News
资讯&洞见
当前位置: 首页 资讯&洞见 洞见
N-of-1 Clinical Trials(Series II)—— Nonclinical Studies
2024-12-04
Milasen and Its Successors Paving the Way for Accessible Patient-Customized Oligonucleotide Therapies.

N-of-1 or single subject clinical trials consider an individual patient as the sole unit of observation in a study investigating the efficacy or adverse effects of different interventions. The ultimate goal of an N-of-1 trial is to determine the optimal or best intervention for an individual patient using objective data-driven criteria


The advent of Milasen, a groundbreaking patient-customized antisense oligonucleotide therapy, has heralded a new era in personalized medicine for rare genetic disorders. This pioneering approach, exemplified by Milasen and its successors, not only offers hope to patients with previously untreatable conditions but also raises critical questions about the regulatory framework governing such individualized treatments. As the field of patient-specific oligonucleotide therapies rapidly evolves, it becomes increasingly crucial to comprehend the FDA's guidances and their far-reaching implications for future N-of-1 clinical trials.

TriApex, leveraging its extensive experience in nonclinical research on oligonucleotide drugs, has proactively initiated nonclinical toxicology studies for certain projects focusing on Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases (SDLT). These studies, conducted in accordance with FDA guidelines, represent a significant effort to expedite the commencement of clinical trials for these innovative therapies. 

This comprehensive review begins with Mila's compelling story and further explores the regulatory considerations emerging from FDA guidances related to patient-customized oligonucleotide therapies. We provide an in-depth analysis of key aspects outlined in these guidances, including the intricate design of N-of-1 clinical trials, essential nonclinical studies, and critical Chemistry, Manufacturing, and Controls requirements. By dissecting the regulatory landscape, we aim to elucidate a clear path forward for making these innovative, tailored therapies more accessible to patients with rare genetic conditions. By bridging the gap between cutting-edge science and regulatory frameworks, we strive to accelerate the development and delivery of life-changing treatments to those who need them most, potentially transforming the landscape of rare disease management.


Mila's Story: A Race Against Time

Timeline from the initial clinical diagnosis to the initiation of treatment[1]



The Disease Discovery

In 2016, a 6-year-old girl named Mila began showing signs of a rare and fatal neurodegenerative condition. Her symptoms started subtly at age 3 with an inward-turning foot, progressed to vision problems at 4, and by 5, she was experiencing language regression and clumsiness. As she approached her 6th birthday, Mila's condition rapidly deteriorated, leading to hospitalization due to vision loss, frequent falls, and difficulty speaking and swallowing.

After extensive testing, a skin biopsy revealed abnormal lysosomal inclusions characteristic of Batten's disease. Genetic testing identified one known pathogenic mutation in the MFSD8 gene, but the second mutation remained elusive. Determined to find answers, Mila's family enrolled her in a research study at Boston Children's Hospital.



The Breakthrough

Using whole-genome sequencing (WGS), researchers discovered a novel mutation: an SVA retrotransposon insertion in MFSD8 intron 6. This insertion caused missplicing of the gene, leading to Mila's condition. With the genetic cause identified, the race was on to develop a treatment.


The Birth of Milasen

Inspired by the success of nusinersen, an antisense oligonucleotide (ASO) for spinal muscular atrophy, researchers designed a custom ASO to correct Mila's specific mutation. After testing multiple candidates, they identified the most effective one, naming it "milasen" after Mila.

Milasen Development[1]


From Lab to Treatment in Record Time

In an unprecedented feat, researchers moved from genetic discovery to clinical treatment in just one year. They conducted rapid toxicology studies, manufactured the drug, and obtained FDA approval for an expanded access investigational new drug application.


A Glimmer of Hope

Mila began receiving milasen treatments in January 2018, when she was 7 years old. The drug was administered intrathecally, with doses gradually increasing over time. Within the first year of treatment, Mila showed no SAEs, and her seizure frequency and duration decreased significantly.

The dosing schedule of milasen[1]


The concentration of milasen in cerebrospinal fluid (CSF) before each administration (trough) and at day 174 without concurrent dose administration. [1]

The trends in seizure frequency and duration as reported in a seizure diary recorded by the parents[1]

The trends in seizure activity as detected by electroencephalography (EEG)[1]

While Mila's story represents a groundbreaking approach to personalized medicine for rare genetic diseases, it also highlights the challenges and ethical considerations in developing patient-specific treatments. Mila's journey offers hope for other patients with rare genetic conditions and paves the way for future advancements in tailored therapies.



Part I:IND Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases:  Clinical Recommendations Guidance for Sponsor-Investigators

The guidance provides recommendations for managing administration, dosing, and clinical monitoring of individualized ASO drug products for SDLT genetic diseases with no alternative treatments. It focuses on ASOs targeting unique genetic variants specific to 1-2 patients, not larger patient populations with same disease[2].

1

Ethical and Human Subject Protection

▶The protocol must be reviewed and approved by an institutional review board (IRB) before proceeding, per 21 CFR part 56.

▶Informed consent must be obtained, including risks, potential benefits, and providing updates.

▶If the individualized ASO drug product is intended for use in children, there are some special considerations

  • The requirements under 21 CFR part 50, subpart D (Additional Safeguards for Children in Clinical Investigations) must be met. 

  • This subpart provides additional protections for children involved as subjects in clinical investigations, including obtaining appropriate informed consent/assent, and having procedures to respect child dissent when applicable. 

  • The IRB review is critical for studies involving children.

2

Diagnostic and Genetic Considerations

▶Confirm the patient's clinical and genetic diagnosis through relevant laboratory testing(e.g., gene sequencing, enzymatic analysis, biochemical testing, imaging evaluations, as applicable).

▶Provide evidence linking the targeted gene variant(s) to the patient's disease pathogenesis. If the evidence is insufficient, additional investigation(s) may be needed before initiating use of the ASO drug product.

▶Show the targeted variant(s) are unique to the patient, including: 

  • prevalence estimates of the targeted gene variant(s) in the disease

  • the projected incidence of the targeted variant(s) in new cases of the disease based on molecular genetic considerations (e.g., type of mutation (missense, frameshift, other), location in the protein).

3

Dosing Considerations

Starting dose

  • Starting dose should be based on available nonclinical data and be scientifically supported by available information from other ASO drug products in the chemical class and/or prior clinical experience. 

  • In general, the starting dose should be expected to have a pharmacologic effect, based on available nonclinical in vitro and/or in vivo data.

Dose escalation approach

Given the limited nonclinical data available when initiating dosing, the sponsor should consider a dose escalation approach. Dosing should be initiated at a low level and escalated to higher doses cautiously based on:

  • Pharmacodynamic (PD) effects observed

  • Trial participant's response (tolerability or perceived benefit)

  • Available nonclinical data

The protocol should clearly outline:

  • The starting dose and justification for selecting it

  • The dosing interval.

  • The plan for dose escalation.

  • The plan for dose reduction in case of toxicity.

If a loading dose is proposed, the protocol should contain a rationale for using a loading dose(s) and maintenance dose interval(s), providing a rationale based on:

  • The ASO's half-life.

  • Likely distribution to target areas (e.g. CNS, kidney).

  • Anticipated toxicities at different dose levels.

The dose escalation should be done cautiously(e.g.,by no more than a two-fold increment at a time), allowing sufficient time to observe the response to a dose change before making further dose modifications.

  • If nonclinical toxicology studies show a steep dose- or exposure-response relationship for severe toxicities, or if no predictive marker of severe toxicity is available in humans, the sponsor should escalate the dose using smaller increments rather than doubling the dose (e.g., fractional increments rather than dose doubling).

  • In general, concurrent changes to both the dose amount and the dosing interval should be avoided unless there is a justification provided.

Dose de-escalation or discontinuation

Dose de-escalation or discontinuation may be necessary to manage unacceptable toxicities and maintain an appropriate benefit-risk profile for the individualized ASO drug product.

  • The protocol should clearly specify a plan for dose de-escalation or discontinuation if a dose-limiting toxicity develops during the trial. Having a pre-specified plan in the protocol allows for proactive management of dose-limiting toxicities that may emerge during the trial.

  • If a decision is made to de-escalate or discontinue the dose due to a dose-limiting toxicity, the sponsor should inform the trial participant or the participant's legally authorized representative about the reason(s) for making such dose changes. 

4

Drug Product Administration Procedures

♦ Administer starting dose and escalations in inpatient (hospital) setting with close monitoring initially.

♦ For intrathecal, use experienced personnel and have sedation/interventions available.

♦ For at least 24 hours after administration, the participant should be closely monitored for acute, serious  adverse events (AEs)  that may be precipitated by:

  • The administration procedure itself (e.g. intrathecal administration)

  • The ASO drug product (e.g. hemodynamic instability, severe hypersensitivity/anaphylaxis, CNS AEs)

♦ After the safety and tolerability at a given dose level is adequately characterized and any AEs are identified and can be appropriately monitored, subsequent doses at the same level can generally be administered in an outpatient (clinic) setting, if needed. Appropriate safety monitoring and interventions should be available in the outpatient setting for possible serious AEs like anaphylaxis.

For intrathecal ASO drug products, experienced medical personnel should perform the intrathecal administration and administer sedation and/or analgesics if needed for the procedure. Appropriate medical monitoring, procedural interventions, and therapeutic interventions should be available in case of complications from the intrathecal procedure and/or sedation.

The guidance recommends an inpatient setting for initial doses and dose escalations to closely monitor for serious AEs, with a potential shift to outpatient setting once the safety at a dose level is characterized. It also outlines specific considerations for intrathecal administration.

5

Safety Assessment

According to the FDA guidance, the key recommendations for safety assessments of investigational ASO drug products are:

♦ Perform routine safety assessments (e.g. AEs, vital signs, ECG, lab tests) while the ASO is being administered and for a period after stopping, based on the anticipated half-life. 

♦ Safety monitoring should be informed by:

  • Nonclinical toxicology findings.

  • Potential off-target gene effects predicted by bioinformatics tools.

  • Previously identified risks with other ASOs of the same class.

♦ Include a detailed schedule of safety assessments in the protocol.

Conduct safety assessments more frequently during the initial dosing period (e.g. first 2-3 months). At minimum, assess before each dose or more frequently if long dosing intervals.

♦ Based on initial results, the frequency of assessments can then be decreased. Clearly outline this in the protocol.

♦ For phosphorothioate (PS) or mixed-backbone ASOs, perform platelet counts at least every 2 weeks or before each dose, whichever is more frequent, due to thrombocytopenia risk. Closely monitor for abnormal findings/bleeding.

♦ For intrathecally administered ASOs, collect cerebrospinal fluid before each dose to evaluate for inflammation/infection. Use remaining samples for PK or biochemical assessments if feasible.

♦ If ASO administration is discontinued for safety reasons, continue safety monitoring until adverse event resolution and evaluate for potential late-onset toxicities.

6

Clinical Response Assessment

Assessing whether the ASO drug product is providing clinical benefit to the participant is an important element throughout the administration period. The guideline emphasizes careful tracking of clinical status, use of relevant assessments/biomarkers, continual benefit-risk evaluation, and having criteria for dosing adjustments or discontinuation based on clinical response.

  • To inform decisions on continuing administration, sponsors should carefully track clinical changes over time and have a specific plan for adjusting the dosing regimen to achieve a desirable response. The protocol should clearly outline the specific methods and timing for clinical assessments.

  • Clinical assessments can include outcome scales, performance tests, or patient/caregiver reported symptom changes to follow important disease symptoms during ASO administration.

  • Assessments will depend on the participant's pre-treatment signs/symptoms, underlying disease stage/severity at initiation. 

  • Administer assessments at specified intervals determined by disease pathophysiology as well as PK/PD of the ASO drug product.

  • Pharmacodynamic biomarkers (lab tests, imaging, etc.) scientifically justified and relevant to the disease may help assess response, with consideration of assay validation issues.

  • Have a pre-specified plan to continually assess clinical response to ensure benefit-risk remains favorable for continued administration.

  • Consider discontinuing if there is inadequate clinical response or if toxicities outweigh observed clinical benefits.

In summary, the key points cover ethical considerations, diagnostic confirmation, dosing strategy, administration procedures, safety monitoring, and assessing clinical response for these individualized ASO drug products in this guidance. Based on this guidance, valuable experience will be accumulated through practice, providing reference for the development of individualized ASO drug products in the future, and further improving this guidance as well.



下期预告:N-of-1 Clinical Trials(Series II)—— Nonclinical Studies

往期精选

1、他山之石-3丨siRNA 药物 Lumasiran 上市历程及临床开发路径总结
2、鼎泰助力 | 热烈祝贺艾码生物自组装核酸药物 ER2001 注射液获 NMPA&FDA IND 许可
3、鼎泰助力 | 热烈祝贺中因科技 AAV 治疗药物 ZVS203e 注射液获 NMPA&FDA IND 许可


Reference:

[1] J. Kim, C. Hu, C. Moufawad El Achkar, et al. Patient-Customized Oligonucleotide Therapy for a Rare Genetic Disease[J]. N Engl J Med 2019;381:1644-52.

[2] FDA. IND Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases:Clinical Recommendations. Dec, 2021.

[3] FDA. Nonclinical Testing of Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life Threatening Diseases Guidance for Sponsor-Investigators. Apr, 2021

[4] FDA. Investigational New Drug Application Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases:  Chemistry, Manufacturing, and Controls Recommendations Guidance for Sponsor-Investigators. Dec, 2021


分享文章:
上一篇 没有了
推荐新闻
N-of-1 Clinical Trials(Series II)—— Nonclinical Studies
2024-12-04
N-of-1 Clinical Trials(Series I)
2024-11-29
众里寻他千百度 —— 首个获批上市的 TCR-T 非临床研究思路和临床开发路径
2024-11-11
探寻行业前沿、聚焦客户需求 —— 专业的见解看这里!鼎泰集团寡核苷酸药物生殖发育毒性研究
2024-10-09
CNS 寡核苷酸药物早期研发:Arrowhead 的经验
2024-10-08
不怕贴秋膘! ALK7 siRNA 脂肪递送、精准减脂,鼎泰 KBI 打造影像学小蛮腰
2024-09-24
寡核苷酸药物:致癌性试验一般考虑
2024-09-12
PD-L1 表达阳性转移性 NSCLC,依沃西单抗(AK112)头对头对比帕博利珠单抗 III 期临床试验期中分析和研发历程回顾
2024-09-09
ANGTPL3 siRNA,降脂新思路
2024-08-30
鼎泰智声 | 毒理学研究结果与首次人体试验(FIH)起始剂量设计、剂量优化
2024-08-30
他山之石-5丨siRNA 迭代药物 Vutrisiran 上市历程及临床开发路径总结
2024-08-23
选择困难?NHP ePPND,鼎泰准备好了!
2024-08-23
减脂保肌,INHBE 有态度,KBI 有招数
2024-07-15
寡核苷酸药物临床药理学考量和案例分析
2024-07-08
寡核苷酸药物的中枢直接递送技术和常见组织病理学改变
2024-07-08
他山之石-4丨siRNA 药物 Inclisiran 上市历程及临床开发路径总结
2024-07-08
ADC 临床剂量优化的一般考虑
2024-07-08
他山之石-3丨siRNA 药物 Lumasiran 上市历程及临床开发路径总结(下)
2024-05-17
他山之石-3丨siRNA 药物 Lumasiran 上市历程及临床开发路径总结(上)
2024-05-17
前沿资讯 | 17th WRIB 白皮书part 3精彩议题回顾
2024-04-30
他山之石-2 | siRNA 药物 Givosiran 上市历程及临床开发路径总结
2024-04-30
他山之石丨6 款 siRNA 药物上市历程及临床开发路径总结(2) —— Givosiran(Givlaari®)
2024-04-30
他山之石丨6 款 siRNA 药物上市历程及临床开发路径总结(1) —— Patisiran(Onpattro®)
2024-04-30
siRNA药物开发不容忽视的关键点 —— 非临床药效学研究
2024-04-02
siRNA药物非临床研究价值的延伸 —— 杂质控制
2024-04-02
临床开发失败siRNA:复盘与启示
2024-04-02
靶向FRα的Mirvetuximab soravtansine (Elahere)的上市路径回顾和对ADC药物非临床开发的启示
2023-12-29
全球已上市ADC药物非临床研究路径调研(3) — 拓扑异构酶抑制剂篇
2023-12-29
全球已上市ADC药物非临床研究一览(2) ——微管抑制剂篇
2023-12-05
全球已上市ADC药物非临床研究一览(1)
2023-11-23
药政解读 | CDE《人源干细胞产品非临床研究技术指导原则(征求意见稿)》学习领会和相关案例分析
2023-10-13
前沿资讯 | FDA CGT产品非临床评价网络研讨会复盘与鼎泰理解(第4期) 及全部会议内容简述
2023-09-27
前沿资讯 | FDA CGT产品非临床评价网络研讨会复盘与鼎泰理解(第3期)
2023-09-22
前沿资讯 | FDA CGT产品非临床评价网络研讨会复盘与鼎泰理解(第2期)
2023-09-14
前沿资讯 | FDA CGT产品非临床评价网络研讨会复盘与鼎泰理解(第1期)
2023-09-14
ICH S12《基因治疗产品非临床生物分布的考虑》要点汇总(上)
鼎泰集团生物分析团队对 S12 进行了要点汇总,并结合已上市 GT 产品的生物分布研究对该指南中的相关内容进行梳理,以期进一步加深对指南的理解和运用,与同业者沟通、交流。
2023-07-07
寡核苷酸类药物免疫原性风险及评估策略
本文简要综述影响寡核苷酸药物免疫原性的因素,以及评估时的考虑因素和分析方法检测策略。
2023-07-07
眼科专题 | 干细胞产品治疗视网膜变性
2023-07-07
浅谈ASO药物非临床安全性评估中的主要考量因素
本文汇总了已上市ASO药物的临床不良反应,并通过对上述药物非临床研究结果的回顾,分析了非临床研究在指导ASO药物临床开发中的价值和局限性,探索非临床安全性评估中主要的考量因素,以期为后续ASO新药的开发与评估提供一定的参考。
2023-07-07
从iPSCs到类器官, 浅析专病领域药物研发的机遇和挑战
类器官(Organoids)作为干细胞研究领域最重要的成果之一,在基础医学研究、转化医学及药物研发领域展现出巨大的应用潜力,特别是在精准医疗以及药物安全性和有效性评价等方向凭借其先天优势引起了极大的市场关注,成为各大药物研发机构、CRO公司争相布局的战略点。
2023-07-07
泪液的采集方法、处理过程及不同采样方式对检测结果的影响分析
眼睛分泌的泪液不仅具有湿润眼球和免疫与防御功能,还含有大量与人体生理病理相关的信息。摄入的药物或毒品也可能经由泪液分泌排出。此外,由于眼睛直接暴露于空气中,空气中的挥发物、气溶胶与可溶性颗粒也可能进入眼睛而转移至泪液中。所以,泪液的成分分析在疾病诊断、药物分析、毒品检测以及环境健康等相关领域具有重要意义。因此,可通过泪液建立非侵入性诊断方法,能够加深我们对眼部和全身性疾病的了解。
2023-01-05
CDE关于ICH S1/S1B(R1)药物致癌性试验指导原则宣讲会回顾及FDA专家对减少致癌试验动物使用最新指南的解读
随着药物研发的推进,致癌性试验成为支持部分药物上市申请的非临床安全性评价的重要内容之一。鼎泰团队对药物致癌性试验的必要性、致癌性试验的设计和数据分析以及致癌性试验相关的资源可及性进行了充分的调研。本文通过回顾《ICH S系列指导原则宣讲会》,及FDA对减少致癌试验动物指南的解读,提出鼎泰团队对药物致癌性试验的思考和建议,力求为国内同行、新药研发企业和审评机构提供参考。
2022-11-23
GalNAc-siRNAs药物ADME特性及PK-PD模型
寡核苷酸药物研发近年来发展迅速,目前FDA已批准 10款反义核苷酸(ASO)和5款siRNA药物上市。在寡核苷酸药物研发过程中,递药系统起着关键作用,其中GalNAc  (N-乙酰化的半乳糖胺)直接与寡核苷酸结合或将其修饰到特定的递送系统作为靶向部分,以nM级高亲和性结合ASGPR,在蛋白介导作用下将GalNAc及核酸摄取进入肝细胞,是当前最常用的小核酸药物递送系统之一
2022-11-23
订阅我们的新闻,实时了解最新资讯,立即订阅