新开发策略!将肿瘤药物研发成功率从5%提高到35%

2024-09-12 08:12

Reads:


近日,《药物化学杂志》刊登了一篇题为《机器学习能否克服95%的失败率以及只有30%获批抗癌药物能有效延长患者生存期的现实?》的文章,作者为密歇根大学孙笃新教授和密歇根大学药学院、工程学院、医学院、密歇根大数据研究所的成员,以及来自Lancaster Life Science、Aurinia和BMS的成员。

 

过去30年,肿瘤治疗已取得显著进展。1991年至2020年间,美国癌症患者死亡率下降33%。2000年至2022年间,FDA批准了250种靶向120个分子靶点的抗肿瘤药物。

 

然而,这些进展背后,美国在2023年仍有超过61万人死于癌症——相当于每天有1670人死亡,这一数字与2020年大流行高峰期间美国每日的COVID-19死亡率相当。2022年,全球因肿瘤死亡的人数仍高达1000万,是2020年全球大流行期间COVID-19死亡人数的三倍。

 

过去几十年间,全球发起了多项攻克肿瘤的研究计划。美国也重新启动了对抗肿瘤的“登月计划”,目标是在未来25年内将肿瘤死亡率降低50%。

 

不幸的是,肿瘤治疗的经济负担给实现这一目标带来挑战。例如,在美国,每名患者接受一个疗程的抗肿瘤药物治疗平均费用为17万至27.7万美元。美国每年在肿瘤治疗上的支出达2000亿美元,其中600亿美元用于抗肿瘤药物,以此计算,相当于每天支出分别为5.4亿美元和1.65亿美元。

 
 
 
这篇评论文章中,作者重点讨论了以下问题:
 

(1)过去30年,为什么尽管用了数百种有效策略来大大改进药物研发中的每一步,但抗肿瘤药物研发的失败率仍然一直高达95%,并无任何改善?

 

(2)为什么只有30%已批准的抗肿瘤药物能够延长肿瘤病人2.5个月的生命,而在美国每名癌症患者药物治疗平均费用却高达17万至27.7万美元?

 

(3)当前的药物研发策略(包括人工智能的应用)是否陷入了“幸存者偏误”陷阱,过多关注许多不重要的问题,而忽略了根本原因?

 

(4)目前研发成功一个药物已经需10-15年并花费10亿至20亿美元,是否有可能在如此漫长且昂贵的过程中,增加更多的标准?

 

(5)对当前的研药物发过程,应用人工智能(AI)和机器学习(ML)方法是否可以提高效率、增加成功率并改善药物的疗效?

 

(6)这些问题的根本原因是什么,未来的研究重点应该放在哪些方面才能找到解决问题的方案?

 
 
 
01

癌症治疗药物存在的两个相互关联的问题

 
 

1.过去30年中,抗肿瘤药物研发的95%失败率,并无任何改善。

 

科学家在药物开发过程中,已经使用了数百种有效的策略和标准,包括靶点验证、高通量筛选、先导化合物优化、临床前测试、成药性质优化、GLP毒性测试、GMP生产以及临床I-III期试验。尽管每个步骤都有很大的改进,但95%的失败率在过去30年中从未提高。(图1)

 

Figure 1. Cancer drug development has a 95% failure rate despite improvements in each step by incorporating hundreds of helpful criteria over the past 30 years. It is impractical to continually add more criteria without streamlining nonessential ones.

 

由于抗肿瘤药物研发过程既漫长又昂贵,不断增加更多的标准而不去剔除非必要的标准是不切实际的。那么,哪些标准是非必要的?

 

此外,目前所采用的所有策略,是否陷入了“幸存者偏误”的陷阱,即过度关注许多不重要的因素而忽视了重大缺陷?

 

这就像在二战期间,盟军为防止战斗机被炮弹击落,对从战场返回的幸存飞机机翼上的炮弹孔进行大量研究,从而提出解决加固机翼方案。由于缺乏那些没能从战场返回的飞机上的数据,盟军最终忽略了要加固发动机和驾驶舱这些重要地方的方案(图2)

 

尽管药物开发过程中许多标准确实很重要,但另一些标准可能只是解决了非关键问题,却忽略类似于“幸存者偏误”陷阱中的关键问题。

 

Figure 2. Survivorship Bias. The survivorship biased focus is fixing the damage on the wings of surviving/returned aircrafts where they can sustain damage and still return home, but missing the damage in the critical locations of engines and cockpit of the non-surviving/un-returned aircrafts from the battle during WWII.

 

2.仅20-42%新批的抗肿瘤药物,能显著延长患者的总生存期超过2.5个月。

 

由于临床需要,许多抗肿瘤药物的获批是基于在临床II期试验中改善PFS。这些药物随后需要在临床III期试验中,验证其延长患者OS的有效性。对于没有其他治疗选择的癌症患者而言,及早获得此类药物对他们的治疗是有益处的。

 

然而,令人失望的是,仅有20-42%的这些新获批抗肿瘤药物,能够达到由ASCO和ESMO设定的临床获益标准,即延长总生存期超过2.5个月。另外40%新批的抗肿瘤药物延长生存期2周至2.5个月,有30%新批的抗肿瘤药物未能改善总生存期。

 

在总生存期没有显著改善的情况下,可能存在几种不同的情形。如果新批的抗肿瘤药物与标准治疗相比,改善总生存期少于2.5个月,它们在疗效上或许与标准治疗相似,但可能具有其他优势,如毒性较低,这仍然是有意义的。

 

然而,假设这些新批准的药物与不适当的对照组相比,只改善少于2.5个月的总生存期,则应对其疗效保持谨慎态度。在许多情况下,这种临床疗效甚微的次优药物对照品仍在市场上销售,甚至被列入癌症治疗指南。辨别这些对照组是否能为癌症患者带来有意义的临床获益非常具有挑战性。

 

此外,如果新批准的抗癌药物(或与标准治疗联合使用)与安慰剂(或与标准治疗联合使用)相比没有改善OS,其疗效就值得怀疑。遗憾的是,许多未能改善患者OS的药物,仍基于其最初“积极”的II期试验留在市场被使用。

 

Figure 3. Only approximately 30% approved cancer drugs, based on positive improvement of progression-free survival (PFS), meaningfully extend patient overall survival (OS) by more than 2.5 months, while 30% of approved cancer drugs do not extend patient overall survivals.

 
 
02

抗肿瘤药物存在的两个相互关联的问题源于三个被忽视的因素,这些因素导致未优化好的药物进入临床试验。

 
 

这三个被忽视的因素,包括不完全的靶点/脱靶点验证、错误应用成药性质评估,以及无法优化的临床剂量从而造成95%抗肿瘤药物研发失败率或已获批药物的低疗效/安全性问题。

 

1.目前用体外靶点结合IC50筛选的药物候选物,忽视了在相关临床剂量下针对肿瘤的靶点/脱靶点的效力/特异性(PS),从而影响了药物在临床剂量下的临床疗效。

 

2.目前基于成药性标准筛选的药物候选物,忽视了由靶点/脱靶点所决定的组织/细胞选择性(TS),从而影响了在临床剂量下对正常器官的不良反应(靶点/脱靶点毒性)。

 

3.临床剂量无法得到优化来平衡临床疗效/安全性,因为药物的效力/特异性和组织/细胞选择性已经决定临床剂量无法优化,而GLP动物毒性实验并不能预测在临床治疗剂量范围内的不良反应(靶点/脱靶点毒性)。

 

文章作者提出“STAR系统”(结构-组织/细胞选择性-活性关系),用这三个因素将药物分为STAR I-IV类药物。STAR系统可以通过简化药物开发流程来指导药物研发策略,以提高成功率和效率。目标是选择STAR I类药物,而不是像目前的流程那样,过于关注于STAR II/IV类化合物。

 

Figure 4. STAR (Structure-Tissue/Cell Selectivity-Activity-Relationship) guides drug development strategies to select STAR class I drugs with high efficacy, high safety, and high success rate, and to avoid class II/IV drugs with low success rates.

 
 
03

如何使用人工智能和机器学习来克服95%抗肿瘤药物失败率以及仅有30%的获批抗肿瘤药物能显著延长患者总生存期的现实?

 
 

如果使用正确并已验证的机器学习(ML)模型,人工智能(AI)和机器学习(ML)会通过降低成本/时间来提高药物研发过程每个步骤的效率。

 

然而,倘若我们仍依赖当前的药物研发流程,仅仅使用AI和ML技术,不一定能够克服95%的抗肿瘤药物开发失败率及提高药物的疗效。

 

1.应用机器学习模型提高药物开发效率:

 

机器学习(ML)模型已被用于改善目前药物开发过程中每个步骤。一个经过充分验证的ML模型能够通过节省时间和成本来提高效率,而无需繁琐的实验室实验,这非常有意义。实际上,ML模型已被应用于当前药物开发过程中的每一个步骤(图1),包括在各种癌症类型中的疾病靶点识别/验证、高通量筛选、药物设计和优化、药物-靶点相互作用(DTI)预测、药代动力学和成药性预测、毒性预测、临床试验设计。用ML模型在药物研发过程每个步中如何通过节省时间和成本来提高效率的例子也不胜枚举。

 

2.开发STAR指导的机器学习模型,以提高药物开发的成功率和效率:

 

我们需要开发基于STAR指导的机器学习(ML)模型,来直接预测临床剂量/疗效/安全性的平衡。基于STAR指导的ML模型可以帮助设计STAR I类药物,从而提高成功率和效率。基于STAR的ML模型,可以通过以下五个参数并使用在相关临床剂量下的体外/离体实验、临床前和临床数据,来预测临床不良反应(AE)(靶点/脱靶点毒性)或临床疗效(Eff)(图5)

 

Clinical adverse effects at relevant clinical doses ŷ(AE) = (PS ˟ TSN ˟ EXN ˟ CP ˟ S)w (1)

Clinical efficacy at relevant clinical doses ŷ(Eff) =  (PS ˟ TST ˟ EXT ˟ CP ˟ S)w (2)

 

基于STAR指导的ML模型可以通过五个步骤构建,来预测在相关临床剂量下的临床不良反应(靶点和脱靶点毒性)ŷ(AE)或临床疗效(Eff)(图5)。ML模型可以使用以下五个指标来建立:药物结构信息(S)可以预测在相关临床剂量下的三个指标PS, TSN or TST, CP),靶点表达量(EXT or EXN是独立于其他四个指标但依赖于肿瘤或正常器官的病理生理变化。

 

Figure. 5. The architecture of STAR-guided machine learning (ML) models to directly predict clinical dose/efficacy/safety by forging links among five features based on in vitro/ex vivo, preclinical, and clinical data: Drug structure (S), potency/specificity (PS) to on/off-targets, on-/off-target-driven tissue/cell selectivity in normal (TSN), tumor tissues (TST), or different cell types (cancer cell, stromal cells, immune cells, normal cells), on/off-target expressions in normal (ExN) and tumor (ExT) as well as in various cell types, and plasma drug concentration (Cp) vs. time profile at relevant clinical doses.

 

为了提高药物开发的成功率和效率,我们建议重点关注以下三个研究领域:

 

(1)开发STAR-guided ML模型,通过预测三大相互依存因素 (临床剂量/疗效/安全性)来设计STAR 一类候选药物,从而提高成功率。

 

(2)应用AI机器人帮助药物的快速合成、优化和验证on-/off-target相关的活性/特异性及组织/细胞选择性来提高药物研发效率。

 

(3)快速临床I+期试验,在原发性/转移性肿瘤与健康人(或治愈肿瘤患者中)确证 on-/off-target相关的活性/特异性及组织/细胞选择性来平衡临床剂量/疗效/安全性。

 

这些方法将会帮助选择最佳候选药物,以平衡临床剂量/疗效/安全性,或者发现已批准抗肿瘤药物的新适应症,最终延长肿瘤患者的生命。这些方法可能在后续I/II/III期临床试验中实现70%的成功率,从而将肿瘤药物研发成功率提升七倍,从5%提高到35%,并通过减少时间/成本提高抗肿瘤药物研发的效率。

 

(上下滑动查看更多)

Can Machine Learning Overcome the 95% Failure Rate and Reality that Only 30% of Approved Cancer Drugs Meaningfully Extend Patient Survival?

 

Duxin Sun*, Christian Macedonia, Zhigang Chen, Sriram Chandrasekaran, Kayvan Najarian, Simon Zhou, Timothy A. Cernak, Vicki L. Ellingrod, Hosagrahar V. Jagadish, Bernard Marini, Manjunath P. Pai, Angela Violi, Jason C. Rech, Shaomeng Wang, Yan Li, Brian Athey, Gilbert S. Omenn

 

Dr. Duxin Sun et al from College of Pharmacy, College of Engineering, School Medicine, Michigan Institute of Data Sciences at University of Michigan, together with colleagues from Lancaster Life Science Group, Aurinia Pharmaceuticals, and Bristol-Meyer Squib Company, jointly published a perspective on J Med Chem, 2024, xxx, entitled: “Can machine learning overcome the 95% failure rate and reality that only 30% of approved cancer drugs meaningfully extend patient survival?”

 

Cancer care has significantly advanced over the past 30 years. The overall cancer mortality rate in the U.S. has dropped by 33% from 1991-2020, and  the FDA has approved 250 cancer drugs targeting 120 molecular targets from 2000-2022. Despite this progress, however, the U.S. still had over 610,000 cancer-related deaths in 2023. This equates to 1,670 deaths per day, a figure comparable to the U.S. daily COVID-19 death rate during the peak of the pandemic in 2020. Globally, there were 10 million cancer-related deaths in 2022, which is three times more than the peak COVID-19 deaths during the worldwide lockdown in 2020.

 

Various global research initiatives have been launched to fight the war on cancer. The U.S. reignited the Cancer Moonshot, aiming to reduce the cancer death rate by 50% over the next 25 years. However, the financial burden in cancer care has posed a challenge to achieve the goal of global research initiatives. For instance, the average cost of a course of cancer drug regimen per patient in the U.S. ranges from $170,000 to $277,000. The U.S. spends $200B annually on cancer care, including $60B on cancer drugs, translating to daily costs of $540M and $165M, respectively.

 

In this perspective, the authors focus on discussion of the following questions:

 

• Why does 95% of cancer drug development fail despite significant improvement at each step of the process using hundreds of helpful strategies in the past 30 years?

 

• Why do only 30% approved cancer drugs meaningfully extend patient survival by more than 2.5 months, while the average cost for a cancer drug regimen per patient in the U.S. ranges from $170,000 to $277,000?

 

• Are current strategies to improve each step of drug development process, including AI-driven approaches, falling into “survivorship bias” trap by overly focusing on many less critical issues but overlooking root causes of key aspects?

 

• Is it practical to add more criteria to the already lengthy and costly drug development process, which takes 10-15 years and costs $1-2 billions per one approved drug?

 

• Can application of AI and machine learning (ML) methodology in the current drug development process improve efficiency, boost success rates, and enhance drug’s efficacy?

 

• What are the root causes of these challenges, and what should be the future research priorities to find potential solutions?

 

1. The two interconnected problems of cancer therapeutics.

 

(1). The 95% failure rate of cancer drug development has not improved over the past 30 years.

 

Hundreds of helpful strategies and criteria have been implemented in the drug development process in the past three decades, which includes target validation, high throughput screening, lead compound optimization, preclinical testing, drug-like property optimization, GLP toxicity testing, GMP manufacturer, and clinical phase I-III trials. Despite the improvements in each step of the process, 95% of small molecule cancer drug development fails from clinical phase I to phase III trials. (Figure 1)

 

Figure 1. Cancer drug development has a 95% failure rate despite improvements in each step by incorporating hundreds of helpful criteria over the past 30 years. It is impractical to continually add more criteria without streamlining nonessential ones.
 

Given the lengthy and costly nature of cancer drug development, it is not practical to continually add more criteria to the process, without eliminating non-essential ones. This raises the question: which criteria are nonessential? This high and persistent failure rate, seen in the in the pharmaceutical industry but not others, may suggest that the existing criteria may have overlooked key aspects of the drug development process (Figure 1). More importantly, do these current strategies fall into the “survivorship bias” trap by overly focusing on many unimportant factors but overlooking major deficiencies? This analogy compares the focus on fixing the damage on the wings of the surviving/returned aircrafts where they can sustain damage and still return home, while overlooking the damage to the critical location, such as engines and cockpits, of the non-surviving/un-returned aircrafts during WWII (Figure 2). While many of current criteria in the drug development process are indeed important, many others may just fix non-essential problems and miss the critical aspects in the process similar to the scenario in survivorship bias.

 

Figure 2. Survivorship Bias. The survivorship biased focus is fixing the damage on the wings of surviving/returned aircrafts where they can sustain damage and still return home, but missing the damage in the critical locations of engines and cockpit of the non-surviving/un-returned aircrafts from the battle during WWII.

 

(2). Only 20-42% of newly approved cancer drugs meaningfully extend patient overall survival beyond 2.5 months.

 

Due to unmet clinical needs, many cancer drugs are approved based on clinical phase II trials demonstrating improvements in surrogate endpoints, such as progression free survival (PFS), delaying the time to tumor progression. These drugs are then required to be tested in clinical phase III trials to validate their effectiveness in prolonging patient overall survival (OS). The early access to such drugs may benefit cancer patients who have no other treatment options. However, it is disappointing that only 20-42% of these newly approved cancer treatments meet the clinical benefit threshold in prolonging overall survival beyond 2.5 months, a criterion set by the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO). Approximately 30% of approved cancer treatments did not improve OS, while 40% improved OS by 2 weeks to 2.5 months.

 

Several different scenarios may exist for the lack of meaningful improvement in overall survival (OS>2.5 months). If newly approved cancer drugs show less than 2.5-month OS improvement compared with standard care, they may offer similar effectiveness to the standard care but may have advantages, such as reduced toxicity, which is still meaningful. However, If these newly approved drugs show a less than 2.5-month OS improvement in comparison with an inappropriate control arm, caution should be exercised regarding their efficacy. In many cases, such suboptimal drug control arms with minimal clinical benefit have remained on the market and have even been listed on cancer treatment guidelines. It is very challenging to discern if these control arms would provide meaningful clinical benefits to cancer patients. Further, If the newly approved cancer drugs (or in combination with standard care) show no OS improvement when compared with placebo (or in combination with standard care), their efficacy is questionable.  Unfortunately, many drugs, which fail to improve patient overall survival, still remain on the market for clinical use based on their original “positive” phase II trials, while effective communication with physicians and patients is often lacking

 

Figure 3. Only approximately 30% approved cancer drugs, based on positive improvement of progression-free survival (PFS), meaningfully extend patient overall survival (OS) by more than 2.5 months, while 30% of approved cancer drugs do not extend patient overall survivals.
 

2.The two interconnected problems of cancer therapeutics stem from three overlooked deficiencies that lead to suboptimal dug candidate selection entering clinical trials.

 

The three overlooked deficiencies, including insufficient on-/off-target validation, incorrect application of drug-like property evaluation, and inability of clinical dose optimization,  contribute to the 95% cancer drug development failure rate or the low efficacy/safety of approved cancer drugs.

 

(1). Current drug candidate selection from in vitro target binding IC50s overlooks potency/specificity (PS) against on-/off-target in tumors at relevant clinical doses

 

(2). Current drug candidate selection from drug-like property criteria overlooks on-/off-target-driven tissue/cell selectivity (TS) influencing adverse effects (on/off-target toxicity) in normal organs at relevant clinical doses.

 

(3). Clinical doses cannot be optimized to balance clinical efficacy/safety due to the overlooked optimization of in vivo potency/specificity and tissue/cell selectivity, while extensive GLP animal toxicity testing is not predictive for clinical adverse effects (on/off-target toxicity) at therapeutic dose ranges.

 

We proposed a  “STAR System” (structure-tissue/cell selectivity-activity relationship) to describe how to use these three factors to classify drugs into STAR class I – IV drugs. The STAR system could guide drug development strategies by streamlining the process to improve success rate and efficiency. The goal is to select STAR class I drugs, rather than focusing on STAR class II/IV compounds as seen in the current process.

 

Figure 4. STAR (Structure-Tissue/Cell Selectivity-Activity-Relationship) guides drug development strategies to select STAR class I drugs with high efficacy, high safety, and high success rate, and to avoid class II/IV drugs with low success rates.
 

3. How can machine learning overcome the 95% failure rate and reality that only 30% of approved cancer drugs meaningfully extend patient overall survival?

 

Emerging technologies like artificial intelligence (AI) and machine learning (ML) may improve the efficiency of each step of the drug development process by reducing cost/time if the ML models are correct and validated. However, it is an oversimplification to assume that applying AI and ML techniques could overcome the 95% cancer drug development failure rates if we still rely on the current and same process in the drug development.

 

(1). Apply ML models to improve efficiency of the drug development:

 

ML models have been used to improve the criteria at each step of the current drug development process. A well validated ML model can improve efficiency by saving time and cost, without the need for tedious wet lab experiments, which is very meaningful. Indeed, ML models have been applied at every step in the current drug development process (Figure 1), which includes disease target identification/validation in various cancer types, high throughput screening, drug design and optimization, drug-target interaction (DTI) prediction, pharmacokinetics and drug-like properties prediction, toxicity prediction, and clinical trial designs and pharmacovigilance. Examples of how ML models have impacted each step of the drug development process to improve efficiency by saving time and cost.

 

(2). Develop STAR-guided ML models to improve success rate and efficiency of the drug development:

 

The STAR-guided ML models need to be developed to directly predict the delicate balance of clinical dose/efficacy/safety as defined in the STAR system. The STAR-guided ML models can help to design STAR class I drugs, which  improve success rate and efficiency. Based on our new hypothesis, the STAR-guided ML models should be built to predict clinical adverse effects (AE) (on/off-target toxicity) or clinical efficacy (Eff) by forging links among the following five features with a dynamic weighting (w) based on in vitro/ex vivo, preclinical and clinical data at relevant clinical doses (Figure 5):

 

Clinical adverse effects at relevant clinical doses ŷ(AE) = (PS ˟ TSN ˟ EXN ˟ CP ˟ S)(1)

Clinical efficacy at relevant clinical doses ŷ(Eff) =  (PS ˟ TST ˟ EXT ˟ CP ˟ S)w (2)

 

The STAR-guided ML models can be built in five steps, described below, to predict clinical adverse effects (on and off-target toxicity) ŷ(AE) or clinical efficacy (Eff) at relevant clinical doses (Figure 5). Among five features, the ML models can use drug structure information (S) to predict three features (PS, TSN or TST, CP) under relevant clinical doses. The expression feature (EXT or EXN) is independent of other four features, but dependent on pathophysiology of tumor or normal organs.

 

Figure. 5. The architecture of STAR-guided machine learning (ML) models to directly predict clinical dose/efficacy/safety by forging links among five features based on in vitro/ex vivo, preclinical, and clinical data: Drug structure (S), potency/specificity (PS) to on/off-targets, on-/off-target-driven tissue/cell selectivity in normal (TSN), tumor tissues (TST), or different cell types (cancer cell, stromal cells, immune cells, normal cells), on/off-target expressions in normal (ExN) and tumor (ExT) as well as in various cell types, and plasma drug concentration (Cp) vs. time profile at relevant clinical doses.

 

To enhance both the success rate and efficiency of drug development, we recommend focusing on the following three research areas:

 

1.Develop STAR-guided ML models to design the STAR I drug candidates by predicting clinical dose/efficacy/safety based on the three interdependent factors, thereby improving the success rate.

 

2.Apply AI robotics to accelerate drug development through robotics-powered synthesis, optimization, and testing of on-/off-target-driven potency/specificity and tissue/cell selectivity.

 

3.Conduct accelerated clinical phase I +  trials in primary/metastatic cancer vs. healthy (or cancer patients in remission) to identify on-/off-target engagement and on-/off-driven tissue/cell selectivity, balancing dose/efficacy/safety.

 

These approaches will select the best drug candidates that balance clinical doses/efficacy/safety, and/or discover new indications for approved cancer drugs, ultimately extending cancer patient survival. These approaches may improve success rate from 5% to 35% by achieving 70% success in each of Phase I/II/III trials, and enhance the efficiency of cancer drug development by reducing time/cost.

Related News

Contact us

Address:Room 62, 6th Floor, Building 1, Zone 1, No.186 South 4th Ring West Road , Fengtai District, Beijing

Tel:010-83634390

Address:Address:Room 1704, Building E, Nanotechnology Park, SIP, Suzhou, Jiangsu Province

TONACEA

TONACEA

XIEYI Release

TONACEA

TONACEA Biotech

TONACEA

TONACEA Micro Service

TONACEA

©2022 TONACEA(beijing)Technology Development Co., Ltd

xueqiu.com zhihu.com MicroBlog