果和预后。此外,这一成功范例的另一个关键是药物再利用。迄今为止,药物都是天价,更不用说那些失败的药物了。药物再利用可以节省大量的药物研发费用和时间,并最大限度地发挥药物的作用。为了应对 "癌症靶向治疗 "对联合治疗的高要求,新药研发,包括药物再利用、伴随诊断和灵活监管审查的适应性临床设计,都应得到全面考虑。
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
Consideration of the Root Causes in Candidate Attrition During Oncology Drug Development

Cancer remained the second-leading cause of death in the United States in 2020, based on the data from the US Centers for Disease Control and Prevention. While there have been lots of money and time devoted to this therapeutic area, the needs from these patients with cancer were still substantial. The fundamental issue is high attrition rate for oncology drugs, which contributes to the higher cost for oncology drug developers. The study for the success rate from first-in-human trials to registration for 10 big pharmaceutical companies in the United States and Europe indicated that the average success rate in all therapeutic fields was about 11% from 1991 to 2000.1 The success rates varied between different therapeutic areas, whereas oncology drugs had a relatively low success rate, approximately 5%. In other words, only 1 in 20 new chemical entities passed through clinical trials and received an approval from the European and/or the US regulatory authorities. Kola and Landis also studied the reasons for drug attrition during drug development from 1991 to 2000. They discovered that the primary reason for drug attrition changed from inappropriate pharmacokinetics (PK) and low bioavailability (approximately 40%) in 1991 to a lack of efficacy and safety (approximately 60%) in 2000.1 Kola and Landis concluded 2 strategies that may reduce the rate of attrition. First, in some therapeutic areas with lower success rates (eg, oncology and central nervous system), appropriate animal models and biomarkers have to be carefully chosen during early drug discovery and development stages.1 For example, a transgenic animal model is more suitable than a xenograft animal model for preclinical studies of oncology drugs. Second, Kola and Landis observed that biologics had a higher success rate to launch from the first-in-human studies, especially in the areas of immunology and cancer, implying that biologics are safer than conventional chemical drugs.1

Antibody drugs, 1 group of biologics, generally have fewer safety concerns and fewer PK issues.2, 3 In general, antibodies possess a few pharmacological characteristics, including high potency, limited off-target toxicity, and a low risk of biotransformation to toxic metabolites.4 Thus, the possibility of drug-drug interactions or renal and hepatic impairment on drug excretion is relatively low, which could significantly eliminate a few matters that could potentially result in drug attrition.

On the other hand, Walker and Newell analyzed the data for small molecular cancer drugs on the attrition from 1995 to 2007, indicating that the attrition rate within the oncology field was 82%; however, the attrition rate of kinase inhibitors was 53%.5 It is worth noticing that kinase inhibitors were more successful in the high-risk transition from Phase 2 to Phase 3.5 In addition, Hutchinson and Kirk concluded that the estimated glomerular filtration rate and vascular endothelial growth factor targeted agents and/or other kinase inhibitors had relative high success rates, especially adjunctly treating with antiangiogenic drugs.6 Overall, for small molecular cancer drugs, molecularly targeted drugs demonstrated the potential to reduce attrition rates.

Moreover, Waring et al found that safety and toxicology were the largest sources of drug failure from 4 major pharmaceutical companies from 2000 to 2010, suggesting a lack of safety was the main factor to contribute drug attrition.7 The links between physicochemical properties and frequent causes of attrition (eg, preclinical toxicology, clinical safety, and human PK) were also assessed. Waring et al concluded that none of the physicochemical properties correlated with the attrition of the drugs. The work was the first study to investigate the relationship between hydrophobicity and clinical failure, implying the stringent control of physicochemical attributes may not be a key to mitigating attrition in small molecular drug development.7

In this study, to understand the root causes of discontinued oncology drugs from 2005 to 2013, the correlated factors were analyzed. Further, a questionnaire was created and disseminated to group leaders in the pharmaceutical industry, the Food and Drug Administration (FDA), and oncology clinicians for first-hand feedback. A few strategies, such as investment on the paradigm-shifting drugs and investigation of biomarkers, were concluded to mitigate attrition. Furthermore, using biomarkers could guide adaptive clinical trials to improve the efficiency of drug discovery and development.

The study sought to assess factors associated with a relatively high attrition rate for oncology drug candidates, which is centric to the problem of how drugs could be adequately designed and how the clinical studies could be employed effectively and efficiently. Recent paradigm shifts in early-stage development and clinical development plans suggest that candidate selection is less influenced by toxicity reduction with more emphasis placed on biologic activity aligned with efficacy expectations. It may suggest that even when the oncology drug candidates had low safety concerns, a lack of efficacy may still be present and impactful. Further, both the attrition cases of antibodies and small molecules increased from 2005 to 2013, which is similar to the fact that the success rate of biologics had leveled out from 2011 to 2013.26 Since efficacy can only be studied by clinical trials, the clinical trials should be unbiasedly designed, and it could be designed in the adaptive ways. For example, with fewer toxicity concerns, the efficacy of biologics could be examined at the early stage of clinical trial to make the process of drug development more efficient and more cost effective. A few options may be considered, such as having Phase 0 to study PK/pharmacodynamics33 and/or moving the proof-of-concept study to Phase 1.19 Regardless of what adaptions are chosen, prior to implementing the adaptions, effective communications with regulatory agencies are needed to ensure that the regulatory infrastructure is flexible enough and ready to review these adaptions. Otherwise, these adjustments may incur even more issues during panel review.

In addition, there are unmet needs for a few cancer types in the cancer community. The current advancing technologies can help us identify genomics and/or biomarker alterations, and yet could guide us to design effective clinical trials and further facilitate targeting the right patients. A recent study revealed that the tests from whole genome sequencing for a patient with metastatic colorectal cancer identified more than 2000 genomic alterations.34 Along with the results from transcriptome sequencing, the most differentially expressed genes were the members of 2 proto-oncogene families, FOS and JUN. These results strongly suggested that blocking the reninangiotensin system could render therapeutic benefit.34 Thus, the antihypertensive angiotensin II receptor antagonist irbesartan was considered for drug repurposing to an oncology treatment, resulting in the patient experiencing a dramatic and persistent response. Although this was the successful application of precision medication, it did demonstrate that these molecular-level technologies can accurately aim at the right patients/targets. Similarly, the application of these tools could lead a correct design in the clinical trials, especially for targeting correct subgroup patients.

Considering cancer as a group of complicated and highly heterogeneous diseases, the majority of tumors should provide multiple targets. Therefore, the combination of drugs for oncology treatment is likely required. Given the successful aforementioned example of precision medicine, the genomic-based or biomarker-driven stratification for targeting correct subpopulation of patients can significantly facilitate the outcomes of treatment and prognosis. Additionally, the other key to this successful example was drug repurposing. Based on the fact that to date drugs are skyrocketed in nature, not to mention the failed drugs, the possibility of drug repurposing for oncology treatment should be scrutinized. Drug repurposing can save lots of money and time on drug discovery and development and maximize the capability of a drug. To cope with high demands of combination treatments for the “targeted cancer therapies,” new drug discovery including drug repurposing, companion diagnostics, and adaptive clinical designs with flexible regulatory review should all be comprehensively considered.

The authors declared no competing interests for this work.

No funding was received for 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
3.70
自引率
10.00%
发文量
154
期刊介绍: Clinical Pharmacology in Drug Development is an international, peer-reviewed, online publication focused on publishing high-quality clinical pharmacology studies in drug development which are primarily (but not exclusively) performed in early development phases in healthy subjects.
期刊最新文献
Population Pharmacokinetics Modeling and Simulation of Deutenzalutamide, A Novel Androgen Receptor Antagonist, in Patients With Metastatic Castration-Resistant Prostate Cancer. Bioequivalence Study of Velpatasvir/Sofosbuvir Oral Coated Tablets in Healthy Volunteers Under Fasting Conditions. A Randomized, Cross-Over Study Investigating the Comparability of Somatrogon-ghla in 2 Different Drug Product Presentations. Bioequivalence of Recombinant Human Teriparatide Injection in Healthy Adult Female Subjects in the Fasting State. Effect of Erdafitinib on the Pharmacokinetics of Midazolam and Metformin in Patients With Advanced Solid Tumors Harboring FGFR Gene Alterations.
×
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