Pub Date : 2024-07-26DOI: 10.1021/acsptsci.4c0011910.1021/acsptsci.4c00119
Ana Semeano, Rian Garland, Alessandro Bonifazi, Kuo Hao Lee, John Famiglietti, Wenqi Zhang, Yoon Jae Jo, Francisco O. Battiti, Lei Shi, Amy Hauck Newman and Hideaki Yano*,
Bitopic ligands bind both orthosteric and allosteric or secondary binding sites within the same receptor, often resulting in an improvement of receptor selectivity, potency, and efficacy. In particular, for both agonists and antagonists of the dopamine D2 and D3 receptors (D2R and D3R), the primary therapeutic targets for several neurological and neuropsychiatric disorders, bitopic ligand design has proved advantageous in achieving better pharmacological profiles in vitro. Although the two pharmacophores within a bitopic ligand are typically considered the main drivers of conformational change for a receptor, the role of the linker that connects the two has not yet been systematically studied for its relevance in receptor activity profiles. Here, we present a comprehensive analysis of sumanirole and PF592,379-based indole-containing bitopic compounds in agonist activity at D2R and D3R, with a focus on linker chemical space and stereochemistry through testing six distinct chirally resolved linkers and a simple aliphatic linker. The structure activity relationships (SARs) of these linkers are examined extensively, beyond the conventional level, by characterizing the activation of all putative transducers over a 44 min time course. Our multiparametric analysis reveals previously unappreciated specific linker-dependent effects on primary pharmacophores, receptors, transducer activation kinetics, and bias, highlighting the utility of this comprehensive approach and the significance of the linker type in shaping transducer bias profiles.
{"title":"Linkers in Bitopic Agonists Shape Bias Profile among Transducers for the Dopamine D2 and D3 Receptors","authors":"Ana Semeano, Rian Garland, Alessandro Bonifazi, Kuo Hao Lee, John Famiglietti, Wenqi Zhang, Yoon Jae Jo, Francisco O. Battiti, Lei Shi, Amy Hauck Newman and Hideaki Yano*, ","doi":"10.1021/acsptsci.4c0011910.1021/acsptsci.4c00119","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00119https://doi.org/10.1021/acsptsci.4c00119","url":null,"abstract":"<p >Bitopic ligands bind both orthosteric and allosteric or secondary binding sites within the same receptor, often resulting in an improvement of receptor selectivity, potency, and efficacy. In particular, for both agonists and antagonists of the dopamine D2 and D3 receptors (D2R and D3R), the primary therapeutic targets for several neurological and neuropsychiatric disorders, bitopic ligand design has proved advantageous in achieving better pharmacological profiles <i>in vitro</i>. Although the two pharmacophores within a bitopic ligand are typically considered the main drivers of conformational change for a receptor, the role of the linker that connects the two has not yet been systematically studied for its relevance in receptor activity profiles. Here, we present a comprehensive analysis of sumanirole and PF592,379-based indole-containing bitopic compounds in agonist activity at D2R and D3R, with a focus on linker chemical space and stereochemistry through testing six distinct chirally resolved linkers and a simple aliphatic linker. The structure activity relationships (SARs) of these linkers are examined extensively, beyond the conventional level, by characterizing the activation of all putative transducers over a 44 min time course. Our multiparametric analysis reveals previously unappreciated specific linker-dependent effects on primary pharmacophores, receptors, transducer activation kinetics, and bias, highlighting the utility of this comprehensive approach and the significance of the linker type in shaping transducer bias profiles.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2333–2349 2333–2349"},"PeriodicalIF":4.9,"publicationDate":"2024-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acsptsci.4c00119","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141956945","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-25DOI: 10.1021/acsptsci.4c0027710.1021/acsptsci.4c00277
Yi-Hua Chiang, Erin C. Berthold, Michelle A. Kuntz, Siva Rama Raju Kanumuri, Alexandria S. Senetra, Sushobhan Mukhopadhyay, Aidan J. Hampson, Christopher R. McCurdy and Abhisheak Sharma*,
This study reports the steady-state pharmacokinetic parameters for mitragynine and characterizes its elimination in male and female rats. Four male and female rats were dosed q12h with 40 mg/kg, and orally administered mitragynine for 5 and 6 days, respectively. Using a validated ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method, the plasma concentrations of mitragynine, its metabolites (7-hydroxymitragynine, 9-hydroxycorynantheidine, and mitragynine acid), and a non-CYP oxidation product (3-dehydromitragynine) were determined at various time points. Sex differences in pharmacokinetics were observed, with females demonstrating significantly higher systemic exposure of mitragynine than males. The mitragynine area under the curve normalized by the dose interval (AUC/τ) was 6741.6 ± 869.5 h*ng/mL in female rats and 1808.9 ± 191.3 h*ng/mL in males (p < 0.05). Both sexes produced similar metabolite profiles; the major metabolites were mitragynine acid and 9-hydroxycorynantheidine. 7-Hydroxymitragynine was a minor metabolite. However, higher exposure (AUCs) and the maximum plasma concentrations (Cmax) of active metabolites, 7-hydroxymitragynine and 9-hydroxycorynantheidine, were observed in female rats and exhibited substantial sex differences. Renal clearance of mitragynine (CLr) was low (0.64 ± 0.3 mL/h in males and 0.98 ± 0.4 mL/h in females), and unchanged mitragynine accounted for <1% of the dose excreted in feces (both sexes). The clinical chemistry, complete blood count, and hematological test results reported no abnormal hematological findings after multiple dosing in either sex.
{"title":"Multiple-Dose Pharmacokinetics and Safety of Mitragynine, the Major Alkaloid of Kratom, in Rats","authors":"Yi-Hua Chiang, Erin C. Berthold, Michelle A. Kuntz, Siva Rama Raju Kanumuri, Alexandria S. Senetra, Sushobhan Mukhopadhyay, Aidan J. Hampson, Christopher R. McCurdy and Abhisheak Sharma*, ","doi":"10.1021/acsptsci.4c0027710.1021/acsptsci.4c00277","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00277https://doi.org/10.1021/acsptsci.4c00277","url":null,"abstract":"<p >This study reports the steady-state pharmacokinetic parameters for mitragynine and characterizes its elimination in male and female rats. Four male and female rats were dosed q12h with 40 mg/kg, and orally administered mitragynine for 5 and 6 days, respectively. Using a validated ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method, the plasma concentrations of mitragynine, its metabolites (7-hydroxymitragynine, 9-hydroxycorynantheidine, and mitragynine acid), and a non-CYP oxidation product (3-dehydromitragynine) were determined at various time points. Sex differences in pharmacokinetics were observed, with females demonstrating significantly higher systemic exposure of mitragynine than males. The mitragynine area under the curve normalized by the dose interval (AUC/τ) was 6741.6 ± 869.5 h*ng/mL in female rats and 1808.9 ± 191.3 h*ng/mL in males (<i>p</i> < 0.05). Both sexes produced similar metabolite profiles; the major metabolites were mitragynine acid and 9-hydroxycorynantheidine. 7-Hydroxymitragynine was a minor metabolite. However, higher exposure (AUCs) and the maximum plasma concentrations (<i>C</i><sub>max</sub>) of active metabolites, 7-hydroxymitragynine and 9-hydroxycorynantheidine, were observed in female rats and exhibited substantial sex differences. Renal clearance of mitragynine (CL<sub>r</sub>) was low (0.64 ± 0.3 mL/h in males and 0.98 ± 0.4 mL/h in females), and unchanged mitragynine accounted for <1% of the dose excreted in feces (both sexes). The clinical chemistry, complete blood count, and hematological test results reported no abnormal hematological findings after multiple dosing in either sex.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2452–2464 2452–2464"},"PeriodicalIF":4.9,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141956349","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Riboflavin transporter 3 (RFVT3) represents a potential cardioprotective biotarget in energetic metabolism reprogramming after myocardial infarction/reperfusion (MI/R). This study investigated the feasibility of noninvasive real-time quantification of RFVT3 expression after MI/R with an radiolabeled probe 18F-RFTA in a preclinical rat model of MI/R. The tracer 18F-RFTA was radio-synthesized manually and characterized on the subjects of radiolabeling yield, radiochemical purity, and stability in vivo. MI/R and sham-operated rat models were confirmed by cardiac magnetic resonance imaging (cMRI) and single-photon-emission computed tomography (SPECT) myocardial perfusion imaging (MPI) with technetium-99m sestamibi (99mTc-MIBI). Positron emission tomography (PET) imaging of MI/R and sham-operated rat models were conducted with 18F-RFTA. Ex vivo autoradiography and RFVT3 immunohistochemical (IHC) staining were conducted to verify the RFVT3 expression in infarcted and normal myocardium. 18F-RFTA injection was prepared with high radiochemical purity (>95%) and kept stable in vitro and in vivo. 18F-RFTA PET revealed significant uptake in the infarcted myocardium at 8 h after reperfusion, as confirmed by lower 99mTc-MIBI perfusion and decreased intensity of cMRI. Conversely, there were only the tiniest uptakes in the normal myocardium and blocked infarcted myocardium, which was further corroborated by ex vivo autoradiography. The RFVT3 expression was further confirmed by IHC staining in the infarcted and normal myocardium. We first demonstrate the feasibility of imaging RFVT3 in infarcted myocardium. 18F-RFTA is an encouraging PET probe for imaging cardioprotective biotarget RFVT3 in mitochondrial energetic metabolism reprogramming after myocardial infarction. Noninvasive imaging of cardioprotective biotarget RFVT3 has potential value in the diagnosis and therapy of patients with MI.
{"title":"MicroPET Imaging of Riboflavin Transporter 3 Expression in Myocardial Infarction/Reperfusion Rat Models with Radiofluorinated Riboflavin","authors":"Jindian Li, Xingfang Hong, Yingxi Chen, Bin Yin, Hongzhang Yang, Changrong Shi, Xinying Zeng, Deliang Zhang*, Zhide Guo* and Xianzhong Zhang*, ","doi":"10.1021/acsptsci.4c0017510.1021/acsptsci.4c00175","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00175https://doi.org/10.1021/acsptsci.4c00175","url":null,"abstract":"<p >Riboflavin transporter 3 (RFVT3) represents a potential cardioprotective biotarget in energetic metabolism reprogramming after myocardial infarction/reperfusion (MI/R). This study investigated the feasibility of noninvasive real-time quantification of RFVT3 expression after MI/R with an radiolabeled probe <sup>18</sup>F-RFTA in a preclinical rat model of MI/R. The tracer <sup>18</sup>F-RFTA was radio-synthesized manually and characterized on the subjects of radiolabeling yield, radiochemical purity, and stability <i>in vivo</i>. MI/R and sham-operated rat models were confirmed by cardiac magnetic resonance imaging (cMRI) and single-photon-emission computed tomography (SPECT) myocardial perfusion imaging (MPI) with technetium-99m sestamibi (<sup>99m</sup>Tc-MIBI). Positron emission tomography (PET) imaging of MI/R and sham-operated rat models were conducted with <sup>18</sup>F-RFTA. <i>Ex vivo</i> autoradiography and RFVT3 immunohistochemical (IHC) staining were conducted to verify the RFVT3 expression in infarcted and normal myocardium. <sup>18</sup>F-RFTA injection was prepared with high radiochemical purity (>95%) and kept stable <i>in vitro</i> and <i>in vivo</i>. <sup>18</sup>F-RFTA PET revealed significant uptake in the infarcted myocardium at 8 h after reperfusion, as confirmed by lower <sup>99m</sup>Tc-MIBI perfusion and decreased intensity of cMRI. Conversely, there were only the tiniest uptakes in the normal myocardium and blocked infarcted myocardium, which was further corroborated by <i>ex vivo</i> autoradiography. The RFVT3 expression was further confirmed by IHC staining in the infarcted and normal myocardium. We first demonstrate the feasibility of imaging RFVT3 in infarcted myocardium. <sup>18</sup>F-RFTA is an encouraging PET probe for imaging cardioprotective biotarget RFVT3 in mitochondrial energetic metabolism reprogramming after myocardial infarction. Noninvasive imaging of cardioprotective biotarget RFVT3 has potential value in the diagnosis and therapy of patients with MI.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2350–2357 2350–2357"},"PeriodicalIF":4.9,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141956351","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-24DOI: 10.1021/acsptsci.4c0030810.1021/acsptsci.4c00308
Jiaqi Huang, Aishik Chakraborty, Lakshmi Suchitra Tadepalli and Arghya Paul*,
DNA nanostructures have been widely researched in recent years as emerging biomedical materials for drug delivery, biosensing, and cancer therapy, in addition to their hereditary function. Multiple precisely designed single-strand DNAs can be fabricated into complex, three-dimensional DNA nanostructures through a simple self-assembly process. Among all of the synthetic DNA nanostructures, tetrahedral DNA nanostructures (TDNs) stand out as the most promising biomedical nanomaterial. TDNs possess the merits of structural stability, cell membrane permeability, and natural biocompatibility due to their compact structures and DNA origin. In addition to their inherent advantages, TDNs were shown to have great potential in delivering therapeutic agents through multiple functional modifications. As a multifunctional material, TDNs have enabled innovative pharmaceutical applications, including antimicrobial therapy, anticancer treatment, immune modulation, and cartilage regeneration. Given the rapid development of TDNs in the biomedical field, it is critical to understand how to successfully produce and fine-tune the properties of TDNs for specific therapeutic needs and clinical translation. This article provides insights into the synthesis and functionalization of TDNs and summarizes the approaches for TDN-based therapeutics delivery as well as their broad applications in the field of pharmaceutics and nanomedicine, challenges, and future directions.
DNA 纳米结构作为一种新兴的生物医学材料,除了具有遗传功能外,还可用于药物输送、生物传感和癌症治疗,近年来已被广泛研究。通过简单的自组装过程,可以将多条精确设计的单链 DNA 制成复杂的三维 DNA 纳米结构。在所有合成的 DNA 纳米结构中,四面体 DNA 纳米结构(TDNs)是最有前途的生物医学纳米材料。四面体 DNA 纳米结构因其紧凑的结构和 DNA 起源而具有结构稳定性、细胞膜渗透性和天然生物相容性等优点。除了其固有的优点外,TDNs 还通过多种功能修饰,在递送治疗药物方面具有巨大潜力。作为一种多功能材料,TDNs 实现了创新药物应用,包括抗菌治疗、抗癌治疗、免疫调节和软骨再生。鉴于 TDNs 在生物医学领域的快速发展,了解如何成功生产和微调 TDNs 的特性以满足特定的治疗需求和临床转化至关重要。本文深入探讨了 TDNs 的合成和功能化,总结了基于 TDN 的治疗递送方法及其在制药学和纳米医学领域的广泛应用、挑战和未来发展方向。
{"title":"Adoption of a Tetrahedral DNA Nanostructure as a Multifunctional Biomaterial for Drug Delivery","authors":"Jiaqi Huang, Aishik Chakraborty, Lakshmi Suchitra Tadepalli and Arghya Paul*, ","doi":"10.1021/acsptsci.4c0030810.1021/acsptsci.4c00308","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00308https://doi.org/10.1021/acsptsci.4c00308","url":null,"abstract":"<p >DNA nanostructures have been widely researched in recent years as emerging biomedical materials for drug delivery, biosensing, and cancer therapy, in addition to their hereditary function. Multiple precisely designed single-strand DNAs can be fabricated into complex, three-dimensional DNA nanostructures through a simple self-assembly process. Among all of the synthetic DNA nanostructures, tetrahedral DNA nanostructures (TDNs) stand out as the most promising biomedical nanomaterial. TDNs possess the merits of structural stability, cell membrane permeability, and natural biocompatibility due to their compact structures and DNA origin. In addition to their inherent advantages, TDNs were shown to have great potential in delivering therapeutic agents through multiple functional modifications. As a multifunctional material, TDNs have enabled innovative pharmaceutical applications, including antimicrobial therapy, anticancer treatment, immune modulation, and cartilage regeneration. Given the rapid development of TDNs in the biomedical field, it is critical to understand how to successfully produce and fine-tune the properties of TDNs for specific therapeutic needs and clinical translation. This article provides insights into the synthesis and functionalization of TDNs and summarizes the approaches for TDN-based therapeutics delivery as well as their broad applications in the field of pharmaceutics and nanomedicine, challenges, and future directions.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2204–2214 2204–2214"},"PeriodicalIF":4.9,"publicationDate":"2024-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141958200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-22DOI: 10.1021/acsptsci.4c0025610.1021/acsptsci.4c00256
Xin Hu*, Paul Shinn, Zina Itkin, Lin Ye, Ya-Qin Zhang, Min Shen, Stephanie Ford-Scheimer and Matthew D. Hall*,
As part of the NIH Helping to End Addiction Long-term (HEAL) Initiative, the National Center for Advancing Translational Sciences is dedicated to the development of new pharmacological tools and investigational drugs for managing and treating pain as well as the prevention and treatment of opioid misuse and addiction. In line with these objectives, we created a comprehensive, annotated small molecule library including drugs, probes, and tool compounds that act on published pain- and addiction-relevant targets. Nearly 3000 small molecules associated with approximately 200 known and hypothesized HEAL targets have been assembled, curated, and annotated in one collection. Physical samples of the library compounds have been acquired and plated in 1536-well format, enabling a rapid and efficient high-throughput screen against a wide range of assays. The creation of the HEAL Targets and Compounds Library, coupled with an integrated computational platform for AI-driven machine learning, structural modeling, and virtual screening, provides a valuable source for strategic drug repurposing, innovative profiling, and hypothesis testing of novel targets related to pain and opioid use disorder (OUD). The library is available to investigators for screening pain and OUD-relevant phenotypes.
作为美国国立卫生研究院(NIH)"帮助戒除毒瘾长期计划"(HEAL)的一部分,美国国家转化科学促进中心(National Center for Advancing Translational Sciences)致力于开发新的药理学工具和研究药物,用于控制和治疗疼痛以及预防和治疗阿片类药物滥用和成瘾。根据这些目标,我们创建了一个全面的、附有注释的小分子化合物库,其中包括作用于已公布的疼痛和成瘾相关靶点的药物、探针和工具化合物。与大约 200 个已知和假设的 HEAL 靶点相关的近 3000 种小分子化合物已被收集、整理和注释在一个集合中。该化合物库的物理样本已采集完毕,并以 1536 孔的格式进行了培养,从而能够针对各种检测方法进行快速、高效的高通量筛选。HEAL 靶点和化合物库的建立,加上人工智能驱动的机器学习、结构建模和虚拟筛选的集成计算平台,为疼痛和阿片类药物使用障碍 (OUD) 相关新靶点的战略药物再利用、创新性分析和假设检验提供了宝贵的资源。研究人员可利用该库筛选与疼痛和阿片类药物使用障碍相关的表型。
{"title":"A Comprehensive Collection of Pain and Opioid Use Disorder Compounds for High-Throughput Screening and Artificial Intelligence-Driven Drug Discovery","authors":"Xin Hu*, Paul Shinn, Zina Itkin, Lin Ye, Ya-Qin Zhang, Min Shen, Stephanie Ford-Scheimer and Matthew D. Hall*, ","doi":"10.1021/acsptsci.4c0025610.1021/acsptsci.4c00256","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00256https://doi.org/10.1021/acsptsci.4c00256","url":null,"abstract":"<p >As part of the NIH Helping to End Addiction Long-term (HEAL) Initiative, the National Center for Advancing Translational Sciences is dedicated to the development of new pharmacological tools and investigational drugs for managing and treating pain as well as the prevention and treatment of opioid misuse and addiction. In line with these objectives, we created a comprehensive, annotated small molecule library including drugs, probes, and tool compounds that act on published pain- and addiction-relevant targets. Nearly 3000 small molecules associated with approximately 200 known and hypothesized HEAL targets have been assembled, curated, and annotated in one collection. Physical samples of the library compounds have been acquired and plated in 1536-well format, enabling a rapid and efficient high-throughput screen against a wide range of assays. The creation of the HEAL Targets and Compounds Library, coupled with an integrated computational platform for AI-driven machine learning, structural modeling, and virtual screening, provides a valuable source for strategic drug repurposing, innovative profiling, and hypothesis testing of novel targets related to pain and opioid use disorder (OUD). The library is available to investigators for screening pain and OUD-relevant phenotypes.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2391–2400 2391–2400"},"PeriodicalIF":4.9,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141957940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Claudin18.2 (CLDN18.2) has emerged as a significant target in the treatment of advanced gastric cancer. The screening of patients positive for CLDN18.2 is crucial for the effective application of targeted therapies specific to CLND18.2. In this study, we developed a novel nanobody-based probe, [99mTc]Tc-PHG102, for use in nuclear medicine. We analyzed its radiochemical yield and stability to ensure accurate probe characterization. Additionally, we assessed the probe’s affinity and specificity toward the CLDN18.2 target and evaluated its efficacy in the BGC82318.2 xenograft model for SPECT/CT imaging of gastric cancer. The binding of [99mTc]Tc-PHG102 to HEK-293T18.2 and BGC82318.2 cells was notably higher than its binding to HEK-293T18.1, HEK-293T, and BGC823 cells, with bound values of 12.87 ± 1.46%, 6.16 ± 0.34%, 1.25 ± 0.22%, 1.14 ± 0.26%, and 1.32 ± 0.07% AD, respectively. The binding ability of [99mTc]Tc-PHG102 was significantly different between CLDN18.2-positive and negative cells (P < 0.001). Imaging results demonstrated a time-dependent tumor accumulation of the radiotracer. Notably, at 0.5 h postinjection, rapid accumulation was observed with an average tumor uptake of 4.63 ± 0.81% ID/cc (n = 3), resulting in clear tumor visualization. By 1 h postinjection, as [99mTc]Tc-PHG102 was rapidly metabolized, a decrease in uptake by other organs was noted. Preliminary clinical imaging trials further confirmed the safety and effectiveness of the probe, indicating specificity for lesions expressing CLDN18.2 in gastric cancer and favorable in vivo metabolic properties. In conclusion, the nanobody-based probe [99mTc]Tc-PHG102 proves to be a safe and effective tool for detecting CLDN18.2 expression levels in gastric cancer tumors and for screening CLDN18.2-positive patients.
{"title":"Claudin18.2-Targeted SPECT/CT Imaging for Gastric Cancer: Preclinical Evaluation and Clinical Translation of the 99mTc-Labeled Nanobody (PHG102) Radiotracer","authors":"Zhidong Bai, Xin Xie, Chenzhen Li, Yuchen Wang, Yuanbo Wang, Huijie Li, Rui Gao* and Bing Jia*, ","doi":"10.1021/acsptsci.4c0028010.1021/acsptsci.4c00280","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00280https://doi.org/10.1021/acsptsci.4c00280","url":null,"abstract":"<p >Claudin18.2 (CLDN18.2) has emerged as a significant target in the treatment of advanced gastric cancer. The screening of patients positive for CLDN18.2 is crucial for the effective application of targeted therapies specific to CLND18.2. In this study, we developed a novel nanobody-based probe, [<sup>99m</sup>Tc]Tc-PHG102, for use in nuclear medicine. We analyzed its radiochemical yield and stability to ensure accurate probe characterization. Additionally, we assessed the probe’s affinity and specificity toward the CLDN18.2 target and evaluated its efficacy in the BGC823<sup>18.2</sup> xenograft model for SPECT/CT imaging of gastric cancer. The binding of [<sup>99m</sup>Tc]Tc-PHG102 to HEK-293T<sup>18.2</sup> and BGC823<sup>18.2</sup> cells was notably higher than its binding to HEK-293T<sup>18.1</sup>, HEK-293T, and BGC823 cells, with bound values of 12.87 ± 1.46%, 6.16 ± 0.34%, 1.25 ± 0.22%, 1.14 ± 0.26%, and 1.32 ± 0.07% AD, respectively. The binding ability of [<sup>99m</sup>Tc]Tc-PHG102 was significantly different between CLDN18.2-positive and negative cells (<i>P</i> < 0.001). Imaging results demonstrated a time-dependent tumor accumulation of the radiotracer. Notably, at 0.5 h postinjection, rapid accumulation was observed with an average tumor uptake of 4.63 ± 0.81% ID/cc (<i>n</i> = 3), resulting in clear tumor visualization. By 1 h postinjection, as [<sup>99m</sup>Tc]Tc-PHG102 was rapidly metabolized, a decrease in uptake by other organs was noted. Preliminary clinical imaging trials further confirmed the safety and effectiveness of the probe, indicating specificity for lesions expressing CLDN18.2 in gastric cancer and favorable in vivo metabolic properties. In conclusion, the nanobody-based probe [<sup>99m</sup>Tc]Tc-PHG102 proves to be a safe and effective tool for detecting CLDN18.2 expression levels in gastric cancer tumors and for screening CLDN18.2-positive patients.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2465–2475 2465–2475"},"PeriodicalIF":4.9,"publicationDate":"2024-07-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141957853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-18DOI: 10.1021/acsptsci.4c0029610.1021/acsptsci.4c00296
Liu Tang, Huiliang Geng, Lei Zhang*, Xinyi Wang, Mengdan Fei, Boyuan Yang, Haijie Sun and Zhongli Zhang*,
The oxidation of the complementarity-determining region (CDR) in monoclonal antibodies (mAbs) is a critical quality attribute that can affect the clinical efficacy and safety of recombinant mAb therapeutics. In this study, a robust hydrophobic interaction chromatography (HIC) method was developed to quantify and characterize CDR oxidation variants in mAb-A by using a Proteomix Butyl-NP5 column. The HIC analysis revealed oxidation variants that eluted earlier than the main species with weaker hydrophobicity. It was found that Met105 in the CDR was more susceptible to oxidation. Additionally, it was noted that the oxidation of Met105 on a single heavy chain resulted in elution at a distinct position compared to the oxidation on two heavy chains. This observation led to the fractionation and enrichment of the oxidized variants for further evaluation of their biofunction. The study also demonstrated that the oxidation of Met105 did not impact the antigen-binding capacity but significantly reduced the PD-1/PD-L1 blockade activity of mAb-A. The HIC method, which was employed to quantify CDR oxidation, underwent validation and was subsequently utilized for stability studies as well as for assessing the similarity between mAb-A and its reference product.
单克隆抗体(mAb)中互补决定区(CDR)的氧化是影响重组 mAb 疗法临床疗效和安全性的关键质量属性。本研究利用 Proteomix Butyl-NP5 色谱柱开发了一种稳健的疏水相互作用色谱(HIC)方法,用于定量和定性 mAb-A 中的 CDR 氧化变体。通过 HIC 分析发现,氧化变体的疏水性较弱,比主变体更早洗脱。研究发现,CDR 中的 Met105 更容易被氧化。此外,研究人员还注意到,与两条重链上的氧化相比,单条重链上的 Met105 氧化导致洗脱位置不同。根据这一观察结果,对氧化变体进行了分馏和富集,以进一步评估其生物功能。研究还表明,Met105 的氧化不会影响抗原结合能力,但会显著降低 mAb-A 的 PD-1/PD-L1 阻断活性。用于量化 CDR 氧化的 HIC 方法经过了验证,随后被用于稳定性研究以及评估 mAb-A 与其参考品之间的相似性。
{"title":"In-Depth Characterization for Methionine Oxidization in Complementary Domain Region by Hydrophobic Interaction Chromatography","authors":"Liu Tang, Huiliang Geng, Lei Zhang*, Xinyi Wang, Mengdan Fei, Boyuan Yang, Haijie Sun and Zhongli Zhang*, ","doi":"10.1021/acsptsci.4c0029610.1021/acsptsci.4c00296","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00296https://doi.org/10.1021/acsptsci.4c00296","url":null,"abstract":"<p >The oxidation of the complementarity-determining region (CDR) in monoclonal antibodies (mAbs) is a critical quality attribute that can affect the clinical efficacy and safety of recombinant mAb therapeutics. In this study, a robust hydrophobic interaction chromatography (HIC) method was developed to quantify and characterize CDR oxidation variants in mAb-A by using a Proteomix Butyl-NP5 column. The HIC analysis revealed oxidation variants that eluted earlier than the main species with weaker hydrophobicity. It was found that Met<sub>105</sub> in the CDR was more susceptible to oxidation. Additionally, it was noted that the oxidation of Met<sub>105</sub> on a single heavy chain resulted in elution at a distinct position compared to the oxidation on two heavy chains. This observation led to the fractionation and enrichment of the oxidized variants for further evaluation of their biofunction. The study also demonstrated that the oxidation of Met<sub>105</sub> did not impact the antigen-binding capacity but significantly reduced the PD-1/PD-L1 blockade activity of mAb-A. The HIC method, which was employed to quantify CDR oxidation, underwent validation and was subsequently utilized for stability studies as well as for assessing the similarity between mAb-A and its reference product.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2476–2483 2476–2483"},"PeriodicalIF":4.9,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141957654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rheumatoid arthritis (RA) is a complex condition that is influenced by various causes, including immunological, genetic, and environmental factors. Several studies using animal models have documented immune system dysfunction and described the clinical characteristics of the disease. These studies have provided valuable insights into the pathogenesis of inflammatory arthritis and the identification of new targets for treatment. Nevertheless, none of these animal models successfully replicated all the characteristics of RA. Additionally, numerous experimental medications, which were developed based on our enhanced comprehension of the immune system’s function in RA, have shown potential in animal research but ultimately proved ineffective during different stages of clinical trials. There have been several novel therapy alternatives, which do not achieve a consistently outstanding therapeutic outcome in all patients. This underscores the importance of employing the progress in in vitro models, particularly 3D models like tissue explants, and diverse multicomponent approaches such as coculture strategies, synovial membrane, articular cartilage, and subchondral bone models that accurately replicate the structural characteristics of RA pathophysiology. These methods are crucial for the advancement of potential therapeutic strategies. This review discusses the latest advancements in in vitro models and their potential to greatly impact research on managing RA.
类风湿性关节炎(RA)是一种复杂的疾病,受多种原因的影响,包括免疫、遗传和环境因素。一些利用动物模型进行的研究记录了免疫系统功能紊乱的情况,并描述了该疾病的临床特征。这些研究为了解炎症性关节炎的发病机制和确定新的治疗靶点提供了宝贵的资料。然而,这些动物模型都没有成功复制出 RA 的所有特征。此外,根据我们对免疫系统在 RA 中的功能的进一步理解而开发的许多实验性药物在动物研究中显示出了潜力,但最终在不同阶段的临床试验中被证明无效。目前已有几种新型替代疗法,但并不是所有患者都能持续获得出色的治疗效果。这凸显了采用体外模型的重要性,尤其是三维模型,如组织外植体,以及多种多成分方法,如共培养策略、滑膜、关节软骨和软骨下骨模型,这些都能准确复制 RA 病理生理学的结构特征。这些方法对于推进潜在的治疗策略至关重要。本综述将讨论体外模型的最新进展及其对控制 RA 的研究产生重大影响的潜力。
{"title":"Emerging Landscape of In Vitro Models for Assessing Rheumatoid Arthritis Management","authors":"Abhay Prakash Mishra, Rajesh Kumar, Seetha Harilal, Manisha Nigam*, Deepanjan Datta and Sudarshan Singh*, ","doi":"10.1021/acsptsci.4c0026010.1021/acsptsci.4c00260","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00260https://doi.org/10.1021/acsptsci.4c00260","url":null,"abstract":"<p >Rheumatoid arthritis (RA) is a complex condition that is influenced by various causes, including immunological, genetic, and environmental factors. Several studies using animal models have documented immune system dysfunction and described the clinical characteristics of the disease. These studies have provided valuable insights into the pathogenesis of inflammatory arthritis and the identification of new targets for treatment. Nevertheless, none of these animal models successfully replicated all the characteristics of RA. Additionally, numerous experimental medications, which were developed based on our enhanced comprehension of the immune system’s function in RA, have shown potential in animal research but ultimately proved ineffective during different stages of clinical trials. There have been several novel therapy alternatives, which do not achieve a consistently outstanding therapeutic outcome in all patients. This underscores the importance of employing the progress in <i>in vitro</i> models, particularly 3D models like tissue explants, and diverse multicomponent approaches such as coculture strategies, synovial membrane, articular cartilage, and subchondral bone models that accurately replicate the structural characteristics of RA pathophysiology. These methods are crucial for the advancement of potential therapeutic strategies. This review discusses the latest advancements in <i>in vitro</i> models and their potential to greatly impact research on managing RA.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2280–2305 2280–2305"},"PeriodicalIF":4.9,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141956002","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-18DOI: 10.1021/acsptsci.4c0018610.1021/acsptsci.4c00186
Youyi Peng*, Allen H. Zhang, Liping Wei and William J. Welsh*,
The global prevalence of diabetes is steadily rising, with an estimated 537 million adults affected by diabetes in 2021, projected to reach 783 million by 2045. A severe consequence of diabetes is the development of painful diabetic neuropathy (PDN), afflicting approximately one in every three diabetic patients and significantly compromising their quality of life. Current pharmacotherapies for PDN provide inadequate pain relief for many patients, underscoring the need for novel treatments that are both safe and effective. The Sigma 1 Receptor (S1R) is a ligand-operated chaperone protein that resides at the mitochondria-associated membrane of the endoplasmic reticulum. The S1R has been shown to play crucial roles in regulating cellular processes implicated in pain modulation. This study explores the potential of PW507, a novel S1R antagonist, as a therapeutic candidate for PDN. PW507 exhibited promising in vitro and in vivo properties in terms of ADME, toxicity, pharmacokinetics, and safety. In preclinical rat models of Streptozotocin-induced diabetic neuropathy, PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following both acute and chronic (2-week) administration, without inducing tolerance and visual evidence of toxicity. To the best of our knowledge, this is the first report to evaluate an S1R antagonist in STZ-induced diabetic rats following both acute and 2-week chronic administration, offering compelling preclinical evidence for the potential use of PW507 as a promising therapeutic option for PDN.
{"title":"Preclinical Evaluation of Sigma 1 Receptor Antagonists as a Novel Treatment for Painful Diabetic Neuropathy","authors":"Youyi Peng*, Allen H. Zhang, Liping Wei and William J. Welsh*, ","doi":"10.1021/acsptsci.4c0018610.1021/acsptsci.4c00186","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00186https://doi.org/10.1021/acsptsci.4c00186","url":null,"abstract":"<p >The global prevalence of diabetes is steadily rising, with an estimated 537 million adults affected by diabetes in 2021, projected to reach 783 million by 2045. A severe consequence of diabetes is the development of painful diabetic neuropathy (PDN), afflicting approximately one in every three diabetic patients and significantly compromising their quality of life. Current pharmacotherapies for PDN provide inadequate pain relief for many patients, underscoring the need for novel treatments that are both safe and effective. The Sigma 1 Receptor (S1R) is a ligand-operated chaperone protein that resides at the mitochondria-associated membrane of the endoplasmic reticulum. The S1R has been shown to play crucial roles in regulating cellular processes implicated in pain modulation. This study explores the potential of PW507, a novel S1R antagonist, as a therapeutic candidate for PDN. PW507 exhibited promising <i>in vitro</i> and <i>in vivo</i> properties in terms of ADME, toxicity, pharmacokinetics, and safety. In preclinical rat models of Streptozotocin-induced diabetic neuropathy, PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following both acute and chronic (2-week) administration, without inducing tolerance and visual evidence of toxicity. To the best of our knowledge, this is the first report to evaluate an S1R antagonist in STZ-induced diabetic rats following both acute and 2-week chronic administration, offering compelling preclinical evidence for the potential use of PW507 as a promising therapeutic option for PDN.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2358–2368 2358–2368"},"PeriodicalIF":4.9,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141955958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-17DOI: 10.1021/acsptsci.4c0031410.1021/acsptsci.4c00314
Kenan Aloss, Pedro Henrique Leroy Viana, Syeda Mahak Zahra Bokhari, Nino Giunashvili, Csaba András Schvarcz, Dániel Bócsi, Zoltán Koós, Zoltán Benyó and Péter Hamar*,
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, with limited treatment options. Modulated electro-hyperthermia (mEHT) is a novel adjuvant cancer therapy that induces selective cancer damage. However, mEHT upregulates heat shock protein beta 1 (HSPB1), a cancer-promoting stress chaperone molecule. Thus, we investigated whether ivermectin (IVM), an anthelmintic drug, may synergize with mEHT and enhance its anticancer effects by inhibiting HSPB1 phosphorylation. Isogenic 4T1 TNBC cells were inoculated into BALB/c mice and treated with mEHT, IVM, or a combination of both. IVM synergistically improved the tumor growth inhibition achieved by mEHT. Moreover, IVM downregulated mEHT-induced HSPB1 phosphorylation. Thus, the strongest cancer tissue damage was observed in the mEHT + IVM-treated tumors, coupled with the strongest apoptosis induction and proliferation inhibition. In addition, there was no significant body weight loss in mice treated with mEHT and IVM, indicating that this combination was well-tolerated. In conclusion, mEHT combined with IVM is a new, effective, and safe option for the treatment of TNBC.
{"title":"Ivermectin Synergizes with Modulated Electro-hyperthermia and Improves Its Anticancer Effects in a Triple-Negative Breast Cancer Mouse Model","authors":"Kenan Aloss, Pedro Henrique Leroy Viana, Syeda Mahak Zahra Bokhari, Nino Giunashvili, Csaba András Schvarcz, Dániel Bócsi, Zoltán Koós, Zoltán Benyó and Péter Hamar*, ","doi":"10.1021/acsptsci.4c0031410.1021/acsptsci.4c00314","DOIUrl":"https://doi.org/10.1021/acsptsci.4c00314https://doi.org/10.1021/acsptsci.4c00314","url":null,"abstract":"<p >Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, with limited treatment options. Modulated electro-hyperthermia (mEHT) is a novel adjuvant cancer therapy that induces selective cancer damage. However, mEHT upregulates heat shock protein beta 1 (HSPB1), a cancer-promoting stress chaperone molecule. Thus, we investigated whether ivermectin (IVM), an anthelmintic drug, may synergize with mEHT and enhance its anticancer effects by inhibiting HSPB1 phosphorylation. Isogenic 4T1 TNBC cells were inoculated into BALB/c mice and treated with mEHT, IVM, or a combination of both. IVM synergistically improved the tumor growth inhibition achieved by mEHT. Moreover, IVM downregulated mEHT-induced HSPB1 phosphorylation. Thus, the strongest cancer tissue damage was observed in the mEHT + IVM-treated tumors, coupled with the strongest apoptosis induction and proliferation inhibition. In addition, there was no significant body weight loss in mice treated with mEHT and IVM, indicating that this combination was well-tolerated. In conclusion, mEHT combined with IVM is a new, effective, and safe option for the treatment of TNBC.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"7 8","pages":"2496–2506 2496–2506"},"PeriodicalIF":4.9,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acsptsci.4c00314","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141955523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}