News & Insight
N-of-1 Clinical Trials(Series Ⅲ)
2024-12-13

Milasen and Its Successors Paving the Way for Accessible Patient-Customized Oligonucleotide Therapies.

Regulatory Insights from FDA Guidances on Patient-Customized Oligonucleotide Therapies for SDLT represent a significant effort to expedite the commencement of clinical trials for these innovative therapies. 

In Series I & , we shared Mila's compelling story and the development history of milasen, while also exploring the FDA guidance on "IND Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases: Clinical Recommendations Guidance for Sponsor-Investigators" and "Nonclinical Testing of Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases Guidance for Sponsor-Investigators". Those guidances offers recommendations for the nonclinical testing and clinical monitoring of individualized ASO drug products intended for SDLT genetic diseases that lack alternative treatments.

In Series Ⅲ, we will further provide an in-depth analysis of key aspects outlined in another FDA guidance document, "Investigational New Drug Application Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases: Chemistry, Manufacturing, and Controls Recommendations." This document emphasizes the fundamental CMC requirements necessary to support an IND application for ASOs developed to treat SDLT diseases.


PART III. Investigational New Drug Application Submissions for Individualized Antisense Oligonucleotide Drug Products for Severely Debilitating or Life-Threatening Diseases: Chemistry, Manufacturing, and Controls Recommendations

The purpose of this FDA guidance is to provide recommendations on the chemistry, manufacturing, and controls (CMC) information that should be included in an IND application submitted by a sponsor-investigator developing an individualized ASO drug for a SDLT disease caused by a unique genetic variant affecting only a small number of individuals (typically 1-2 patients).

The guidance covers individualized unconjugated ASO products manufactured using conventional methods with simple aqueous or lyophilized powder formulations to be reconstituted before administration. These individualized ASO drug should be from a well-characterized chemical class for which there is substantial clinical and nonclinical experience that is either publicly available or to which the sponsor has a right to reference [4].

The guidance provides recommendations on CMC information for the IND, including:


◆ Nomenclature, structure, and general ASO drug substance properties 

◆ Manufacture

◆ Characterization

◆ Control of excipients

◆ Control of drug substance and drug product

◆ Reference standards or materials

◆ Container closure systems

◆ Stability  


The CMC recommendations support FIH exposure but do not address long-term administration, non-SDLT diseases, broader patient populations beyond 1-2, or commercial development requirements.



1

General considerations on CMC 

information for an initial IND


A. Regulatory considerations

♦ For drug products intended to treat SDLT diseases like individualized ASOs, FDA allows flexibility in requirements while preserving appropriate safety and efficacy guarantees per 21 CFR 312 Subpart E.

♦ Sponsors, including sponsor-investigators, are generally required to submit an IND with CMC information per 21 CFR 312.23(a)(7) to ensure proper identification, quality, purity and strength of the investigational drug product.

♦ This guidance provides recommendations on satisfying the general CMC requirements for an IND for individualized ASO products within its scope.

♦ For phase 1 clinical trials, investigational drug production is generally exempt from full compliance with 21 CFR 211 cGMP regulations, as described in the guidance "cGMP for Phase 1 Investigational Drugs."

♦ Individualized ASOs may not follow the traditional phase 1-3 clinical development

  • For the first clinical batch, manufacturing per the Phase 1 cGMP guidance is generally acceptable.

  • For subsequent clinical batches needed for continued administration, FDA generally expects compliance with 21 CFR 211 cGMP per the "Preparation of Investigational New Drug Products" guidance to ensure consistent quality.


2

CMC Information for Drug Substance


A. Description of the Drug Substance

The sponsor must provide a detailed description of the drug substance, including its structure, nomenclature, structural formula, molecular formula, molecular weight, and, if applicable, the molecular weight of the salt form. Additionally, the sponsor should include information about the base moieties and backbone, carbohydrate moieties, internucleoside linkages, and counter ions relevant to the individualized ASO drug substance. Physical properties such as hygroscopicity, aqueous solubility, and melting temperature (Tm) should also be reported.

B. Manufacturer Information

The sponsor must submit the full street address of the drug substance manufacturer, including any contract manufacturers or testing laboratories involved in producing the batches for clinical trials.

C. General Method of Preparation

Submissions should provide a flow diagram and a comprehensive narrative describing the manufacturing process, including purification steps. The diagram must detail all representative coupling/chain elongation and deprotection steps, along with any purification or impurity reduction processes (e.g., chromatography, lyophilization, solvent removal, desalting). The narrative should outline the chemical structures and configurations, including stereochemical details for starting materials, intermediates, and significant side products. Unique or critical manufacturing steps must be described in detail. While the FDA does not require identification of specific column or equipment model numbers, the IND must include a clear description of process controls that ensure drug substance quality (e.g., impurity clearance). Lastly, a list of materials used in the manufacturing process (starting materials, reagents, solvents, auxiliary materials) should be provided. For sterile drug substances, a description of the sterilization process (e.g., moist or dry heat terminal sterilization, aseptic filtration) is necessary, but validation information is not required.

D. Characterization

The sponsor must confirm the chemical structure of the drug substance using various physical and chemical techniques, including nucleotide sequencing, melting temperature (Tm) analysis, and mass spectral analysis. Sequence determination for the ASO drug substance is required; if not included in the initial IND, the sponsor must provide a justification and submit this information in an amendment as soon as possible. Additionally, the sponsor should detail impurities associated with the ASO drug substance, summarizing both actual and potential impurities that may arise during manufacturing, purification, and storage. The FDA recommends categorizing ASO-related impurities according to their structural class or retention times.

The sponsor is also responsible for discussing non-ASO related impurities (such as elemental impurities and residual solvents) and outlining measures for controlling these impurities. The submission must include specifications for these non-ASO impurities or a scientific rationale for why testing is not necessary, such as descriptions of manufacturing steps that remove specific non-ASO impurities. If the clinical batch differs from the nonclinical batch, comparative data (e.g., HPLC chromatograms) demonstrating the quality of both batches—such as homogeneity and purity—should be provided.

E. Control of Drug Substance

1. Specification

This section should present a comprehensive table of specifications for the drug substance batch, detailing tests, acceptance criteria, and references to the analytical procedures used. The sponsor should briefly describe the analytical methods employed. In addition to guidelines for IND submissions, the specifications should include:

♦ Identification of the ASO drug substance, using a combination of methods for verification (e.g., sequencing and molecular weight determination).

♦ Testing for the salt form (if applicable).

♦ A strength assay that reports the full-length drug product content, excluding any P=O impurities.

♦ Quantities of specified impurities and total impurities content.

♦ Residual solvents, moisture content, microbiological testing (e.g., microbial limits and sterility), and bacterial endotoxins.

♦ The certificate of analysis (CoA) for both the proposed clinical batch and any nonclinical batches must also be included.

F. Stability

The sponsor should provide stability information for the synthetic ASO drug substance, including available stability data and the monitoring protocol for ongoing stability. Preliminary data should be presented in a tabular format, along with a description of the container closure system used.


3

CMC Data for Drug Product

A. Components

The sponsor must provide a comprehensive list of all components utilized in the manufacture of the investigational drug product, including both those intended for the final product and those used in the manufacturing process. For inactive ingredients, quality should be referenced through a compendial monograph (e.g., USP or National Formulary) or include the supplier's Certificate of Analysis (CoA).

B. Quantitative Composition

A brief summary, preferably in table format, of the individualized ASO drug product composition should be submitted. This summary must detail the amounts of any processing aids used and those removed during the manufacturing process (e.g., Water for Injection).

C. Manufacturer Information

The full street address of the drug product manufacturer must be provided, including any contract manufacturers, packing facilities, or testing laboratories involved in producing the clinical trial batches.

D. Manufacturing and Packaging Procedures

A flow diagram and a concise written description outlining the manufacturing process should be submitted, detailing any bioburden reduction and sterilization steps (e.g., membrane filtration, terminal sterilization). The description must also specify the air classification of the manufacturing rooms (e.g., Class 100, Grade A, ISO 5).

E. Control of Drug Product

The IND should include detailed specifications and corresponding test methods. The drug product must be tested for identity, strength, impurities, foreign and particulate matter, sterility, bacterial endotoxins, and any dosage form-specific tests. Acceptance criteria for bacterial endotoxins should be determined based on the proposed maximum dose and administration route, as specified in relevant guidelines.

F. Container Closure System

A description of the container closure system for the individualized ASO drug product should be included, identifying the materials used for each primary packaging component (e.g., USP Type 1 glass vial, bromobutyl rubber stopper). Specifications for each component or the manufacturer's CoA should be provided, along with methods for depyrogenation and sterilization for non-sterile components.

G. Stability

The sponsor must monitor the stability of the individualized ASO drug product in its proposed container under specified storage conditions. A stability protocol for the clinical batch should be included, detailing the stability study, test methods, and any initial stability data in tabular format. If modifications to the drug product are made prior to use (e.g., reconstitution or dilution), guidelines for storage duration to minimize microbial contamination should be outlined, along with justifications for any extended storage conditions.


4

Immediate Packaging Labeling

The sponsor is required to submit a copy of the proposed immediate packaging label as part of the IND submission. This label must prominently feature the statement: “Caution: New Drug — Limited by Federal (or United States) law to investigational use.”

5

Environmental Exclusion

IND sponsors must either claim a categorical exclusion from environmental analysis requirements under 21 CFR 25.30 or 25.31, or provide a comprehensive environmental assessment as outlined in 21 CFR 25.40. For an individualized ASO drug product under an IND, it is recommended that the sponsor include a claim for categorical exclusion under 21 CFR 25.31(e).

TriApex's commitment to advancing these cutting-edge treatments demonstrates our dedication to addressing urgent unmet medical needs. By leveraging our deep expertise in oligonucleotide drug development and aligning our research with regulatory guidelines, we aim to accelerate the translation of these promising therapies from bench to bedside.

We sincerely hope that these tailored oligonucleotide drugs will offer promising treatment options for patients with urgent needs, potentially transforming the landscape of care for those suffering from severe and life-threatening conditions. Our efforts underscore the potential of personalized medicine to provide hope and improved outcomes for individuals facing critical health challenges.



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:
推荐新闻
N-of-1 Clinical Trials(Series Ⅲ)
2024-12-13
N-of-1 Clinical Trials(Series II)—— Nonclinical Studies
2024-12-04
N-of-1 Clinical Trials(Series I)
2024-11-29
Analysis of the influence of different tear collection, processing and sampling methods on the results
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
Review of CDE on the ICH S1/S1B(R1) Testing for Carcinogenicity of Pharmaceuticals and interpretation by FDA experts of the latest guidelines for reducing the use of carcinogenic laboratory animals
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
ADME characteristics and PK-PD models of GalNAc-siRNAs
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