Noha F Ghazi, Jonathan C Burley, Ian L Dryden, Clive J Roberts
{"title":"High-Throughput Microarray Approaches for Predicting the Stability of Drug-Polymer Solid Dispersions.","authors":"Noha F Ghazi, Jonathan C Burley, Ian L Dryden, Clive J Roberts","doi":"10.1021/acs.molpharmaceut.4c00955","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c00955","url":null,"abstract":"<p><p>Amorphous solid dispersions (ASDs) offer a well-recognized strategy to improve the effective solubility and, hence, bioavailability of poorly soluble drugs. In this study, we developed an extensive library of a significant number of solid dispersion formulations using a library of chemically diverse drugs combined with a water-soluble polymer (polyvinylpyrrolidone vinyl acetate, PVPVA) at different loadings. These formulations were printed as microarrays of solid dispersion formulations, utilizing minimal material amounts (nanograms). They were subjected to a six-month stability study under accelerated conditions (40 °C and 75% relative humidity). Physical stability outcomes varied significantly among the different drug-polymer combinations, with stability ranging from immediate drug crystallization to several days of stability. The comprehensive data set obtained from this high-throughput screening was used to construct multiple linear regression models to correlate the stability of ASDs with the physicochemical properties of the used Active Pharmaceutical Ingredients (APIs). Our findings reveal that increased stability of ASDs is associated with a lower number of hydrogen bond acceptors alongside a higher overall count of heteroatoms and oxygen atoms in the drug molecules. This suggests that, while heteroatoms and oxygen are abundant, their role as hydrogen bond acceptors is limited due to their specific chemical environments, contributing to overall stability. Additionally, drugs with lower melting points formed more stable ASDs within the polymer matrix. This study, hence, highlights the importance of minimizing repulsive drug-polymer interactions to yield a physically stable ASD. The developed models, validated through Leave-One-Out Cross-Validation, demonstrated good predictability of stability trends. Hence, the high-throughput 2D inkjet printing technique that was used to manufacture the microarrays proved valuable for assessing drug-polymer crystallization onset risks and predicting stability outcomes. In conclusion, this study demonstrates a novel approach to solid dispersion formulation physical stability screening, enhancing efficiency, minimizing material requirements, and expanding the range of samples evaluated. Our findings provide insights into the critical physicochemical properties influencing ASD stability, offering a significant advancement in developing stable ASDs.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advances in Dendritic Systems and Dendronized Nanoparticles: Paradigm Shifts in Cancer Targeted Therapy and Diagnostics.","authors":"Pawan Kedar, Apeksha Saraf, Rahul Maheshwari, Mayank Sharma","doi":"10.1021/acs.molpharmaceut.4c00856","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c00856","url":null,"abstract":"<p><p>Cancer has emerged as a global health crisis, claiming millions of lives annually. Dendrimers and dendronized nanoparticles, a novel class of nanoscale molecules with highly branched three-dimensional macromolecular structures, have gained significant attention in cancer treatment and diagnosis due to their unique properties. These dendritic macromolecules offer a precisely controlled branching architecture, enabling functionalization with specific targeting molecules to enhance the selective delivery of therapeutic agents to tumor cells while minimizing systemic toxicity. Through surface modifications and the incorporation of various components, dendrimers demonstrate remarkable adaptability as nanocarriers for biomedical imaging and theranostic applications. Surface functionalization strategies, including PEGylation and ligand attachment (e.g., folic acid, RGD peptide, lactobionic acid), further enhance biocompatibility and facilitate targeted tumor cell imaging. Leveraging their improved biocompatibility and target specificity, dendritic nanosystems offer heightened sensitivity and precision in cancer diagnostics. Notably, the encapsulation of metal nanoparticles within dendrimers, such as gold nanoparticles, has shown promise in enhancing tumor imaging capabilities. Ongoing advancements in nanotechnology are poised to increase the sophistication and complexity of dendrimer-based systems, highlighting their potential as nanocarriers in drug delivery platforms, with a growing number of clinical trials on the horizon. This review provides a comprehensive overview of the potential and future prospects of dendrimers and dendrimer-based nanocarriers in targeted cancer therapy and diagnosis, exploring their ability to enhance biocompatibility, reduce toxicity, and improve therapeutic outcomes across various malignancies.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142870576","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abbigael Harthorn, Tse-Han Kuo, Sarah W Torres, Roy R Lobb, Benjamin J Hackel
{"title":"Expression-Dependent Tumor Pretargeting via Engineered Avidity.","authors":"Abbigael Harthorn, Tse-Han Kuo, Sarah W Torres, Roy R Lobb, Benjamin J Hackel","doi":"10.1021/acs.molpharmaceut.4c01177","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c01177","url":null,"abstract":"<p><p>Selective delivery of therapeutic modalities to tumor cells via binding of tumor-selective cell-surface biomarkers has empowered substantial advances in cancer treatment. Yet, tumor cells generally lack a truly specific biomarker that is present in high density on tumor tissue while being completely absent from healthy tissue. Rather, low but nonzero expression in healthy tissues results in on-target, off-tumor activity with detrimental side effects that constrain the therapeutic window or prevent use altogether. Advanced technologies to enhance the selectivity for tumor targeting are sorely needed. We have engineered a binding platform that is quantitatively dependent upon expression levels, via avidity-driven specificity, rather than binarily reliant on the presence or absence of a biomarker. We systematically varied monomeric binding affinity by engineering affibodies to target carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and folate receptor 1 (FolR1). Two identical affibody ligands were tethered, with varying polypeptide linker lengths, to a nanobody that binds Alfa peptide to create a bispecific, trivalent protein for use in pretargeted radioligand therapy. Expression-dependent targeting was achieved in both systems: with 110 nM monomeric affinity to CEACAM5 with a two-amino-acid linker or with 250 nM monomeric affinity for FolR1 and a 10 amino acid linker. The latter bispecific, trivalent achieved over 25-fold differentiation between FolR1<sup>high</sup> and FolR1<sup>low</sup> cells in a mixed culture. Similar selectivity was achieved in a size-efficient bivalent molecule lacking a central nanobody. Moreover, the avid bivalent affibody molecule exhibited minimal inhibition by soluble antigen, whereas high-affinity bivalent antibody was inhibited by 97 ± 2%, which is indicative of serum inhibition of shed antigen. This work advances design principles for achieving expression-dependent tumor targeting via low-affinity, high-avidity ligands.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142862534","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Impact of HPMCAS Grade on the Release of Weakly Basic Drugs from Amorphous Solid Dispersions.","authors":"Pradnya Bapat, Lynne S Taylor","doi":"10.1021/acs.molpharmaceut.4c00986","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c00986","url":null,"abstract":"<p><p>Oppositely charged species can form electrostatic interactions in aqueous solution, and these may lead to reduced solubility of the interacting components. Herein, insoluble complex formation between the lipophilic weakly basic drugs, cinnarizine or loratadine, and the enteric polymer, hydroxypropyl methylcellulose acetate succinate (HPMCAS), was studied and used to better understand drug and polymer release from their corresponding amorphous solid dispersions (ASDs). Surface area normalized release experiments were performed at various pH conditions for three different grades of HPMCAS, LF, MF and HF, as well as their ASDs. Both polymer and drug release rates were measured for the ASDs. Complexation tendency was evaluated by measuring the extent of polymer loss from the aqueous phase in the presence of the drug. Results showed that release from ASDs with HPMCAS-LF was less impacted by the presence of a cationic form of the drug than ASDs prepared with the HF grade. Furthermore, an increase in pH, leading to a reduction in the extent of ionized drug also led to an improvement in release rate. These observations provide a baseline to understand the role of drug-polymer electrostatic interactions on release from ASDs formulated with HPMCAS. Future studies should focus on adding complexity to media conditions by employing simulated intestinal fluids with solubilizing components.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142862535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jack D Murray, Harriet Bennett-Lenane, Patrick J O'Dwyer, Brendan T Griffin
{"title":"Establishing a Pharmacoinformatics Repository of Approved Medicines: A Database to Support Drug Product Development.","authors":"Jack D Murray, Harriet Bennett-Lenane, Patrick J O'Dwyer, Brendan T Griffin","doi":"10.1021/acs.molpharmaceut.4c00991","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c00991","url":null,"abstract":"<p><p>Advanced predictive modeling approaches have harnessed data to fuel important innovations at all stages of drug development. However, the need for a machine-readable drug product library which consolidates many aspects of formulation design and performance remains largely unmet. This study presents a scripted, reproducible approach to database curation and explores its potential to streamline oral medicine development. The Product Information files for all centrally authorized drug products containing a small molecule active ingredient were retrieved programmatically from the European Medicines Agency Web site. Text processing isolated relevant information, including the maximum clinical dose, dosage form, route of administration, excipients, and pharmacokinetic performance. Chemical and bioactivity data were integrated through automated linking to external curated databases. The capability of this database to inform oral medicine development was assessed in the context of drug-likeness evaluation, excipient selection, and prediction of oral fraction absorbed. Existing filters of drug-likeness, such as the Rule of Five, were found to poorly capture the chemical space of marketed oral drug products. Association rule learning identified frequent patterns in tablet formulation compositions that can be used to establish excipient combinations that have seen clinical success. Binary prediction models of oral fraction absorbed constructed exclusively from regulatory data achieved acceptable performance (balanced accuracy<sub>test</sub> = 0.725), demonstrating its modelability and potential for use during early stage molecule prioritization tasks. This study illustrates the impact of highly linked drug product data in accelerating clinical translation and underlines the ongoing need for accuracy and completeness of data reported in the regulatory datasphere.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142869275","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Self-Nanoemulsifying Formulation Improves Oral Bioavailability and Insulin Sensitizing Potency of Formononetin-Vitamin E Conjugate in Type 2 Diabetic Mice.","authors":"Rakesh Kumar Dhritlahre, Navneet Thakur, Abhishek Goel, Vikram Patial, Yogendra Padwad, Ankit Saneja","doi":"10.1021/acs.molpharmaceut.4c00886","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c00886","url":null,"abstract":"<p><p>The escalating incidence of obesity, diabetes, and insulin resistance has become a significant global health concern. In this study, we have developed a self-nanoemulsifying delivery system (SNEDS) of formononetin-vitamin E conjugate (VESylated-FMN) for improving its oral bioavailability and improving insulin sensitivity and glycemic control. The developed SNEDS were characterized using dynamic light scattering and transmission electron microscopy. Thereafter, the loading capacity, <i>in vitro</i> release, thermodynamic, and gastrointestinal stability of the developed formulation were evaluated. The safety and oral bioavailability of VESylated-FMN-SNEDS were assessed in Sprague-Dawley rats, whereas insulin-sensitizing potency was assessed in high-fat diet-induced type 2 diabetic mice. The VESylated-FMN-SNEDS quickly emulsified on dilution (droplet size ∼79.17 nm) and showed remarkable thermodynamic and gastrointestinal stability. The developed formulation demonstrated enhanced oral bioavailability (∼1.3-fold higher AUC<sub>0-t</sub>) of VESylated-FMN without liver and kidney injury. Consequently, VESylated-FMN-SNEDS significantly improves insulin sensitivity and glycemic control in HFD-fed mice compared to VESylated-FMN by upregulating the transcript level of insulin-sensitizing genes. Therefore, the SNEDS formulation could be an effective strategy to augment the oral bioavailability and insulin-sensitizing potency of VESylated-FMN.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142851641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jing Ling, Ryan Schroder, W Peter Wuelfing, John Higgins, Filippos Kesisoglou, Allen C Templeton, Yongchao Su
{"title":"Molecular Investigation of SNAC as an Oral Peptide Permeation Enhancer in Lipid Membranes via Solid-State NMR.","authors":"Jing Ling, Ryan Schroder, W Peter Wuelfing, John Higgins, Filippos Kesisoglou, Allen C Templeton, Yongchao Su","doi":"10.1021/acs.molpharmaceut.4c01061","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c01061","url":null,"abstract":"<p><p>Oral peptide therapeutics are increasingly favored in the pharmaceutical industry for their ease of use and better patient adherence. However, they face challenges with poor oral bioavailability due to their high molecular weight and surface polarity. Permeation enhancers (PEs) like salcaprozate sodium (SNAC) have shown promise in clinical trials, achieving about 1% bioavailability. One proposed mechanism for enhancing permeation is membrane perturbation or fluidization, though direct experimental proof and quantitative analysis of these effects are still needed. This study employs solid-state NMR (ssNMR) to investigate how SNAC interacts with hydrated DMPC liposomes, measuring enhancements in membrane fluidity across interfacial and transmembrane regions. The methodology involves analyzing phosphate lipid headgroups and acyl chains using static <sup>31</sup>P chemical shift anisotropy and <sup>2</sup>H quadrupolar coupling measurements alongside <sup>1</sup>H and <sup>13</sup>C magic angle spinning NMR for motional averaging of <sup>1</sup>H-<sup>1</sup>H and <sup>1</sup>H-<sup>13</sup>C dipolar couplings. Our findings indicate an overall increase in the uniaxial motion of phospholipids with SNAC in a PE concentration-dependent manner. It boosts lipid headgroup dynamics and enhancement plateaus at 25% between 24 and 72 mM concentrations. SNAC effectively enhances the fluidity of the hydrophobic center by 43% at 72 mM PE concentration, more significantly than the interfacial region. It is worth noting that the extent of liposome dissolution and conversion to micelles increases as SNAC concentration rises. Including a model peptide drug, octreotide, introduces a competitive equilibrium in this complex PE-lipid-peptide system, further influencing membrane dynamics for peptide permeation. Interestingly, the membrane enhancement does not show the expected plateau, and a less significant lipid mobility increase is observed in the presence of octreotide, suggesting a less substantial impact compared to peptide-free systems, which is likely due to peptide-PE interactions that consume monomeric SNAC, reducing its interaction with the lipid membrane. This study provides the first quantitative and site-specific ssNMR measurements of membrane mobility influenced by one representative PE as a snapshot of PE lipid interaction in a liposome model, demonstrating how peptide drugs modulate competitive equilibria and PE-induced lipid dynamics.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142845314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Enhancing Buccal Drug Delivery: The Impact of Glycerol in Slot-Die-Coated Pectin Films.","authors":"Eleftheria Pantazoglou, Matteo Tollemeto, Nazanin Zanjanizadeh Ezazi, Tien-Jen Chang, Leticia Hosta Rigau, Jette Jacobsen, Line Hagner Nielsen","doi":"10.1021/acs.molpharmaceut.4c01051","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c01051","url":null,"abstract":"<p><p>Buccal delivery offers a promising alternative to e.g., oral or parenteral drug administrations by leveraging the mucosal membranes of the mouth to enhance drug absorption and enhance patient compliance. Buccal films offer a promising approach for enhancing drug delivery by utilizing the mucoadhesive properties of the biopolymer pectin and glycerol's plasticizing effects. Designed to provide fast drug release, these films address the challenges of patient compliance, particularly among the elderly, children, and individuals with dysphagia. This study characterized the physicochemical properties of slot-die-coated films with pectin containing varying amounts of glycerol, including swelling behavior, disintegration rate, mechanical properties, mucoadhesion, and drug release profiles, using paracetamol as a model drug. Different methods such as quartz crystal microbalance with dissipation and open-source force analyzer were employed for the characterization. The results demonstrated that a high glycerol content in the films led to slower drug release with 95% paracetamol released for film without glycerol (GLY0) compared to only 74% released for film with 20% w/v glycerol (GLY20) after 60 min Additionally, higher glycerol levels resulted in enhanced mucoadhesive properties. Films containing 20% glycerol also showed superior permeability of paracetamol through ex vivo porcine buccal mucosa, with double the amount of paracetamol permeating in the first 120 min from GLY20 films compared to GLY0 films. These findings suggest that the pectin-glycerol buccal films, fabricated with slot-die coating as a novel technique, are user-friendly, exhibit interaction with the mucosa, and can be adjusted for specific disintegration and drug release rates, presenting a promising option for efficient, targeted drug delivery.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142845313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Christer Tannergren, Sumit Arora, Andrew Babiskin, Luiza Borges, Parnali Chatterjee, Yi-Hsien Cheng, André Dallmann, Anitha Govada, Tycho Heimbach, Martin Hingle, Sivacharan Kollipara, Evangelos Kotzagiorgis, Anders Lindahl, Claire Mackie, Maria Malamatari, Amitava Mitra, Rebecca Moody, Xavier Pepin, James Polli, Kimberly Raines, Gregory Rullo, Maitri Sanghavi, Rajesh Savkur, Rajendra Singh, Erik Sjögren, Sandra Suarez-Sharp, Sherin Thomas, Shereeni Veerasingham, Kevin Wei, Fang Wu, Yunming Xu, Miyoung Yoon, Bhagwant Rege
{"title":"Current State and New Horizons in Applications of Physiologically Based Biopharmaceutics Modeling (PBBM): A Workshop Report.","authors":"Christer Tannergren, Sumit Arora, Andrew Babiskin, Luiza Borges, Parnali Chatterjee, Yi-Hsien Cheng, André Dallmann, Anitha Govada, Tycho Heimbach, Martin Hingle, Sivacharan Kollipara, Evangelos Kotzagiorgis, Anders Lindahl, Claire Mackie, Maria Malamatari, Amitava Mitra, Rebecca Moody, Xavier Pepin, James Polli, Kimberly Raines, Gregory Rullo, Maitri Sanghavi, Rajesh Savkur, Rajendra Singh, Erik Sjögren, Sandra Suarez-Sharp, Sherin Thomas, Shereeni Veerasingham, Kevin Wei, Fang Wu, Yunming Xu, Miyoung Yoon, Bhagwant Rege","doi":"10.1021/acs.molpharmaceut.4c01148","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c01148","url":null,"abstract":"<p><p>This report summarizes the proceedings for Day 3 of the workshop titled \"<i>Physiologically Based Biopharmaceutics Modeling (PBBM) Best Practices for Drug Product Quality: Regulatory and Industry Perspectives</i>\". This day focused on the current and future drug product quality applications of PBBM from the innovator and generic industries as well as the regulatory agencies perspectives. The presentations, which included several case studies, covered the applications of PBBM in generic drug product development, applications of virtual bioequivalence trials to support formulation bridging and the utility of absorption modeling in clinical pharmacology assessments. In addition, recent progress in the prediction of colon absorption and <i>in vivo</i> performance of extended-release drug products was shared. The morning session was concluded by representatives from FDA, ANVISA, MHRA, Health Canada, EMA, and PMDA giving their perspectives on the application of PBBM in regulatory submissions. The afternoon breakout sessions focused on four parallel topics: 1) PBBM in generic drug product development; 2) virtual bioequivalence trials applications; 3) safe space and extrapolation; and 4) regional absorption and modified release PBBM applications. This allowed the participants to engage in in-depth discussions of best practices as well to identify key points of consideration to allow further progress on the applications of PBBM.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142833143","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Colleen P Olkowski, Falguni Basuli, Bruna Fernandes, Behnaz Ghaemi, Jianfeng Shi, Hongwei H Zhang, Joshua M Farber, Freddy E Escorcia, Peter L Choyke, Orit Jacobson
{"title":"Comparative Kidney Uptake of Nanobody-Based PET Tracers Labeled with Various Fluorine-18-Labeled Prosthetic Groups.","authors":"Colleen P Olkowski, Falguni Basuli, Bruna Fernandes, Behnaz Ghaemi, Jianfeng Shi, Hongwei H Zhang, Joshua M Farber, Freddy E Escorcia, Peter L Choyke, Orit Jacobson","doi":"10.1021/acs.molpharmaceut.4c01101","DOIUrl":"https://doi.org/10.1021/acs.molpharmaceut.4c01101","url":null,"abstract":"<p><p>Nanobodies, or single-domain antibody fragments, are promising candidates for molecular imaging due to their small size, rapid tissue penetration, and high target specificity. However, a significant challenge in their use is high renal uptake and retention, which can limit the therapeutic efficacy and complicate image interpretation. This study compares five different fluorine-18-labeled prosthetic groups for nanobodies, aiming to optimize pharmacokinetics and minimize kidney retention while maintaining tumor targeting. Using an epidermal growth factor receptor (EGFR) targeting nanobody as a model, two labeling approaches were evaluated; direct labeling of RESCA (with and without polyethylene glycol (PEG))-conjugated nanobody using Al[<sup>18</sup>F]F and indirect labeling using ([<sup>18</sup>F]F-fluoropyridine ([<sup>18</sup>F]F-FPy)-based prosthetic groups (site-specific and nonsite-specific). Labeled nanobodies were characterized in vitro for binding affinity and cell uptake with in vivo behavior assessed in EGFR + A431 tumor-bearing mice using PET imaging and biodistribution studies. Labeling with Al[<sup>18</sup>F]F showed high renal retention, which was partially mitigated by PEGylation. However, PEGylation also led to a decreased tumor uptake, particularly with longer PEG chains. Labeling using [<sup>18</sup>F]F-FPy prosthetic groups exhibited the most favorable pharmacokinetics, with rapid renal clearance and minimal kidney retention while maintaining high tumor uptake. These constructs showed excellent tumor-to-background contrast as early as 1 h postinjection. The study confirms that the selection of the prosthetic group significantly impacts the in vivo behavior of nanobodies, particularly regarding kidney accumulation. [<sup>18</sup>F]F-FPy-based prosthetic groups show the most promising results, with high tumor and minimal kidney uptake. Robust production of [<sup>18</sup>F]F-FPy on Sep-Pak is adaptable to clinical translation. Moreover, the potential substitution of <sup>18</sup>F with therapeutic radioisotopes such as <sup>131</sup>I or <sup>211</sup>At could expand the application of these nanobodies from diagnostics to targeted radionuclide therapy while maintaining a low kidney exposure. These findings have important implications for optimizing nanobody-based radiopharmaceuticals for molecular imaging and targeted radionuclide therapy.</p>","PeriodicalId":52,"journal":{"name":"Molecular Pharmaceutics","volume":" ","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142833141","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}