The United States Food and Drug Administration’s Platform Technology Designation to Expedite the Development of Drugs
Abstract
:1. Background
- Designated platform technology: A technology that meets the eligibility factors for designation under section 506K(b), (d), and (h) of the FD&C Act;
- Platform technology: A well-understood and reproducible technology essential to a drug’s structure or function, adaptable for multiple drugs, and facilitating standardized production or manufacturing processes;
- Preliminary evidence: Information from tests or studies comparing the platform technology’s use in approved and proposed drugs;
- Prior knowledge: Expertise and understanding gained from developing similar products and processes, including established scientific principles;
- Significant efficiencies: Leveraging previous tests or processes to streamline drug development, manufacturing, and review;
- Drug: For this guidance, the terms drug, drug product, and product refer to a drug as defined in section 201(g)(1) of the FD&C Act (21 U.S.C. 321(g)(1)). This includes biological products as defined in section 351(i) of the Public Health Service Act (PHS Act) (42 U.S.C. 262(i)). The term drug also applies to a drug or biological product constituent part (21 CFR 4.2) of a combination product being developed for review under section 505 of the FD&C Act (21 U.S.C. 355) or section 351 of the PHS Act.
2. Introduction
3. Potential Benefits of a Platform Technology Designation
- Engaging in early interactions with the FDA to discuss using a platform technology, including information relevant to establishing, as applicable, safety, purity, potency, or quality.
- Receiving timely advice from and engaging with the FDA during the development program, such as additional interactions and/or meetings on the use of the platform technology. Depending on resources, the FDA might prioritize interactions or additional engagements regarding a designated platform technology for those products where the FDA has determined the most significant public health benefit or impact.
- Leveraging data from a prior product that used the designated platform technology, such as leveraging batch and stability data from a related product as prior knowledge that can supplement product development studies (e.g., in-use stability studies to define administration conditions and/or light exposure studies to inform the design of the container closure system) or support shelf-life extrapolation and determination for structurally alike products.
- Leveraging certain nonclinical safety data from prior products that used the designated platform technology such that a product-specific assessment for specific, designated endpoints might not be warranted.
- Considering previous inspection findings by the FDA for subsequent marketing applications related to the manufacture of a drug that incorporates or uses the designated platform technology.
4. Eligibility for the Platform Technology Designation Program
- Structurally similar drug substances, such as similarly sized nucleic acid sequences with comparable backbone chemistry, subunit modifications, and targeting moieties;
- Minimal qualitative and quantitative differences in drug product formulation; and/or
- Nearly identical manufacturing processes for drug substance and/or drug product manufacturing and purification.
5. Designation Request
5.1. Recommended Content for a Designation Request
- Specifically, the request should explain how the technology meets the definition under 506K(h)(1) and how it is eligible under 506K(b).
- Identification of an approved application (NDA, BLA, or ANDA) where the technology was incorporated, with applicable cross-references to other applications or submissions that the sponsor owns or has full right of reference to as part of a business agreement, and an appropriate eCTD link to the relevant and identified information (in INDs, NDAs, BLAs, or ANDAs).
- Identification of the shared structural element between drug products and how the shared structural element facilitates the use of the platform technology. Such a demonstration for a shared element could be based on a logical assertion supported using relevant prior knowledge and/or experimental studies.
- Justification and scientific support for using a platform technology across multiple drugs, including how utilizing the technology in subsequent proposed products would not affect safety, quality, or manufacturing. The justification should include information to demonstrate, for example, how the technology can be incorporated into other drugs with no or only minor differences in the relevant parts of the manufacturing process, as well as how the technology functions and is appropriate for the safety and quality profile.
- Risk assessment to evaluate how the differences between a prior product and the subsequent proposed product (for this guidance, a subsequent proposed product is a proposed drug product that is the subject of a marketing application and/or a candidate product that is the subject of an IND application) could affect the use of the platform technology; the relevance of prior information; and, therefore, how much prior information would be appropriate to be leveraged in support of the subsequent proposed product.
- Information to justify why the use of the platform technology would bring significant efficiencies to the drug development or manufacturing process and to the review process for the application (e.g., allow for testing or validation performed as part of the development of one of the products to reduce testing or validation for the other products and, thus, increase efficiency). The ability to reduce testing and validation for manufacturing and/or analytical methods will depend on the drug class. Whether the reduction in testing or validation constitutes a significant efficiency would depend on the nature of the testing or validation.
5.2. Submitting a Designation Request
5.3. Timing of Designation Request Submissions by the Requester and Timeline for FDA Evaluation of Designation Requests
6. Post-Approval Changes to a Designated Platform Technology
7. Revocation of a Platform Technology Designation
8. General Considerations for Eligibility
8.1. LNP Platforms
- a.
- Composition, including type, amount, and manufacture of the lipids;
- b.
- Manufacturing process unit operations (e.g., transcribing RNA, synthesizing lipid moieties, and formation of the lipid nanoparticles) that are not sensitive to inputs (e.g., template sequences) and yield consistent outputs across multiple products and where sequence differences of the mRNA do not affect product quality;
- c.
- Manufacturing process parameters, in-process controls, and equipment critical to the manufacture of the mRNA LNP vaccine or gene therapy;
- d.
- Process-related impurity clearance across a defined downstream purification process.
- e.
- Composition, including type and amount of the lipids;
- f.
- Demonstration that, within a narrow range of double-stranded or single-stranded oligonucleotide length, there is no effect on product quality arising from sequence differences of the oligonucleotides (product-specific stability data should be provided to demonstrate that the sequence changes, modifications to the sugar backbone, phosphorothioate incorporation, or nucleobase modifications of the single-stranded or double-stranded oligonucleotide will not impact product quality);
- g.
- Manufacturing process parameters, in-process controls, and equipment critical to the formation of the lipid nanoparticles
8.2. mAbs
- Approaches for cell substrate and expression construct engineering that can be used with multiple products with the same upstream manufacturing process developed for the specific cell substrate and expression construct backbone refer to strategies in biotechnology and pharmaceutical manufacturing where a consistent and standardized upstream process is applied to produce various biological products using the same foundational cell substrate and expression construct [32].
- Cell substrate engineering involves selecting and genetically modifying a host cell line (cell substrate) to produce a desired biological product. Common host cell lines include Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) cells, and microbial cells like Escherichia coli [33]. For instance, CHO cells are frequently engineered to produce monoclonal antibodies (mAbs). By optimizing these cells to express high levels of recombinant proteins, manufacturers can use the same engineered CHO cell line to produce different mAbs, leveraging the same upstream process conditions, like media composition, growth conditions, and bioreactor settings [34]
- Expression construct engineering refers to the design and insertion of genetic constructs into the host cells to drive the expression of the target protein. The construct includes the gene of interest and regulatory elements such as promoters, enhancers, and terminators. For example, an expression construct might include a strong promoter like the cytomegalovirus (CMV) promoter to ensure high levels of protein expression in HEK293 cells. Once an effective expression construct backbone is developed and optimized, it can be reused with different genes of interest, simplifying the production of various recombinant proteins using the same upstream processes [35].
- Monoclonal Antibody Production:
- a.
- Cell substrate: CHO cells are engineered to produce various therapeutic monoclonal antibodies. The same upstream process, including cell culture media and bioreactor conditions, is applied across different mAbs, ensuring consistent product quality and streamlined regulatory approval [36].
- b.
- Expression construct: A well-characterized expression vector with a CMV promoter and other regulatory elements can be adapted to express different antibody genes, reducing development time for new products [37].
- Vaccine Manufacturing:
- a.
- Cell substrate: Vero cells (derived from African green monkey kidney cells) are used to produce viral vaccines like those for poliovirus and influenza. The upstream process, such as cell culture techniques and infection protocols, remains consistent across different vaccines produced in Vero cells [38].
- b.
- Expression construct: For recombinant protein vaccines, a plasmid vector with a strong viral promoter can be engineered to express different antigens, facilitating the rapid development of vaccines against emerging pathogens [39].
- Gene Therapy Products:
- a.
- Cell substrate: HEK293 cells are commonly used to produce viral vectors for gene therapy. The upstream process involving cell culture and transfection methods is standardized, enabling the production of various gene therapy vectors with the same manufacturing platform [39].
- b.
- Expression construct: The viral vector backbone, such as adeno-associated virus (AAV), is engineered with different therapeutic genes while keeping the regulatory elements constant, allowing for the efficient production of different gene therapy products using the same upstream process [40].
8.3. siRNA
- a.
- Identification of the targeting moiety, including its synthesis, incorporation into the final drug substance, and quality control;
- b.
- Modification of the synthetic siRNA sequence has no biological effect on the product quality or safety arising from the differences such that some pharmacology/toxicology and CMC data are potentially appropriate to be leveraged;
- c.
- The safety of the targeting moiety is not altered when used with multiple different siRNA moieties such that some pharmacology/toxicology data are potentially appropriate to be leveraged;
- d.
- Use of a unique method of manufacturing, purification approach, or purification strategies that simplify downstream characterization of the drug product and that can be used for multiple products with little modification;
- e.
- Recent advancements in delivery methods, such as the use of lipid nanoparticles, have further enhanced siRNA’s stability and cellular uptake, making it a more viable option for clinical applications [52].
9. Exclusions
- Approaches to viral clearance for certain unit operations;
- Manufacturing unit operations sensitive to inputs (e.g., the general use of roller compaction that might be sensitive to material properties).
- Technologies that rely on established manufacturing unit operations (e.g., blending, compressing, or film coating) (this prior knowledge can already be leveraged in formulation and manufacturing process development; demonstrated prior knowledge can also be used in applications to demonstrate unit operation robustness).
- Established formulation technologies traditionally used for immediate-release and extended-release solid oral dosage forms (e.g., matrices and osmotic pumps), established formulation technologies for oral and parenteral dosage forms, and other established drug delivery systems;
- Near-infrared technologies for monitoring in-process material attributes;
- Analytical methods that leverage prior knowledge as described in the draft ICH guidance for industry Q14 Analytical Procedure Development (August 2022);
- Device delivery technologies (e.g., syringes and autoinjectors). (For the purposes of this guidance, generally, such device delivery technologies are not essential to the structure (e.g., chemical or molecular formula) or function (e.g., the molecular mechanism of action or the drug or biological product’s characteristics or chemical or biological interaction with the body) of the drug or biological product. In addition, such device delivery technologies are generally not expected to facilitate the manufacture or development of a drug because, generally, drug manufacture is complete before the drug interacts with the delivery device. Also, the devices are not expected to bring significant efficiencies to the review process because of the existing leveraging options for delivery devices already incorporated in the review process).
10. Conclusions
10.1. Biosimilars
10.2. Advanced Manufacturing
10.3. Advanced Manufacturing Program
Funding
Conflicts of Interest
References
- Vass, P.; Akdag, D.S.; Broholm, G.E.; Kjaer, J.; Humphreys, A.J.; Ehmann, F. Enabling technologies driving drug research and development. Front. Med. 2023, 10, 1122405. [Google Scholar] [CrossRef] [PubMed]
- Ayon, N.J. High-Throughput Screening of Natural Product and Synthetic Molecule Libraries for Antibacterial Drug Discovery. Metabolites 2023, 13, 625. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.; Hawkins, B.A.; Du, J.J.; Groundwater, P.W.; Hibbs, D.E.; Lai, F. A Guide to In Silico Drug Design. Pharmaceutics 2022, 15, 49. [Google Scholar] [CrossRef] [PubMed]
- Vora, L.K.; Gholap, A.D.; Jetha, K.; Thakur, R.R.S.; Solanki, H.K.; Chavda, V.P. Artificial Intelligence in Pharmaceutical Technology and Drug Delivery Design. Pharmaceutics 2023, 15, 1916. [Google Scholar] [CrossRef] [PubMed]
- FDA. Real-World Evidence. 2023. Available online: https://www.fda.gov/science-research/science-and-research-special-topics/real-world-evidence (accessed on 12 May 2024).
- Bakker, E.; Starokozhko, V.; Kraaijvanger, J.W.M.; Heerspink, H.J.L.; Mol, P.G.M. Precision medicine in regulatory decision making: Biomarkers used for patient selection in European Public Assessment Reports from 2018 to 2020. Clin. Transl. Sci. 2023, 16, 2394–2412. [Google Scholar] [CrossRef] [PubMed]
- Zhang, G.; Tang, T.; Chen, Y.; Huang, X.; Liang, T. mRNA vaccines in disease prevention and treatment. Signal Transduct. Target. Ther. 2023, 8, 365. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Yang, Y.; Qi, H.; Cui, W.; Zhang, L.; Fu, X.; He, X.; Liu, M.; Li, P.-F.; Yu, T. CRISPR/Cas9 therapeutics: Progress and prospects. Signal Transduct. Target. Ther. 2023, 8, 36. [Google Scholar] [CrossRef]
- Kaspute, G.; Arunagiri, B.D.; Alexander, R.; Ramanavicius, A.; Samukaite-Bubniene, U. Development of Essential Oil Delivery Systems by ‘Click Chemistry’ Methods: Possible Ways to Manage Duchenne Muscular Dystrophy. Materials 2023, 16, 6537. [Google Scholar] [CrossRef]
- Singh, R.; Chandley, P.; Rohatgi, S. Recent Advances in the Development of Monoclonal Antibodies and Next-Generation Antibodies. ImmunoHorizons 2023, 7, 886–897. [Google Scholar] [CrossRef]
- Uscanga-Palomeque, A.C.; Chávez-Escamilla, A.K.; Alvizo-Báez, C.A.; Saavedra-Alonso, S.; Terrazas-Armendáriz, L.D.; Tamez-Guerra, R.S.; Rodríguez-Padilla, C.; Alcocer-González, J.M. CAR-T Cell Therapy: From the Shop to Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 15688. [Google Scholar] [CrossRef]
- Giannuzzi, V.; Bertolani, A.; Torretta, S.; Reggiardo, G.; Toich, E.; Bonifazi, D.; Ceci, A. Innovative research methodologies in the EU regulatory framework: An analysis of EMA qualification procedures from a pediatric perspective. Front. Med. 2024, 11, 1369547. [Google Scholar] [CrossRef]
- FDA. Platform Technology Designation Program for Drug Development Guidance for Industry. 2024. Available online: https://www.fda.gov/media/178928/download (accessed on 12 May 2024).
- Register, F. Critical Path Initiative; Establishment of Docket. 2004. Available online: https://www.federalregister.gov/documents/2004/04/22/04-9147/critical-path-initiative-establishment-of-docket (accessed on 12 May 2024).
- Sherman, R.E.; Li, J.; Shapley, S.; Robb, M.; Woodcock, J. Expediting Drug Development—The FDA’s New “Breakthrough Therapy” Designation. New Engl. J. Med. 2013, 369, 1877–1880. [Google Scholar] [CrossRef] [PubMed]
- Register, F. Adaptive Designs for Clinical Trials of Drugs and Biologics; Guidance for Industry. 2019. Available online: https://www.federalregister.gov/documents/2019/12/02/2019-25986/adaptive-designs-for-clinical-trials-of-drugs-and-biologics-guidance-for-industry-availability (accessed on 12 May 2024).
- Register, F. Q13 Continuous Manufacturing of Drug Substances and Drug Products; International Council for Harmonisation; Guidance for Industry. 2023. Available online: https://www.federalregister.gov/documents/2023/03/01/2023-04212/q13-continuous-manufacturing-of-drug-substances-and-drug-products-international-council-for (accessed on 12 May 2024).
- FDA. Chemistry, Manufacturing, and Controls Changes to an Approved Application: Certain Biological Products; Guidance for Industry. 2021. Available online: https://www.federalregister.gov/documents/2021/06/24/2021-13392/chemistry-manufacturing-and-controls-changes-to-an-approved-application-certain-biological-products (accessed on 12 May 2024).
- Kramps, T. Introduction to RNA Vaccines Post COVID-19. In RNA Vaccines. Methods in Molecular Biology; Humana: New York, NY, USA, 2024; Volume 2786, pp. 1–22. [Google Scholar]
- Wu, L.; Li, X.; Qian, X.; Wang, S.; Liu, J.; Yan, J. Lipid Nanoparticle (LNP) Delivery Carrier-Assisted Targeted Controlled Release mRNA Vaccines in Tumor Immunity. Vaccines 2024, 12, 186. [Google Scholar] [CrossRef]
- Masarwy, R.; Stotsky-Oterin, L.; Elisha, A.; Hazan-Halevy, I.; Peer, D. Delivery of nucleic acid based genome editing platforms via lipid nanoparticles: Clinical applications. Adv. Drug Deliv. Rev. 2024, 211, 115359. [Google Scholar] [CrossRef] [PubMed]
- Jogdeo, C.M.; Siddhanta, K.; Das, A.; Ding, L.; Panja, S.; Kumari, N.; Oupický, D. Beyond Lipids: Exploring Advances in Polymeric Gene Delivery in the Lipid Nanoparticles Era. Adv. Mater. 2024, e2404608. [Google Scholar] [CrossRef]
- US Pharmacopeia (USP). Uncovering RNA Payload, Empty Particles, and Physicochemical Heterogeneity of Lipid Nanoparticles via A Single Nanoparticle Analyzer. 2024. Available online: https://www.usp.org/sites/default/files/usp/document/events-and-training/02%20Uncovering%20RNA%20Payload%2C%20Empty%20Particles%2C%20and%20Physicochemical%20-%20Sixuan%20Li.pdf (accessed on 12 May 2024).
- Schober, G.B.; Story, S.; Arya, D.P. A careful look at lipid nanoparticle characterization: Analysis of benchmark formulations for encapsulation of RNA cargo size gradient. Sci. Rep. 2024, 14, 2403. [Google Scholar] [CrossRef]
- Finston, S.K.; Davey, N.S.; Davé, E.; Ravichandran, V.; Davey, S.R.; Davé, R.S. Mayo, Myriad, America Invents Act and BPCIA: How has the United States biopharmaceutical market been affected? Pharm. Pat. Anal. 2016, 5, 159–167. [Google Scholar] [CrossRef]
- World Trade Organization (WTO). WTO Members Agree to Extend TRIPS Transition Period for LDCs Until 1 July 2034. 2021. Available online: https://www.wto.org/english/news_e/news21_e/trip_30jun21_e.htm (accessed on 12 May 2024).
- UN Trade and Development (UNCTAD). United Nations List of Least Developed Countries. 2024. Available online: https://unctad.org/topic/least-developed-countries/list (accessed on 12 May 2024).
- US Pharmacopeia (USP). USP mRNA Vaccine Chapter. 2023. Available online: https://www.uspnf.com/sites/default/files/usp_pdf/EN/USPNF/usp-nf-notices/gc-xxx-analytical-procedures-mRNA-vaccines.pdf (accessed on 12 May 2024).
- Hashiba, K.; Taguchi, M.; Sakamoto, S.; Otsu, A.; Maeda, Y.; Ebe, H.; Okazaki, A.; Harashima, H.; Sato, Y. Overcoming thermostability challenges in mRNA–lipid nanoparticle systems with piperidine-based ionizable lipids. Commun. Biol. 2024, 7, 556. [Google Scholar] [CrossRef] [PubMed]
- Kulkarni, J.A.; Cullis, P.R.; van der Meel, R. Lipid Nanoparticles Enabling Gene Therapies: From Concepts to Clinical Utility. Nucleic Acid Ther. 2018, 28, 146–157. [Google Scholar] [CrossRef]
- Kazemian, P.; Yu, S.-Y.; Thomson, S.B.; Birkenshaw, A.; Leavitt, B.R.; Ross, C.J.D. Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Components. Mol. Pharm. 2022, 19, 1669–1686. [Google Scholar] [CrossRef]
- Wurm, F.M. Production of recombinant protein therapeutics in cultivated mammalian cells. Nat. Biotechnol. 2004, 22, 1393–1398. [Google Scholar] [CrossRef] [PubMed]
- Balbás, P. Understanding the art of producing protein and nonprotein molecules in Escherichia coli. Mol. Biotechnol. 2001, 19, 251–268. [Google Scholar] [CrossRef] [PubMed]
- Jayapal, K.; Wlaschin, K.F.; Hu, W.S.; Yap, M.G.S. Recombinant Protein Therapeutics from CHO Cells—20 Years and Counting. Chem. Eng. Prog. 2007, 103, 40–47. [Google Scholar]
- Thomas, P.; Smart, T.G. HEK293 cell line: A vehicle for the expression of recombinant proteins. J. Pharmacol. Toxicol. Methods 2005, 51, 187–200. [Google Scholar] [CrossRef] [PubMed]
- Butler, M.; Spearman, M. The choice of mammalian cell host and possibilities for glycosylation engineering. Curr. Opin. Biotechnol. 2014, 30, 107–112. [Google Scholar] [CrossRef] [PubMed]
- Walsh, G. Biopharmaceutical benchmarks 2014. Nat. Biotechnol. 2014, 32, 992–1000. [Google Scholar] [CrossRef] [PubMed]
- Barrett, P.N.; Mundt, W.; Kistner, O.; Howard, M.K. Vero cell platform in vaccine production: Moving towards cell culture-based viral vaccines. Expert Rev. Vaccines 2009, 8, 607–618. [Google Scholar] [CrossRef] [PubMed]
- Thomas, C.E.; Ehrhardt, A.; Kay, M.A. Progress and problems with the use of viral vectors for gene therapy. Nat. Rev. Genet. 2003, 4, 346–358. [Google Scholar] [CrossRef]
- Kotin, R.M. Large-scale recombinant adeno-associated virus production. Hum. Mol. Genet. 2011, 20, R2–R6. [Google Scholar] [CrossRef]
- Elbashir, S.M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber, K.; Tuschl, T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001, 411, 494–498. [Google Scholar] [CrossRef]
- Zamore, P.D.; Tuschl, T.; Sharp, P.A.; Bartel, D.P. RNAi: Double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell 2000, 101, 25–33. [Google Scholar] [CrossRef] [PubMed]
- Perry, N.B.; Swingler, M.M.; Calkins, S.D.; Bell, M.A. Neurophysiological correlates of attention behavior in early infancy: Implications for emotion regulation during early childhood. J. Exp. Child Psychol. 2016, 142, 245–261. [Google Scholar] [CrossRef]
- Lu, Z.; Xie, J.; Yan, R.; Yu, Z.; Sun, Z.; Yu, F.; Zhang, Y. A pilot study of pancreatic islet amyloid PET imaging with [18F]FDDNP. Nucl. Med. Commun. 2018, 39, 659–664. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.U.; Hayles, J.; Kim, D.; Wood, V.; Park, H.O.; Won, M.; Hoe, K.L. Analysis of a genome-wide set of gene deletions in the fission yeast Schizosaccharomyces pombe. Nat. Biotechnol. 2010, 28, 617–623. [Google Scholar] [CrossRef] [PubMed]
- Sanmukhani, J.J.; Pawar, P.; Mittal, R. Ramosetron hydrochloride for the prevention of cancer chemotherapy induced nausea and vomiting: The Indian experience. South Asian J. Cancer 2014, 3, 132–137. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Wang, Z.; Zhang, L.; Qiao, L.; Shen, D. Remodeling Pearson’s Correlation for Functional Brain Network Estimation and Autism Spectrum Disorder Identification. Front. Neuroinform. 2017, 11, 55. [Google Scholar] [CrossRef] [PubMed]
- Dzimianski, J.V.; Scholte, F.E.M.; Williams, I.L.; Langley, C.; Freitas, B.T.; Spengler, J.R.; Bergeron, É.; Pegan, S.D. Determining the molecular drivers of species-specific interferon-stimulated gene product 15 interactions with nairovirus ovarian tumor domain proteases. PLoS ONE 2019, 14, e0226415. [Google Scholar] [CrossRef] [PubMed]
- Behlke, M.A. Progress towards in vivo use of siRNAs. Mol. Ther. 2006, 13, 644–670. [Google Scholar] [CrossRef] [PubMed]
- Schwarz, D.S.; Hutvágner, G.; Du, T.; Xu, Z.; Aronin, N.; Zamore, P.D. Asymmetry in the assembly of the RNAi enzyme complex. Cell 2003, 115, 199–208. [Google Scholar] [CrossRef]
- Brummelkamp, T.R.; Bernards, R.; Agami, R. A system for stable expression of short interfering RNAs in mammalian cells. Science 2002, 296, 550–553. [Google Scholar] [CrossRef]
- Whitehead, K.A.; Langer, R.; Anderson, D.G. Knocking down barriers: Advances in siRNA delivery. Nat. Rev. Drug Discov. 2009, 8, 129–138. [Google Scholar] [CrossRef] [PubMed]
- FDA. Advanced Manufacturing. 2024. Available online: https://www.fda.gov/emergency-preparedness-and-response/mcm-issues/advanced-manufacturing (accessed on 12 May 2024).
Type | Moderna | BioNtech-Pfizer | CureVac |
---|---|---|---|
Ionizable lipids | heptadecan-9-yl 8-((2-hydroxyethyl)(6-oxo-6-(undecyloxy)hexyl)amino)octanoate (SM-102) | (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315) | Probably, (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis(2-hexyldecanoate) (ALC-0315) |
Phospholipid | 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) | 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) | 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) |
Cholesterol | Yes | Yes | Yes |
PEG lipid | 1,2-dimyristoylrac-glycero-3-methoxypolyethylene glycol-2000 (PEG2000 DMG) | 2[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide | Probably, 2[(polyethylene glycol)-2000]-N,N-ditetradecylacetamide |
Total Lipids, mg/dose | 1.93 | 0.77 | 0.31 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Niazi, S.K. The United States Food and Drug Administration’s Platform Technology Designation to Expedite the Development of Drugs. Pharmaceutics 2024, 16, 918. https://doi.org/10.3390/pharmaceutics16070918
Niazi SK. The United States Food and Drug Administration’s Platform Technology Designation to Expedite the Development of Drugs. Pharmaceutics. 2024; 16(7):918. https://doi.org/10.3390/pharmaceutics16070918
Chicago/Turabian StyleNiazi, Sarfaraz K. 2024. "The United States Food and Drug Administration’s Platform Technology Designation to Expedite the Development of Drugs" Pharmaceutics 16, no. 7: 918. https://doi.org/10.3390/pharmaceutics16070918
APA StyleNiazi, S. K. (2024). The United States Food and Drug Administration’s Platform Technology Designation to Expedite the Development of Drugs. Pharmaceutics, 16(7), 918. https://doi.org/10.3390/pharmaceutics16070918