1
|
Martínez-Navarrete M, Correia M, Bernabeu-Martínez JA, Paiva-Santos AC, Borrego-Sánchez A, Guillot AJ. Enhanced ex vivo skin retention of bicalutamide using a nano-in-micro composite: Drug-loaded lipid vesicles in a dissolving microarray patch. Eur J Pharm Biopharm 2025:114728. [PMID: 40300672 DOI: 10.1016/j.ejpb.2025.114728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/07/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Antiandrogens are a family of drugs that block the biological effects of androgens, which are commonly used to treat a plethora of androgen-dependent conditions. However, their effectiveness highly depends on treatment compliance and their use is not exempt from side effects. Bicalutamide (BIC) is a testosterone blocker that, due to its peripheral tissue selectivity, offers the advantage of fewer side effects compared to other antiandrogens such as finasteride or dutasteride. In this study, BIC-loaded lipid vesicles (LVs) were developed and incorporated into a PVP/PVA-based Dissolving Microarray Patches (BIC-LVs@DMAPs) for BIC skin local delivery. First, the mechanical and insertion properties of the BIC-LVs@DMAPs were assessed. Then, the in vitro biocompatibility of the formulation was determined in keratinocytes. Finally, the effectiveness of the formulation to deliver BIC through skin was explored ex vivo using murine skin. BIC-LVs@DMAPs exhibited optimal mechanical and insertion properties to effectively perforate the stratum corneum and facilitate the passage of BIC-LVs. Ex vivo studies demonstrated the efficacy of BIC-LVs@DMAPs to improve the skin retention of BIC, while in vitro cytotoxicity results ensured their safety profile. Overall, these results pave the way for further in vivo studies and clinical application to improve the current early stages androgenetic alopecia topical treatments.
Collapse
Affiliation(s)
- Miquel Martínez-Navarrete
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avenue Vicent Andrés Estelles s/n. Spain. Valencia, Burjassot 46100, Spain
| | - Mafalda Correia
- Department of Pharmaceutical Technology, University of Coimbra. Polo das Ciências da Saúde, Azinhaga de Santa Comba. Portugal, Coimbra 3000-548, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra. Polo das Ciências da Saúde, Azinhaga de Santa Comba. Portugal, Coimbra 3000-548, Portugal
| | - J Alejandro Bernabeu-Martínez
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avenue Vicent Andrés Estelles s/n. Spain. Valencia, Burjassot 46100, Spain
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, University of Coimbra. Polo das Ciências da Saúde, Azinhaga de Santa Comba. Portugal, Coimbra 3000-548, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, University of Coimbra. Polo das Ciências da Saúde, Azinhaga de Santa Comba. Portugal, Coimbra 3000-548, Portugal
| | - Ana Borrego-Sánchez
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avenue Vicent Andrés Estelles s/n. Spain. Valencia, Burjassot 46100, Spain
| | - Antonio José Guillot
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avenue Vicent Andrés Estelles s/n. Spain. Valencia, Burjassot 46100, Spain.
| |
Collapse
|
2
|
Li H, Chen X, Rao S, Zhou M, Lu J, Liang D, Zhu B, Meng L, Lin J, Ding X, Zhang Q, Hu D. Recent development of micro-nano carriers for oral antineoplastic drug delivery. Mater Today Bio 2025; 30:101445. [PMID: 39866789 PMCID: PMC11762190 DOI: 10.1016/j.mtbio.2025.101445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Chemotherapy is widely recognized as a highly efficacious modality for cancer treatment, involving the administration of chemotherapeutic agents to target and eradicate tumor cells. Currently, oral administration stands as the prevailing and widely utilized method of delivering chemotherapy drugs. However, the majority of anti-tumor medications exhibit limited solubility and permeability, and poor stability in harsh gastrointestinal environments, thereby impeding their therapeutic efficacy for chemotherapy. Therefore, more and more micro-nano drug delivery carriers have been developed and used to effectively deliver anti-cancer drugs, which can overcome physiological barriers, facilitate oral administration, and ultimately improve drug efficacy. In this paper, we first discuss the effects of various biological barriers on micro-nano drug carriers and oral administration approach. Then, the development of micro-nano drug carriers based on various biomedical components, such as micelles, dendrimers, hydrogels, liposomes, inorganic nanoparticles, etc. were introduced. Finally, the current dilemma and the potential of oral drug delivery for clinical treatment were discussed. The primary objective of this review is to introduce various oral delivery methods and serve as a point of reference for the advancement of novel oral delivery carriers, with the ultimate goal of informing the development of future clinical applications.
Collapse
Affiliation(s)
- Hongzheng Li
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Xiang Chen
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shangrui Rao
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Minyu Zhou
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jianhua Lu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Danna Liang
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Bingzi Zhu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Letian Meng
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ji Lin
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoya Ding
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Qingfei Zhang
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Danhong Hu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
3
|
Meng J, Chan MY, Peng C, Jiang X, Qian F. Enhancing leuprolide penetration through enterocytes via the ER-Golgi pathway using lipophilic complexation. Eur J Pharm Biopharm 2025; 207:114624. [PMID: 39733960 DOI: 10.1016/j.ejpb.2024.114624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/12/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Oral delivery of peptide drugs remains one of the most formidable challenges in the frontier of pharmaceutical research. Peptide drugs typically suffer from exceptionally low oral bioavailability, primarily attributed to rigorous enzymatic degradation within the gastrointestinal (GI) tract, limited ability to traverse the enterocyte barrier, and significant first-pass hepatic metabolism. Absorption of peptide drugs via the lymphatic route could potentially bypass intracellular lysosome degradation and hepatic first-pass metabolism. In this study, we present a strategy to enhance the lymphatic absorption of the model peptide leuprolide (LEU) by diverting its intracellular trafficking towards the endoplasmic-reticulum (ER)-Golgi pathway. Complexes were formed between LEU and lipophilic excipient and then formulated as an oral emulsion. We observed that the penetration of LEU in the emulsion across the Caco-2 cell monolayer model was diverted from the endosome-lysosome pathway, and LEU entered the bloodstream via the mesenteric lymph nodes (MLNs). The data obtained illustrates that the lipophilic LEU complexes could improve enterocyte permeability and bypass lysosomal degradation, and the change of absorption pathway may reduce hepatic first pass metabolism.
Collapse
Affiliation(s)
- Jia Meng
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - May Yee Chan
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Cheng Peng
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Xuling Jiang
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Frontier Research Center for Biological Structure, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
4
|
Li J, Chen J, Shao X, Zhang N, Wang Y, Li Y. Flaxseed Linusorb Alleviates Collagen-Induced Rheumatoid Arthritis in Rats via Inhibiting the TLR4/NF-κb/MAPK Signal Pathway and Modulating Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27991-28004. [PMID: 39639764 DOI: 10.1021/acs.jafc.4c09582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Flaxseed linusorb (FL) has gradually garnered widespread attention in recent years because of its intriguing bioactivities like anti-inflammation, antimelanogenesis, and even anticancer effects. Based on its proven in vitro anti-inflammatory activity and mechanism, it is supposed that FL may also exhibit an in vivo effect in treating and preventing rheumatoid arthritis (RA). Collagen-induced arthritis (CIA) rat models were established to investigate the potential therapeutic effect of FL, which were intervened with FL via gavage (50 and 100 mg/kg B.W.) and intraperitoneal injection (10 and 20 mg/kg B.W.). After FL treatment, RA clinical symptoms were significantly alleviated, including reduced toe swelling volume and mitigated bone damage in CIA rats. Moreover, a decline in the expression of pro-inflammatory factors (i.e., TNF-α, IL-1β, and IL-6) and RA-related proteins (i.e., MMP-3, COX-2, and 5-LOX) was observed to effectively block the TLR4/NF-κB/MAPK signaling pathway. In addition, FL was discovered to modulate the diversity and composition of intestinal microbiota in CIA rats, where the level of g_Parvibacte, g_Allobaculum, g_Enterococcus, and unclassified_o_Lactobacillales could be significantly increased, whereas the level of Gram-negative bacteria g_Parabacteroides, g_Parasutterella, and g_Paludicola was notably reduced. In conclusion, FL shows promise in RA treatment by inhibiting inflammatory pathways and regulating the gut microbiota.
Collapse
Affiliation(s)
- Jialong Li
- Guangdong International Joint Research Center for Oilseed Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, College of Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou 510632, China
| | - Xin Shao
- Guangdong International Joint Research Center for Oilseed Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, College of Science and Engineering, Jinan University, Guangzhou 510632, China
- Department of Critical Care Medicine, Maoming People's Hospital, Maoming 512500, China
| | - Ning Zhang
- Guangdong International Joint Research Center for Oilseed Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, College of Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Wang
- Guangdong International Joint Research Center for Oilseed Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, College of Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Ying Li
- Guangdong International Joint Research Center for Oilseed Biorefinery, Nutrition and Safety, Department of Food Science and Engineering, College of Science and Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
5
|
Robinson KE, Moniz HA, Stokes AN, Feldman CR. Where Does All the Poison Go? Investigating Toxicokinetics of Newt (Taricha) Tetrodotoxin (TTX) in Garter Snakes (Thamnophis). J Chem Ecol 2024; 50:489-502. [PMID: 38842636 DOI: 10.1007/s10886-024-01517-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
Animals that consume toxic diets provide models for understanding the molecular and physiological adaptations to ecological challenges. Garter snakes (Thamnophis) in western North America prey on Pacific newts (Taricha), which employ tetrodotoxin (TTX) as an antipredator defense. These snakes possess mutations in voltage-gated sodium channels (Nav), the molecular targets of TTX, that decrease the binding ability of TTX to sodium channels (target-site resistance). However, genetic variation at these loci that cannot explain all the phenotypic variation in TTX resistance in Thamnophis. We explored a separate means of resistance, toxin metabolism, to determine if TTX-resistant snakes either rapidly remove TTX or sequester TTX. We examined the metabolism and distribution of TTX in the body (toxicokinetics), to determine differences between TTX-resistant and TTX-sensitive snakes in the rates at which TTX is eliminated from organs and the whole body (using TTX half-life as our metric). We assayed TTX half-life in snakes from TTX-resistant and TTX-sensitive populations of three garter snake species with a coevolutionary history with newts (T. atratus, T. couchii, T. sirtalis), as well as two non-resistant "outgroup" species (T. elegans, Pituophis catenifer) that seldom (if ever) engage newts. We found TTX half-life varied across species, populations, and tissues. Interestingly, TTX half-life was shortest in T. elegans and P. catenifer compared to all other snakes. Furthermore, TTX-resistant populations of T. couchii and T. sirtalis eliminated TTX faster (shorter TTX half-life) than their TTX-sensitive counterparts, while populations of TTX-resistant and TTX-sensitive T. atratus showed no difference rates of TTX removal (same TTX half-life). The ability to rapidly eliminate TTX may have permitted increased prey consumption, which may have promoted the evolution of additional resistance mechanisms. Finally, snakes still retain substantial amounts of TTX, and we projected that snakes could be dangerous to their own predators days to weeks following the ingestion of a single newt. Thus, aspects of toxin metabolism may have been key in driving predator-prey relationships, and important in determining other ecological interactions.
Collapse
Affiliation(s)
- Kelly E Robinson
- Department of Biology and Program in Ecology, Evolution and Conservation Biology, University of Nevada, Reno, Reno, NV, USA.
- Program in Ecology, Evolution and Conservation Biology, University of Nevada, Reno, NV, USA.
| | - Haley A Moniz
- Department of Biology and Program in Ecology, Evolution and Conservation Biology, University of Nevada, Reno, Reno, NV, USA
- Program in Ecology, Evolution and Conservation Biology, University of Nevada, Reno, NV, USA
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Amber N Stokes
- Department of Biology, California State University Bakersfield, Bakersfield, CA, USA
| | - Chris R Feldman
- Department of Biology and Program in Ecology, Evolution and Conservation Biology, University of Nevada, Reno, Reno, NV, USA
- Program in Ecology, Evolution and Conservation Biology, University of Nevada, Reno, NV, USA
| |
Collapse
|
6
|
Chu H, Xue J, Yang Y, Zheng H, Luo D, Li Z. Advances of Smart Stimulus-Responsive Microneedles in Cancer Treatment. SMALL METHODS 2024; 8:e2301455. [PMID: 38148309 DOI: 10.1002/smtd.202301455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/09/2023] [Indexed: 12/28/2023]
Abstract
Microneedles (MNs) have emerged as a highly promising technology for delivering drugs via the skin. They provide several benefits, including high drug bioavailability, non-invasiveness, painlessness, and high safety. Traditional strategies for intravenous delivery of anti-tumor drugs have risks of systemic toxicity and easy development of drug resistance, while MN technology facilitates precise delivery and on-demand release of drugs in local tissues. In addition, by further combining with stimulus-responsive materials, the construction of smart stimulus-responsive MNs can be achieved, which can respond to specific physical/chemical stimuli from the internal or external environment, thereby further improving the accuracy of tumor treatment and reducing toxicity to surrounding tissues/cells. This review systematically summarizes the classification, materials, and reaction mechanisms of stimulus-responsive MNs, outlines the benefits and challenges of various types of MNs, and details their application and latest progress in cancer treatment. Finally, the development prospects of smart MNs in tumor treatment are also discussed, bringing inspiration for future precision treatment of tumors.
Collapse
Affiliation(s)
- Huaqing Chu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Jiangtao Xue
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuan Yang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dan Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| |
Collapse
|
7
|
Pace J, Lee JJ, Srinivasarao M, Kallepu S, Low PS, Niedre M. In Vivo Labeling and Detection of Circulating Tumor Cells in Mice Using OTL38. Mol Imaging Biol 2024; 26:603-615. [PMID: 38594545 PMCID: PMC11281960 DOI: 10.1007/s11307-024-01914-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/04/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE We recently developed an optical instrument to non-invasively detect fluorescently labeled circulating tumor cells (CTCs) in mice called 'Diffuse in vivo Flow Cytometry' (DiFC). OTL38 is a folate receptor (FR) targeted near-infrared (NIR) contrast agent that is FDA approved for use in fluorescence guided surgery of ovarian and lung cancer. In this work, we investigated the use OTL38 for in vivo labeling and detection of FR + CTCs with DiFC. PROCEDURES We tested OTL38 labeling of FR + cancer cell lines (IGROV-1 and L1210A) as well as FR- MM.1S cells in suspensions of Human Peripheral Blood Mononuclear cells (PBMCs) in vitro. We also tested OTL38 labeling and NIR-DIFC detection of FR + L1210A cells in blood circulation in nude mice in vivo. RESULTS 62% of IGROV-1 and 83% of L1210A were labeled above non-specific background levels in suspensions of PBMCs in vitro compared to only 2% of FR- MM.1S cells. L1210A cells could be labeled with OTL38 directly in circulation in vivo and externally detected using NIR-DiFC in mice with low false positive detection rates. CONCLUSIONS This work shows the feasibility of labeling CTCs in vivo with OTL38 and detection with DiFC. Although further refinement of the DiFC instrument and signal processing algorithms and testing with other animal models is needed, this work may eventually pave the way for human use of DiFC.
Collapse
Affiliation(s)
- Joshua Pace
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Jane J Lee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | | | | | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, 047906, USA
| | - Mark Niedre
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Hosseini MAH, Alizadeh AA, Shayanfar A. Prediction of the First-Pass Metabolism of a Drug After Oral Intake Based on Structural Parameters and Physicochemical Properties. Eur J Drug Metab Pharmacokinet 2024; 49:449-465. [PMID: 38733548 DOI: 10.1007/s13318-024-00892-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND AND OBJECTIVE The oral first-pass metabolism is a crucial factor that plays a key role in a drug's pharmacokinetic profile. Prediction of the oral first-pass metabolism based on chemical structural parameters can be useful in the drug-design process. Developing an orally administered drug with an acceptable pharmacokinetic profile is necessary to reduce the cost and time associated with evaluating the extent of the first-pass metabolism of a candidate compound in preclinical studies. The aim of this study is to estimate the first-pass metabolism of an orally administered drug. METHODS A set of compounds with reported first-pass metabolism data were collected. Moreover, human intestinal absorption percentage and oral bioavailability data were extracted from the literature to propose a classification system that split the drugs up based on their first-pass metabolism extents. Various structural parameters were calculated for each compound. The relations of the structural and physicochemical values of each compound to the class the compound belongs to were obtained using logistic regression. RESULTS Initial analysis showed that compounds with logD7.4 > 1 or a rugosity factor of > 1.5 are more likely to have high first-pass metabolism. Four different models that can predict the oral first-pass metabolism with acceptable error were introduced. The overall accuracies of the models were in the range of 72% (for models with simple descriptors) to 78% (for models with complex descriptors). Although the models with simple descriptors have lower accuracies compared to complex models, they are more interpretable and easier for researchers to utilize. CONCLUSION A novel classification of drugs based on the extent of the oral first-pass metabolism was introduced, and mechanistic models were developed to assign candidate compounds to the appropriate proposed classes.
Collapse
Affiliation(s)
- Mir Amir Hossein Hosseini
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Alizadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shayanfar
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Golgasht St., Tabriz, 51664-14766, Iran.
| |
Collapse
|
9
|
Esquea EM, Ciraku L, Young RG, Merzy J, Talarico AN, Ahmed NN, Karuppiah M, Ramesh A, Chatoff A, Crispim CV, Rashad AA, Cocklin S, Snyder NW, Beld J, Simone NL, Reginato MJ, Dick A. Selective and brain-penetrant ACSS2 inhibitors target breast cancer brain metastatic cells. Front Pharmacol 2024; 15:1394685. [PMID: 38818373 PMCID: PMC11137182 DOI: 10.3389/fphar.2024.1394685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
Breast cancer brain metastasis (BCBM) typically results in an end-stage diagnosis and is hindered by a lack of brain-penetrant drugs. Tumors in the brain rely on the conversion of acetate to acetyl-CoA by the enzyme acetyl-CoA synthetase 2 (ACSS2), a key regulator of fatty acid synthesis and protein acetylation. Here, we used a computational pipeline to identify novel brain-penetrant ACSS2 inhibitors combining pharmacophore-based shape screen methodology with absorption, distribution, metabolism, and excretion (ADME) property predictions. We identified compounds AD-5584 and AD-8007 that were validated for specific binding affinity to ACSS2. Treatment of BCBM cells with AD-5584 and AD-8007 leads to a significant reduction in colony formation, lipid storage, acetyl-CoA levels and cell survival in vitro. In an ex vivo brain-tumor slice model, treatment with AD-8007 and AD-5584 reduced pre-formed tumors and synergized with irradiation in blocking BCBM tumor growth. Treatment with AD-8007 reduced tumor burden and extended survival in vivo. This study identifies selective brain-penetrant ACSS2 inhibitors with efficacy towards breast cancer brain metastasis.
Collapse
Affiliation(s)
- Emily M. Esquea
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Lorela Ciraku
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Riley G. Young
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jessica Merzy
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Alexandra N. Talarico
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nusaiba N. Ahmed
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Mangalam Karuppiah
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Anna Ramesh
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Adam Chatoff
- Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Claudia V. Crispim
- Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Adel A. Rashad
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Simon Cocklin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nathaniel W. Snyder
- Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Joris Beld
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nicole L. Simone
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
- Cancer Risk and Control Program, Philadelphia, PA, United States
| | - Mauricio J. Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
- Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexej Dick
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
10
|
Yang Q, Fan L, Hao E, Hou X, Deng J, Xia Z, Du Z. Construction of An Oral Bioavailability Prediction Model Based on Machine Learning for Evaluating Molecular Modifications. J Pharm Sci 2024; 113:1155-1167. [PMID: 38430955 DOI: 10.1016/j.xphs.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE This study aims to explore the impact of ADME on the Oral Bioavailability (OB) of drugs and to construct a machine learning model for OB prediction. The model is then applied to predict the OB of modified berberine and atenolol molecules to obtain structures with higher OB. METHODS Initially, a drug OB database was established, and corresponding ADME characteristics were obtained. The relationship between ADME and OB was analyzed using machine learning, with Morgan fingerprints serving as molecular descriptors. Compounds from the database were input into Random Forest, XGBoost, CatBoost, and LightGBM machine learning models to train the OB 7prediction model and evaluate its performance. Subsequently, berberine and atenolol were modified using Chemdraw software with ten different substituents for mono-substitution, and chlorine atoms for a full range of double substitutions. The modified molecular structures were converted into the same format as the training set for OB prediction. The predicted OB values of the modified structures of berberine and atenolol were compared. RESULTS An OB database of 386 drugs was obtained. It was found that smaller molecular weight and a higher number of rotatable bonds (ten or less) could potentially lead to higher OB. The four machine learning models were evaluated using MSE, R2 score, MAE, and MFE as metrics, with Random Forest performing the best. The models' predictions for the test set were particularly accurate when OB ranged from 30% to 90%. After mono-substitution and double substitution of berberine and atenolol, the OB of both drugs was significantly improved. CONCLUSIONS This study found that some ADME properties of molecules do not have an absolute impact on OB. The database played a decisive role in the process of the machine learning OB prediction model, and the performance of the model was evaluated based on predictions within a range of strong generalization ability. In most cases, mono-substitution and double substitution were beneficial for enhancing the OB of berberine and atenolol. In summary, this study successfully constructed a machine learning regression prediction model that can accurately predict drug OB, which can guide drug design to achieve higher OB to some extent.
Collapse
Affiliation(s)
- Qi Yang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Lili Fan
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Key Laboratory of Traditional Chinese Medicine Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xiaotao Hou
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Key Laboratory of Traditional Chinese Medicine Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Key Laboratory of Traditional Chinese Medicine Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhongshang Xia
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Key Laboratory of Traditional Chinese Medicine Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Key Laboratory of Traditional Chinese Medicine Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning 530200, China; Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
11
|
Hurwitz SJ, De R, LeCher JC, Downs-Bowen JA, Goh SL, Zandi K, McBrayer T, Amblard F, Patel D, Kohler JJ, Bhasin M, Dobosh BS, Sukhatme V, Tirouvanziam RM, Schinazi RF. Why Certain Repurposed Drugs Are Unlikely to Be Effective Antivirals to Treat SARS-CoV-2 Infections. Viruses 2024; 16:651. [PMID: 38675992 PMCID: PMC11053489 DOI: 10.3390/v16040651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Most repurposed drugs have proved ineffective for treating COVID-19. We evaluated median effective and toxic concentrations (EC50, CC50) of 49 drugs, mostly from previous clinical trials, in Vero cells. Ratios of reported unbound peak plasma concentrations, (Cmax)/EC50, were used to predict the potential in vivo efficacy. The 20 drugs with the highest ratios were retested in human Calu-3 and Caco-2 cells, and their CC50 was determined in an expanded panel of cell lines. Many of the 20 drugs with the highest ratios were inactive in human Calu-3 and Caco-2 cells. Antivirals effective in controlled clinical trials had unbound Cmax/EC50 ≥ 6.8 in Calu-3 or Caco-2 cells. EC50 of nucleoside analogs were cell dependent. This approach and earlier availability of more relevant cultures could have reduced the number of unwarranted clinical trials.
Collapse
Affiliation(s)
- Selwyn J. Hurwitz
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Ramyani De
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Julia C. LeCher
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Jessica A. Downs-Bowen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Shu Ling Goh
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Keivan Zandi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Tamara McBrayer
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Franck Amblard
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Dharmeshkumar Patel
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - James J. Kohler
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Manoj Bhasin
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Brian S. Dobosh
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Vikas Sukhatme
- Morningside Center for Innovative and Affordable Medicine, Departments of Medicine and Hematology and Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Rabindra M. Tirouvanziam
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Raymond F. Schinazi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| |
Collapse
|
12
|
Maedera S, Mizuno T, Kusuhara H. Investigation of latent representation of toxicopathological images extracted by CNN model for understanding compound properties in vivo. Comput Biol Med 2024; 168:107748. [PMID: 38016375 DOI: 10.1016/j.compbiomed.2023.107748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/25/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
Toxicopathological images acquired during safety assessment elucidate an individual's biological responses to a given compound, and their numerization can yield valuable insights contributing to the assessment of compound properties. Currently, toxicopathological images are mainly encoded as pathological findings, evaluated by pathologists, which introduces challenges when used as input for modeling, specifically in terms of representation capability and comparability. In this study, we assessed the usefulness of latent representations extracted from toxicopathological images using Convolutional Neural Network (CNN) in estimating compound properties in vivo. Special emphasis was placed on examining the impact of learning pathological findings, the depth of frozen layers during learning, and the selection of the layer for latent representation. Our findings demonstrate that a machine learning model fed with the latent representation as input surpassed the performance of a model directly employing pathological findings as input, particularly in the classification of a compound's Mechanism of Action and in predicting late-phase findings from early-phase images in repeated-dose tests. While learning pathological findings did improve accuracy, the magnitude of improvement was relatively modest. Similarly, the effect of freezing layers during learning was also limited. Notably, the selection of the layer for latent representation had a substantial impact on the accurate estimation of compound properties in vivo.
Collapse
Affiliation(s)
- Shotaro Maedera
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, Japan
| | - Tadahaya Mizuno
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, Japan.
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, Japan
| |
Collapse
|
13
|
Kenchegowda M, Hani U, Al Fatease A, Haider N, Ramesh KVRNS, Talath S, Gangadharappa HV, Kiran Raj G, Padmanabha SH, Osmani RAM. Tiny titans- unravelling the potential of polysaccharides and proteins based dissolving microneedles in drug delivery and theranostics: A comprehensive review. Int J Biol Macromol 2023; 253:127172. [PMID: 37793514 DOI: 10.1016/j.ijbiomac.2023.127172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/06/2023]
Abstract
In recent years, microneedles (MNs) have emerged as a promising alternative to traditional drug delivery systems in transdermal drug delivery. The use of MNs has demonstrated significant potential in improving patient acceptance and convenience while avoiding the invasiveness of traditional injections. Dissolving, solid, hollow, coated, and hydrogel microneedles are among the various types studied for drug delivery. Dissolving microneedles (DMNs), in particular, have gained attention for their safety, painlessness, patient convenience, and high delivery efficiency. This comprehensive review primarily focuses on different types of microneedles, fabrication methods, and materials used in fabrication of DMNs such as hyaluronic acid, chitosan, alginate, gelatin, collagen, silk fibroin, albumin, cellulose and starch, to list a few. The review also provides an exhaustive discussion on the applications of DMNs, including the delivery of vaccines, cosmetic agents, contraceptives, hormone and genes, and other therapeutic applications like for treating cancer, skin diseases, and diabetes, among others, are covered in this review. Additionally, this review highlights some of the DMN systems that are presently undergoing clinical trials. Finally, the review discusses current advances and trends in DMNs, as well as future prospective directions for this ground-breaking technology in drug delivery.
Collapse
Affiliation(s)
- Madhuchandra Kenchegowda
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Nazima Haider
- Department of Pathology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - K V R N S Ramesh
- Department of Pharmaceutics, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Hosahalli V Gangadharappa
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - G Kiran Raj
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Sharath Honganoor Padmanabha
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India.
| |
Collapse
|
14
|
Esquea E, Ciraku L, Young RG, Merzy J, Talarico AN, Rashad AA, Cocklin S, Simone NL, Beld J, Reginato MJ, Dick A. Discovery of novel brain permeable human ACSS2 inhibitors for blocking breast cancer brain metastatic growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573073. [PMID: 38187734 PMCID: PMC10769402 DOI: 10.1101/2023.12.22.573073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Breast-cancer brain metastasis (BCBM) poses a significant clinical challenge, resulting in an end-stage diagnosis and hindered by limited therapeutic options. The blood-brain barrier (BBB) acts as an anatomical and physiological hurdle for therapeutic compounds, restricting the effective delivery of therapies to the brain. In order to grow and survive in a nutrient-poor environment, tumors in the brain must adapt to their metabolic needs, becoming highly dependent on acetate. These tumors rely on the conversion of acetate to acetyl-CoA by the enzyme Acetyl-CoA synthetase 2 (ACSS2), a key metabolic enzyme involved in regulating fatty acid synthesis and protein acetylation in tumor cells. ACSS2 has emerged as a crucial enzyme required for the growth of tumors in the brain. Here, we utilized a computational pipeline, combining pharmacophore-based shape screen methodology with ADME property predictions to identify novel brain-permeable ACSS2 inhibitors. From a small molecule library, this approach identified 30 potential ACSS2 binders, from which two candidates, AD-5584 and AD-8007, were validated for their binding affinity, predicted metabolic stability, and, notably, their ability to traverse the BBB. We show that treatment of BCBM cells, MDA-MB-231BR, with AD-5584 and AD-8007 leads to a significant reduction in lipid storage, reduction in colony formation, and increase in cell death in vitro . Utilizing an ex vivo orthotopic brain-slice tumor model, we show that treatment with AD-8007 and AD-5584 significantly reduces tumor size and synergizes with radiation in blocking BCBM tumor growth ex vivo. Importantly, we show that following intraperitoneal injections with AD-5584 and AD-8007, we can detect these compounds in the brain, confirming their BBB permeability. Thus, we have identified and validated novel ACSS2 inhibitor candidates for further drug development and optimization as agents for treating patients with breast cancer brain metastasis.
Collapse
|
15
|
Tay JH, Lim YH, Zheng M, Zhao Y, Tan WS, Xu C, Ramamurty U, Song J. Development of hyaluronic acid-silica composites via in situ precipitation for improved penetration efficiency in fast-dissolving microneedle systems. Acta Biomater 2023; 172:175-187. [PMID: 37865280 DOI: 10.1016/j.actbio.2023.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023]
Abstract
Fast-dissolving microneedles (DMNs) hold significant promise for transdermal drug delivery, offering improved patient compliance, biocompatibility, and functional adaptability for various therapeutic purposes. However, the mechanical strength of the biodegradable polymers used in DMNs often proves insufficient for effective penetration into human skin, especially under high humidity conditions. While many composite strategies have been developed to reinforce polymer-based DMNs, simple mixing of the reinforcements with polymers often results in ineffective penetration due to inhomogeneous dispersion of the reinforcements and the formation of undesired micropores. In response to this challenge, this study aimed to enhance the mechanical performance of hyaluronic acid (HA)-based microneedles (MNs), one of the most commonly used DMN systems. We introduced in situ precipitation of silica nanoparticles (Si) into the HA matrix in conjunction with conventional micromolding. The precipitated silica nanoparticles were uniformly distributed, forming an interconnected network within the HA matrix. Experimental results demonstrated that the mechanical properties of the HA-Si composite MNs with up to 20 vol% Si significantly improved, leading to higher penetration efficiency compared to pure HA MNs, while maintaining structural integrity without any critical defects. The composite MNs also showed reduced degradation rates and preserved their drug delivery capabilities and biocompatibility. Thus, the developed HA-Si composite MNs present a promising solution for efficient transdermal drug delivery and address the mechanical limitations inherent in DMN systems. STATEMENT OF SIGNIFICANCE: HA-Si composite dissolving microneedle (DMN) systems were successfully fabricated through in situ precipitation and conventional micromolding processes. The precipitated silica nanoparticles formed an interconnected network within the HA matrix, ranging in size from 25 to 230 nm. The optimal silica content for HA-Si composite MN systems should be up to 20 % by volume to maintain structural integrity and mechanical properties. HA-Si composite MNs with up to 20 % Si showed improved penetration efficiency and reduced degradation rates compared to pure HA MNs, thereby expanding the operational window. The HA-Si composite MNs retained good drug delivery capabilities and biocompatibility.
Collapse
Affiliation(s)
- Jie Hao Tay
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 639798, Singapore
| | - Yu Han Lim
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 639798, Singapore
| | - Mengjia Zheng
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Yakai Zhao
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 138634, Singapore
| | - Wen See Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 639798, Singapore; Singapore Centre for 3D Printing, Nanyang Technological University, Singapore, 639798, Singapore
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Upadrasta Ramamurty
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 138634, Singapore; Singapore Centre for 3D Printing, Nanyang Technological University, Singapore, 639798, Singapore; School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
| | - Juha Song
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 639798, Singapore; Singapore Centre for 3D Printing, Nanyang Technological University, Singapore, 639798, Singapore.
| |
Collapse
|
16
|
Benayad S, Wahnou H, El Kebbaj R, Liagre B, Sol V, Oudghiri M, Saad EM, Duval RE, Limami Y. The Promise of Piperine in Cancer Chemoprevention. Cancers (Basel) 2023; 15:5488. [PMID: 38001748 PMCID: PMC10670142 DOI: 10.3390/cancers15225488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer, characterized by the unregulated growth and dissemination of malignantly transformed cells, presents a significant global health challenge. The multistage process of cancer development involves intricate biochemical and genetic alterations within target cells. Cancer chemoprevention has emerged as a vital strategy to address this complex issue to mitigate cancer's impact on healthcare systems. This approach leverages pharmacologically active agents to block, suppress, prevent, or reverse invasive cancer development. Among these agents, piperine, an active alkaloid with a wide range of therapeutic properties, including antioxidant, anti-inflammatory, and immunomodulatory effects, has garnered attention for its potential in cancer prevention and treatment. This comprehensive review explores piperine's multifaceted role in inhibiting the molecular events and signaling pathways associated with various stages of cancer development, shedding light on its promising prospects as a versatile tool in cancer chemoprevention. Furthermore, the review will also delve into how piperine enhances the effectiveness of conventional treatments such as UV-phototherapy and TRAIL-based therapy, potentially synergizing with existing therapeutic modalities to provide more robust cancer management strategies. Finally, a crucial perspective of the long-term safety and potential side effects of piperine-based therapies and the need for clinical trials is also discussed.
Collapse
Affiliation(s)
- Salma Benayad
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (S.B.); (R.E.K.); (E.M.S.)
| | - Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20100, Morocco; (H.W.); (M.O.)
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (S.B.); (R.E.K.); (E.M.S.)
| | - Bertrand Liagre
- Le Laboratoire des Agroressources, Biomolécules et Chimie pour l’Innovation en Santé (LABCiS), University Limoges, UR 22722, F-87000 Limoges, France; (B.L.); (V.S.)
| | - Vincent Sol
- Le Laboratoire des Agroressources, Biomolécules et Chimie pour l’Innovation en Santé (LABCiS), University Limoges, UR 22722, F-87000 Limoges, France; (B.L.); (V.S.)
| | - Mounia Oudghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20100, Morocco; (H.W.); (M.O.)
| | - El Madani Saad
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (S.B.); (R.E.K.); (E.M.S.)
| | - Raphaël Emmanuel Duval
- The Franch Center for Scientific Research (CNRS), Université de Lorraine, L2CM, F-54000 Nancy, France
| | - Youness Limami
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco; (S.B.); (R.E.K.); (E.M.S.)
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20100, Morocco; (H.W.); (M.O.)
| |
Collapse
|
17
|
Kus M, Ibragimow I, Piotrowska-Kempisty H. Caco-2 Cell Line Standardization with Pharmaceutical Requirements and In Vitro Model Suitability for Permeability Assays. Pharmaceutics 2023; 15:2523. [PMID: 38004503 PMCID: PMC10674574 DOI: 10.3390/pharmaceutics15112523] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
The Caco-2 cell line derived from human colon carcinoma is commonly used to assess the permeability of compounds in in vitro conditions. Due to the significant increase in permeability studies using the Caco-2 cell line in recent years, the need to standardize this biological model seems necessary. The pharmaceutical requirements define only the acceptance criteria for the validation of the Caco-2 cell line and do not specify the protocol for its implementation. Therefore, the aim of this study is to review the conditions for permeability studies across the Caco-2 monolayer reported in the available literature concerning validation guidelines. We summarized the main aspects affecting the validation process of the Caco-2 cell line, including the culture conditions, cytotoxicity, cell differentiation process, and monolayer transport conditions, and the main conclusions may be useful in developing individual methods for preparing the cell line for validation purposes and further permeability research.
Collapse
Affiliation(s)
- Marta Kus
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
- Research and Development Department of Ethifarm, Ethifarm Manufacturing Plant, 9 Stefana Zeromskiego St., 60-544 Poznan, Poland;
| | - Izabela Ibragimow
- Research and Development Department of Ethifarm, Ethifarm Manufacturing Plant, 9 Stefana Zeromskiego St., 60-544 Poznan, Poland;
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
- Department of Basic and Preclinical Science, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 7 Gagarina St., 87-100 Torun, Poland
| |
Collapse
|
18
|
Pereira CDS, Cruz JN, Ferreira MKM, Baia-da-Silva DC, Fontes-Junior EA, Lima RR. Global Research Trends and Hotspots Analysis of the Scientific Production of Amitriptyline: A Bibliometric Approach. Pharmaceuticals (Basel) 2023; 16:1047. [PMID: 37513958 PMCID: PMC10386017 DOI: 10.3390/ph16071047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Amitriptyline was first introduced as a medication to treat depression. Over time, this substance has been used to treat other conditions, such as gastrointestinal disorders, fibromyalgia, neuropathic pain, and analgesia, among others. However, there are no published studies that provide a broad view of the possible motivations that have led to changes in the use of amitriptyline. In this study, we have identified the landscape of use for amitriptyline based on knowledge mapping of the 100 most-cited articles about this drug. We searched Web of Science Core Collection without time and language restrictions. We obtained 14,446 results, but we only used the 100 most-cited articles that had amitriptyline as the object of study. We collected the following information from each article: authors, country of the corresponding authors, year of publication, citation count, citation density (number of citations per year), and keywords. In addition, we seek to map in the chosen articles study design and research findings. We found that since 1980, the use of amitriptyline has expanded beyond depression, moving to off-label use to treat a variety of diseases and conditions, including post-herpetic neuralgia, neuropathic pain, primary fibrosis, fibromyalgia, and migraine, can be considered a drug with more clinical applicability than its original clinical indication.
Collapse
Affiliation(s)
- Cristian Dos Santos Pereira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University do Pará, Belém 66075-110, Brazil
| | - Jorddy Neves Cruz
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University do Pará, Belém 66075-110, Brazil
| | - Maria Karolina Martins Ferreira
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University do Pará, Belém 66075-110, Brazil
| | - Daiane Claydes Baia-da-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University do Pará, Belém 66075-110, Brazil
| | - Eneas Andrade Fontes-Junior
- Laboratory of Pharmacology of Inflammation and Behavior, Federal University of Pará, Belém 66075-110, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University do Pará, Belém 66075-110, Brazil
| |
Collapse
|
19
|
Boutin JA, Kennaway DJ, Jockers R. Melatonin: Facts, Extrapolations and Clinical Trials. Biomolecules 2023; 13:943. [PMID: 37371523 DOI: 10.3390/biom13060943] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Melatonin is a fascinating molecule that has captured the imagination of many scientists since its discovery in 1958. In recent times, the focus has changed from investigating its natural role as a transducer of biological time for physiological systems to hypothesized roles in virtually all clinical conditions. This goes along with the appearance of extensive literature claiming the (generally) positive benefits of high doses of melatonin in animal models and various clinical situations that would not be receptor-mediated. Based on the assumption that melatonin is safe, high doses have been administered to patients, including the elderly and children, in clinical trials. In this review, we critically review the corresponding literature, including the hypotheses that melatonin acts as a scavenger molecule, in particular in mitochondria, by trying not only to contextualize these interests but also by attempting to separate the wheat from the chaff (or the wishful thinking from the facts). We conclude that most claims remain hypotheses and that the experimental evidence used to promote them is limited and sometimes flawed. Our review will hopefully encourage clinical researchers to reflect on what melatonin can and cannot do and help move the field forward on a solid basis.
Collapse
Affiliation(s)
- J A Boutin
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, University of Normandy, INSERM U1239, 76000 Rouen, France
| | - D J Kennaway
- Robinson Research Institute and Adelaide School of Medicine, University of Adelaide, Adelaide Health and Medical Science Building, North Terrace, Adelaide, SA 5006, Australia
| | - R Jockers
- Institut Cochin, Université Paris Cité, INSERM, CNRS, 75014 Paris, France
| |
Collapse
|
20
|
Singh V, Dziwornu GA, Chibale K. The implication of Mycobacterium tuberculosis-mediated metabolism of targeted xenobiotics. Nat Rev Chem 2023; 7:340-354. [PMID: 37117810 PMCID: PMC10026799 DOI: 10.1038/s41570-023-00472-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 03/29/2023]
Abstract
Drug metabolism is generally associated with liver enzymes. However, in the case of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), Mtb-mediated drug metabolism plays a significant role in treatment outcomes. Mtb is equipped with enzymes that catalyse biotransformation reactions on xenobiotics with consequences either in its favour or as a hindrance by deactivating or activating chemical entities, respectively. Considering the range of chemical reactions involved in the biosynthetic pathways of Mtb, information related to the biotransformation of antitubercular compounds would provide opportunities for the development of new chemical tools to study successful TB infections while also highlighting potential areas for drug discovery, host-directed therapy, dose optimization and elucidation of mechanisms of action. In this Review, we discuss Mtb-mediated biotransformations and propose a holistic approach to address drug metabolism in TB drug discovery and related areas. ![]()
Mycobacterium tuberculosis-mediated metabolism of xenobiotics poses an important research question for antitubercular drug discovery. Identification of the metabolic fate of compounds can inform requisite structure–activity relationship strategies early on in a drug discovery programme towards improving the properties of the compound.
Collapse
Affiliation(s)
- Vinayak Singh
- grid.7836.a0000 0004 1937 1151Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- grid.7836.a0000 0004 1937 1151South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, South Africa
- grid.7836.a0000 0004 1937 1151Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | - Godwin Akpeko Dziwornu
- grid.7836.a0000 0004 1937 1151Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
| | - Kelly Chibale
- grid.7836.a0000 0004 1937 1151Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- grid.7836.a0000 0004 1937 1151South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, South Africa
- grid.7836.a0000 0004 1937 1151Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
- grid.7836.a0000 0004 1937 1151Department of Chemistry, University of Cape Town, Rondebosch, South Africa
| |
Collapse
|
21
|
Marcello E, Chiono V. Biomaterials-Enhanced Intranasal Delivery of Drugs as a Direct Route for Brain Targeting. Int J Mol Sci 2023; 24:ijms24043390. [PMID: 36834804 PMCID: PMC9964911 DOI: 10.3390/ijms24043390] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/22/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Intranasal (IN) drug delivery is a non-invasive and effective route for the administration of drugs to the brain at pharmacologically relevant concentrations, bypassing the blood-brain barrier (BBB) and minimizing adverse side effects. IN drug delivery can be particularly promising for the treatment of neurodegenerative diseases. The drug delivery mechanism involves the initial drug penetration through the nasal epithelial barrier, followed by drug diffusion in the perivascular or perineural spaces along the olfactory or trigeminal nerves, and final extracellular diffusion throughout the brain. A part of the drug may be lost by drainage through the lymphatic system, while a part may even enter the systemic circulation and reach the brain by crossing the BBB. Alternatively, drugs can be directly transported to the brain by axons of the olfactory nerve. To improve the effectiveness of drug delivery to the brain by the IN route, various types of nanocarriers and hydrogels and their combinations have been proposed. This review paper analyzes the main biomaterials-based strategies to enhance IN drug delivery to the brain, outlining unsolved challenges and proposing ways to address them.
Collapse
Affiliation(s)
- Elena Marcello
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research, Centro 3R, 56122 Pisa, Italy
- Institute for Chemical-Physical Processes, National Research Council (CNR-IPCF), 56124 Pisa, Italy
- Correspondence:
| |
Collapse
|
22
|
Bakulina NV, Tikhonov SV, Okovityi SV, Lutaenko EA, Bolshakov AO, Prikhodko VA, Nekrasova AS. [Pharmacokinetics and pharmacodynamics of rebamipide. New possibilities of therapy: A review]. TERAPEVT ARKH 2023; 94:1431-1437. [PMID: 37167190 DOI: 10.26442/00403660.2022.12.202000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
The MedLine database contains 570 publications, including 71 randomized clinical trials and 6 meta-analyses on the rebamipide molecule in 2022. Indications for the use of rebamipide are gastric ulcer, chronic gastritis with hyperacidityin the acute stage, erosive gastritis, prevention of damage to the gastrointestinal mucosa while taking non-steroidal anti-inflammatory drugs, eradication of Helicobacter pylori. Currently trials are studying the efficacy and safety of the drug in gouty and rheumatoid arthritis, osteoarthritis, Sjögren's syndrome, bronchial asthma, vitiligo, atherosclerosis, diseases of the kidneys and liver; using in traumatology to accelerate bone regeneration; in ophthalmology to improve the regeneration of corneal epithelium; in oncology to reduce inflammatory changes in the oral mucosa after chemoradiotherapy. The review article is about the main pharmacokinetic and pharmacodynamic characteristics of rebamipide. A detailed understanding of pharmacodynamics and pharmacokinetics allows for individual selection of therapy based on the characteristics of the patient's body - gender, age, comorbidities; choose the optimal route of administration and dosing regimen; predict adverse effects and drug interactions; be determined with new clinical indications.
Collapse
Affiliation(s)
- N V Bakulina
- Mechnikov North-Western State Medical University
| | - S V Tikhonov
- Mechnikov North-Western State Medical University
| | - S V Okovityi
- Saint Petersburg State Chemical Pharmaceutical University
| | - E A Lutaenko
- Mechnikov North-Western State Medical University
| | | | - V A Prikhodko
- Saint Petersburg State Chemical Pharmaceutical University
| | | |
Collapse
|
23
|
Flexible polymeric patch based nanotherapeutics against non-cancer therapy. Bioact Mater 2022; 18:471-491. [PMID: 35415299 PMCID: PMC8971585 DOI: 10.1016/j.bioactmat.2022.03.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/16/2022] Open
Abstract
Flexible polymeric patches find widespread applications in biomedicine because of their biological and tunable features including excellent patient compliance, superior biocompatibility and biodegradation, as well as high loading capability and permeability of drug. Such polymeric patches are classified into microneedles (MNs), hydrogel, microcapsule, microsphere and fiber depending on the formed morphology. The combination of nanomaterials with polymeric patches allows for improved advantages of increased curative efficacy and lowered systemic toxicity, promoting on-demand and regulated drug administration, thus providing the great potential to their clinic translation. In this review, the category of flexible polymeric patches that are utilized to integrate with nanomaterials is briefly presented and their advantages in bioapplications are further discussed. The applications of nanomaterials embedded polymeric patches in non-cancerous diseases were also systematically reviewed, including diabetes therapy, wound healing, dermatological disease therapy, bone regeneration, cardiac repair, hair repair, obesity therapy and some immune disease therapy. Alternatively, the limitations, latest challenges and future perspectives of such biomedical therapeutic devices are addressed. The most explored polymeric patches, such as microneedle, hydrogel, microsphere, microcapsule, and fiber are summarized. Polymeric patches integrated with a diversity of nanomaterials are systematically overviewed in non-cancer therapy. The future prospective for the development of polymeric patch based nanotherapeutics is discussed.
Collapse
|
24
|
Liu X, Hou S, Xiang R, Hu C, Chen Z, Li N, Yan H, Yu X, Li X, Chi Y, Yang J. Imipramine activates FAM3A-FOXA2-CPT2 pathway to ameliorate hepatic steatosis. Metabolism 2022; 136:155292. [PMID: 35995281 DOI: 10.1016/j.metabol.2022.155292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/19/2022] [Accepted: 08/12/2022] [Indexed: 10/31/2022]
Abstract
Mitochondrial FAM3A has been revealed to be a viable target for treating diabetes and nonalcoholic fatty liver disease (NAFLD). However, its distinct mechanism in ameliorating hepatic steatosis remained unrevealed. High-throughput RNA sequencing revealed that carnitine palmityl transferase 2 (CPT2), one of the key enzymes for lipid oxidation, is the downstream molecule of FAM3A signaling pathway in hepatocytes. Intensive study demonstrated that FAM3A-induced ATP release activated P2 receptor to promote the translocation of calmodulin (CaM) from cytoplasm into nucleus, where it functioned as a co-activator of forkhead box protein A2 (FOXA2) to promote the transcription of CPT2, increasing free fatty acid oxidation and reducing lipid deposition in hepatocytes. Furthermore, antidepressant imipramine activated FAM3A-ATP-P2 receptor-CaM-FOXA2-CPT2 pathway to reduce lipid deposition in hepatocytes. In FAM3A-deficient hepatocytes, imipramine failed to activate CaM-FOXA2-CPT2 axis to increase lipid oxidation. Imipramine administration significantly ameliorated hepatic steatosis, hyperglycemia and obesity of obese mice mainly by activating FAM3A-ATP-CaM-FOXA2-CPT2 pathway in liver and thermogenesis in brown adipose tissue (BAT). In FAM3A-deficient mice fed on high-fat-diet, imipramine treatment failed to correct the dysregulated lipid and glucose metabolism, and activate thermogenesis in BAT. In conclusion, imipramine activates FAM3A-ATP-CaM-FOXA2-CPT2 pathway to ameliorate steatosis. For depressive patients complicated with metabolic disorders, imipramine may be recommended in priority as antidepressive drug.
Collapse
Affiliation(s)
- Xiangyang Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Song Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Rui Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Chengqing Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Zhenzhen Chen
- Hypertension Center, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Na Li
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China
| | - Han Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoxing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Xin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Yujing Chi
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China.
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
25
|
Kimura S, Ichikawa M, Sugawara S, Katagiri T, Hirasawa Y, Ishikawa T, Matsunaga W, Gotoh A. Nicotinamide Mononucleotide Is Safely Metabolized and Significantly Reduces Blood Triglyceride Levels in Healthy Individuals. Cureus 2022; 14:e28812. [PMID: 36225528 PMCID: PMC9534732 DOI: 10.7759/cureus.28812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2022] [Indexed: 11/05/2022] Open
|
26
|
Pace J, Ivich F, Marple E, Niedre M. Near-infrared diffuse in vivo flow cytometry. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220101GR. [PMID: 36114606 PMCID: PMC9478904 DOI: 10.1117/1.jbo.27.9.097002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/07/2022] [Indexed: 06/15/2023]
Abstract
Significance Diffuse in vivo flow cytometry (DiFC) is an emerging technique for enumerating rare fluorescently labeled circulating cells noninvasively in the bloodstream. Thus far, we have reported red and blue-green versions of DiFC. Use of near-infrared (NIR) fluorescent light would in principle allow use of DiFC in deeper tissues and would be compatible with emerging NIR fluorescence molecular contrast agents. Aim We describe the design of a NIR-DiFC instrument and demonstrate its use in optical flow phantoms in vitro and in mice in vivo. Approach We developed an improved optical fiber probe design for efficient collection of fluorescence from individual circulating cells and efficient rejection of instrument autofluorescence. We built a NIR-DiFC instrument. We tested this with NIR fluorescent microspheres and cell lines labeled with OTL38 fluorescence contrast agent in a flow phantom model. We also tested NIR-DiFC in nude mice injected intravenously with OTL38-labeled L1210A cells. Results NIR-DiFC allowed detection of circulating tumor cells (CTCs) in flow phantoms with mean signal-to-noise ratios (SNRs) of 19 to 32 dB. In mice, fluorescently labeled CTCs were detectable with mean SNR of 26 dB. NIR-DiFC also exhibited orders significantly lower autofluorescence and false-alarm rates than blue-green DiFC. Conclusions NIR-DiFC allows use of emerging NIR contrast agents. Our work could pave the way for future use of NIR-DiFC in humans.
Collapse
Affiliation(s)
- Joshua Pace
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Fernando Ivich
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Eric Marple
- EmVision LLC, Loxahatchee, Florida, United States
| | - Mark Niedre
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| |
Collapse
|
27
|
Singh K, Nawabjan SA, Zhang L, El-Nezami H, Annapureddy RR, Chow BKC. Discovery of small-molecule modulators of the secretin receptor: Purmorphamine as novel anti-hypertensive agent. Eur J Med Chem 2022; 242:114642. [DOI: 10.1016/j.ejmech.2022.114642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/17/2022] [Accepted: 07/27/2022] [Indexed: 11/04/2022]
|
28
|
Jackstadt MM, Chamberlain CA, Doonan SR, Shriver LP, Patti GJ. A multidimensional metabolomics workflow to image biodistribution and evaluate pharmacodynamics in adult zebrafish. Dis Model Mech 2022; 15:dmm049550. [PMID: 35972155 PMCID: PMC9411795 DOI: 10.1242/dmm.049550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/13/2022] [Indexed: 12/16/2022] Open
Abstract
An integrated evaluation of the tissue distribution and pharmacodynamic properties of a therapeutic is essential for successful translation to the clinic. To date, however, cost-effective methods to measure these parameters at the systems level in model organisms are lacking. Here, we introduce a multidimensional workflow to evaluate drug activity that combines mass spectrometry-based imaging, absolute drug quantitation across different biological matrices, in vivo isotope tracing and global metabolome analysis in the adult zebrafish. As a proof of concept, we quantitatively determined the whole-body distribution of the anti-rheumatic agent hydroxychloroquine sulfate (HCQ) and measured the systemic metabolic impacts of drug treatment. We found that HCQ distributed to most organs in the adult zebrafish 24 h after addition of the drug to water, with the highest accumulation of both the drug and its metabolites being in the liver, intestine and kidney. Interestingly, HCQ treatment induced organ-specific alterations in metabolism. In the brain, for example, HCQ uniquely elevated pyruvate carboxylase activity to support increased synthesis of the neuronal metabolite, N-acetylaspartate. Taken together, this work validates a multidimensional metabolomics platform for evaluating the mode of action of a drug and its potential off-target effects in the adult zebrafish. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Madelyn M. Jackstadt
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Casey A. Chamberlain
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Steven R. Doonan
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Leah P. Shriver
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Gary J. Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO 63130, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
29
|
Lee JH, Kuhar S, Seo JH, Pasricha PJ, Mittal R. Computational modeling of drug dissolution in the human stomach: Effects of posture and gastroparesis on drug bioavailability. PHYSICS OF FLUIDS (WOODBURY, N.Y. : 1994) 2022; 34:081904. [PMID: 35971381 PMCID: PMC9372820 DOI: 10.1063/5.0096877] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/23/2022] [Indexed: 05/25/2023]
Abstract
The oral route is the most common choice for drug administration because of several advantages, such as convenience, low cost, and high patient compliance, and the demand and investment in research and development for oral drugs continue to grow. The rate of dissolution and gastric emptying of the dissolved active pharmaceutical ingredient (API) into the duodenum is modulated by gastric motility, physical properties of the pill, and the contents of the stomach, but current in vitro procedures for assessing dissolution of oral drugs are limited in their ability to recapitulate this process. This is particularly relevant for disease conditions, such as gastroparesis, that alter the anatomy and/or physiology of the stomach. In silico models of gastric biomechanics offer the potential for overcoming these limitations of existing methods. In the current study, we employ a biomimetic in silico simulator based on the realistic anatomy and morphology of the stomach (referred to as "StomachSim") to investigate and quantify the effect of body posture and stomach motility on drug bioavailability. The simulations show that changes in posture can potentially have a significant (up to 83%) effect on the emptying rate of the API into the duodenum. Similarly, a reduction in antral contractility associated with gastroparesis can also be found to significantly reduce the dissolution of the pill as well as emptying of the API into the duodenum. The simulations show that for an equivalent motility index, the reduction in gastric emptying due to neuropathic gastroparesis is larger by a factor of about five compared to myopathic gastroparesis.
Collapse
Affiliation(s)
| | - S. Kuhar
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | | | - P. J. Pasricha
- Division of Gastroenterology and Hepatology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| | - R. Mittal
- Author to whom correspondence should be addressed:
| |
Collapse
|
30
|
The Molecular Mechanism of Traditional Chinese Medicine Prescription: Gu-tong Formula in Relieving Osteolytic Bone Destruction. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4931368. [PMID: 35872837 PMCID: PMC9300326 DOI: 10.1155/2022/4931368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/20/2022] [Indexed: 01/01/2023]
Abstract
Bone metastasis is a common complication in patients with advanced tumors, causing pain and bone destruction and affecting their quality of life. Typically, complementary and alternative medicine (CAM), with unique theoretical guidance, has played key roles in the treatment of tumor-related diseases. Gu-tong formula (GTF), as a representative prescription of traditional Chinese medicine, has been demonstrated to be an effective clinical medication for the relief of cancer pain. However, the molecular mechanism of GTF in the treatment of osteolytic metastasis is still unclear. Herein, we employ network pharmacology and molecular dynamics methods to uncover the potential treatment mechanism, indicating that GTF can reduce the levels of serum IL6 and TGFB1 and thus limit the scope of bone cortical damage. Among the active compounds, sesamin and deltoin can bind stably with IL6 and TGFB1, respectively, and have the potential to become anti-inflammatory and anticancer drugs. Although the reasons for the therapeutic effect of GTF are complex and comprehensive, this work provides biological plausibility in the treatment of osteolytic metastases, which has a guiding significance for the treatment of cancer pain with CAM.
Collapse
|
31
|
Kiesel BF, Deppas JJ, Guo J, Parise RA, Clump DA, Bakkenist CJ, Beumer JH. Dose-dependent bioavailability, absorption-rate limited elimination, and tissue distribution of the ATR inhibitor BAY-1895344 (elimusertib) in mice. Cancer Chemother Pharmacol 2022; 89:795-807. [PMID: 35507041 PMCID: PMC10082586 DOI: 10.1007/s00280-022-04436-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Ataxia Telangiectasia and Rad3-related (ATR) is a pivotal component of the DNA damage response and repair pathways that is activated in responses to cytotoxic cancer treatments. Several ATR inhibitors (ATRi) are in development that block the ATR mediated DNA repair and enhance the damage associated with cytotoxic therapy. BAY-1895344 (elimusertib) is an orally available ATRi with preclinical efficacy that is in clinical development. Little is known about the pharmacokinetics (PK) which is of interest, because tissue exposure and ATR inhibition may relate to toxicities or responses. METHODS To evaluate BAY-1895344 PK, a sensitive LC-MS/MS method was utilized for quantitation in mouse plasma and tissues. PK studies in mice were first conducted to determine dose linearity. In vivo metabolites were identified and analyzed semi-quantitatively. A compartmental PK model was developed to describe PK behavior. An extensive PK study was then conducted in tumor-bearing mice to quantitate tissue distribution for relevant tissues. RESULTS Dose linearity was observed from 1 to 10 mg/kg PO, while at 40 mg/kg PO bioavailability increased approximately fourfold due to saturation of first-pass metabolism, as suggested by metabolite analyses and a developed compartmental model. Longer half-lives in PO treated mice compared to IV treated mice indicated absorption-rate limited elimination. Tissue distribution varied but showed extensive distribution to bone marrow, brain, and spinal cord. CONCLUSIONS Complex PK behavior was limited to absorption processes which may not be recapitulated clinically. Tissue partition coefficients may be used to contrast ATR inhibitors with respect to their efficacy and toxicity.
Collapse
Affiliation(s)
- Brian F Kiesel
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Room G27e, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joshua J Deppas
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Room G27e, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianxia Guo
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Room G27e, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Robert A Parise
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Room G27e, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
| | - David A Clump
- Department of Radiation Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher J Bakkenist
- Department of Radiation Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jan H Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Room G27e, 5117 Centre Ave, Pittsburgh, PA, 15213, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
Umfress A, Singh S, Ryan KJ, Chakraborti A, Plattner F, Sonawane Y, Mallareddy JR, Acosta EP, Natarajan A, Bibb JA. Systemic Administration of a Brain Permeable Cdk5 Inhibitor Alters Neurobehavior. Front Pharmacol 2022; 13:863762. [PMID: 35645825 PMCID: PMC9134315 DOI: 10.3389/fphar.2022.863762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/01/2022] [Indexed: 01/09/2023] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a crucial regulator of neuronal signal transduction. Cdk5 activity is implicated in various neuropsychiatric and neurodegenerative conditions such as stress, anxiety, depression, addiction, Alzheimer's disease, and Parkinson's disease. While constitutive Cdk5 knockout is perinatally lethal, conditional knockout mice display resilience to stress-induction, enhanced cognition, neuroprotection from stroke and head trauma, and ameliorated neurodegeneration. Thus, Cdk5 represents a prime target for treatment in a spectrum of neurological and neuropsychiatric conditions. While intracranial infusions or treatment of acutely dissected brain tissue with compounds that inhibit Cdk5 have allowed the study of kinase function and corroborated conditional knockout findings, potent brain-penetrant systemically deliverable Cdk5 inhibitors are extremely limited, and no Cdk5 inhibitor has been approved to treat any neuropsychiatric or degenerative diseases to date. Here, we screened aminopyrazole-based analogs as potential Cdk5 inhibitors and identified a novel analog, 25-106, as a uniquely brain-penetrant anti-Cdk5 drug. We characterize the pharmacokinetic and dynamic responses of 25-106 in mice and functionally validate the effects of Cdk5 inhibition on open field and tail-suspension behaviors. Altogether, 25-106 represents a promising preclinical Cdk5 inhibitor that can be systemically administered with significant potential as a neurological/neuropsychiatric therapeutic.
Collapse
Affiliation(s)
- Alan Umfress
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sarbjit Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kevin J. Ryan
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ayanabha Chakraborti
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Yogesh Sonawane
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jayapal Reddy Mallareddy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - Edward P. Acosta
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, United States
| | - James A. Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
- Departments of Neurobiology and Neurology, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
33
|
Wu J, Sahoo JK, Li Y, Xu Q, Kaplan DL. Challenges in delivering therapeutic peptides and proteins: A silk-based solution. J Control Release 2022; 345:176-189. [PMID: 35157939 PMCID: PMC9133086 DOI: 10.1016/j.jconrel.2022.02.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023]
Abstract
Peptide- and protein-based therapeutics have drawn significant attention over the past few decades for the treatment of infectious diseases, genetic disorders, oncology, and many other clinical needs. Yet, protecting peptide- and protein-based drugs from degradation and denaturation during processing, storage and delivery remain significant challenges. In this review, we introduce the properties of peptide- and protein-based drugs and the challenges associated with their stability and delivery. Then, we discuss delivery strategies using synthetic polymers and their advantages and limitations. This is followed by a focus on silk protein-based materials for peptide/protein drug processing, storage, and delivery, as a path to overcome stability and delivery challenges with current systems.
Collapse
Affiliation(s)
- Junqi Wu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
34
|
Abstract
Exosomes are natural nanoparticles that originate in the endocytic system. Exosomes play an important role in cell-to-cell communication by transferring RNAs, lipids, and proteins from donor cells to recipient cells or by binding to receptors on the recipient cell surface. The concentration of exosomes and the diversity of cargos are high in milk. Exosomes and their cargos resist degradation in the gastrointestinal tract and during processing of milk in dairy plants. They are absorbed and accumulate in tissues following oral administrations, cross the blood-brain barrier, and dietary depletion and supplementation elicit phenotypes. These features have sparked the interest of the nutrition and pharmacology communities for exploring milk exosomes as novel bioactive food compounds and for delivering drugs to diseased tissues. This review discusses the current knowledgebase, uncertainties, and controversies in these lines of scholarly endeavor and health research.
Collapse
Affiliation(s)
- Alice Ngu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Shu Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Haichuan Wang
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Afsana Khanam
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
35
|
Kalenga CZ, Dumanski SM, Metcalfe A, Robert M, Nerenberg KA, MacRae JM, Premji Z, Ahmed SB. The effect of non-oral hormonal contraceptives on hypertension and blood pressure: A systematic review and meta-analysis. Physiol Rep 2022; 10:e15267. [PMID: 35510324 PMCID: PMC9069167 DOI: 10.14814/phy2.15267] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 06/14/2023] Open
Abstract
Oral contraceptives (OC) are associated with increased risk of hypertension and elevated blood pressure (BP). Whether non-oral hormonal contraceptives have similar associations is unknown. We sought to investigate the effect of non-oral hormonal contraceptive (NOHC) use on the risk of hypertension and changes in BP, compared to non-hormonal contraceptive and OC use. We searched bibliographic databases (MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials) until August 2020. Studies reporting risk of hypertension or changes in systolic and diastolic BP with NOHC use compared with either non-hormonal contraceptive or OC use. Abstract screening, full-text review, data extraction, and quality assessment were completed in duplicate. For studies reporting dichotomous outcomes, we reported results as relative risk with 95% confidence intervals (CI). A random-effects model was used to estimate pooled weighted mean difference and 95% CI of change in BP. Twenty-five studies were included. A lower incidence of hypertension was observed with injectable contraceptive use compared to non-hormonal contraceptive and OC use, although it was unclear if this was statistically significant. Compared to non-hormonal contraceptive use, injectable contraceptive use was associated with increased BP (SBP: 3.24 mmHg, 95%CI 2.49 to 3.98 mmHg; DBP: 3.15 mmHg, 95%CI 0.09 to 6.20 mmHg), the hormonal intra-uterine device use was associated with reduced BP (SBP: -4.50 mmHg, 95%CI -8.44 to -0.57 mmHg; DBP: -7.48 mmHg, 95% -14.90 to -0.05 mmHg), and the vaginal ring was associated with reduced diastolic BP (-3.90 mmHg, 95%CI -6.67 to -1.13 mmHg). Compared to OC use, the injectable contraceptive use was associated with increased diastolic BP (2.38 mmHg, 95%CI 0.39 to 4.38 mmHg). NOHC use is associated with changes in BP which differ by type and route of administration. Given the strong association between incremental increases in BP and cardiovascular risk, prospective studies are required.
Collapse
Affiliation(s)
- Cindy Z. Kalenga
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Libin Cardiovascular InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | - Sandra M. Dumanski
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Libin Cardiovascular InstituteUniversity of CalgaryCalgaryAlbertaCanada
- Alberta Kidney Disease NetworkCalgaryAlbertaCanada
| | - Amy Metcalfe
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Libin Cardiovascular InstituteUniversity of CalgaryCalgaryAlbertaCanada
- Alberta Children's Hospital Research InstituteCalgaryAlbertaCanada
| | - Magali Robert
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Kara A. Nerenberg
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Libin Cardiovascular InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | - Jennifer M. MacRae
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Libin Cardiovascular InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | - Zahra Premji
- University of VictoriaVictoriaBritish ColumbiaCanada
| | - Sofia B. Ahmed
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Libin Cardiovascular InstituteUniversity of CalgaryCalgaryAlbertaCanada
- Alberta Kidney Disease NetworkCalgaryAlbertaCanada
| |
Collapse
|
36
|
Zhang H, Cui Y, Zhang X, Yuan X, Xu D, Zhang L. Sustained delivery of salbutamol from cubosomal gel for management of pediatric asthma: In vitro and in vivo evaluation. J Microencapsul 2022; 39:252-260. [PMID: 35384781 DOI: 10.1080/02652048.2022.2060362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIM In the current study, efforts are being made to formulate transdermal salbutamol-cubosomal gel to manage pediatric asthma. METHODS Salbutamol-loaded cubosomal gels were prepared by melt emulsification and sonication. The cubosomal gels were characterized by morphology, particle size, zeta potential, entrapment efficacy, assay, viscosity, and texture profiles. Ex vivo permeation and pharmacokinetic studies were performed using rats. RESULTS The mean cubosomal particle size (208-361 ± 12.5-32.5 nm), PDI (0.06-0.11 ± 0.01-0.02), viscosity (8527-9019 cp), and entrapment efficacy (76.3-91.0% w/w) increase with the level of monoolein. The ex vivo permeation study showed a biphasic release pattern, with salbutamol cleared from control gel within 8 h, while cubosomal gels showed sustained release up to 72 h. The pharmacokinetic profiles in the rat model showed 8.62-fold higher bioavailability with cubosomal gel. CONCLUSION The study demonstrated the potential of cubosomal nanoparticle-laden gel to sustain the release of salbutamol to treat pediatric asthma.
Collapse
Affiliation(s)
- Huifang Zhang
- Department of Pediatrics, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, 230001, China
| | - Yanjie Cui
- Department of Pediatrics, Dongda Hospital of Shanxian County, Heze, Shandong, 274300, China
| | - Xiaochun Zhang
- Department of Pediatrics, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Xunling Yuan
- Department of Pediatrics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, 150036, China
| | - Dandan Xu
- Department of Pediatrics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, 150036, China
| | - Lei Zhang
- Department of Pediatrics, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, 150036, China
| |
Collapse
|
37
|
Uluc K, Neuwelt EA, Ambady P. Advances in Intraarterial Chemotherapy Delivery Strategies and Blood-Brain Barrier Disruption. Neurosurg Clin N Am 2022; 33:219-223. [PMID: 35346454 DOI: 10.1016/j.nec.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Chemotherapeutics play a significant role in the management of most brain tumors. First pass effect, systemic toxicity, and more importantly, the blood-brain barrier pose significant challenges to the success of chemotherapy. Over the last 80 years, different techniques of intraarterial chemotherapy delivery have been performed in many studies but failed to become standard of care. The purpose of this article is to review the history of intraarterial drug delivery and osmotic blood-brain barrier disruption, identify the challenges for clinical translation, and identify future directions for these approaches.
Collapse
Affiliation(s)
- Kutluay Uluc
- Neurosurgery, Northernlight Eastern Maine Medical Center, Bangor, ME, USA
| | - Edward A Neuwelt
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Neurosurgery, Oregon Health & Science University, Portland, OR, USA; Portland Veterans Affairs Medical Center, Portland, OR, USA
| | - Prakash Ambady
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
38
|
A Nanoparticle's Journey to the Tumor: Strategies to Overcome First-Pass Metabolism and Their Limitations. Cancers (Basel) 2022; 14:cancers14071741. [PMID: 35406513 PMCID: PMC8996837 DOI: 10.3390/cancers14071741] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Traditional cancer therapeutics suffer from off-target toxicity, limiting their effective dose and preventing patients’ tumors from being sufficiently treated by chemotherapeutics alone. Nanomedicine is an emerging class of therapeutics in which a drug is packaged into a nanoparticle that promotes uptake of the drug at a tumor site, shielding it from uptake by peripheral organs and enabling the safe delivery of chemotherapeutics that have poor aqueous solubility, short plasma half-life, narrow therapeutic window, and toxic side effects. Despite the advantages of nanomedicines for cancer, there remains significant challenges to improve uptake at the tumor and prevent premature clearance from the body. In this review, we summarize the effects of first-pass metabolism on a nanoparticle’s journey to a tumor and outline future steps that we believe will improve the efficacy of cancer nanomedicines. Abstract Nanomedicines represent the cutting edge of today’s cancer therapeutics. Seminal research decades ago has begun to pay dividends in the clinic, allowing for the delivery of cancer drugs with enhanced systemic circulation while also minimizing off-target toxicity. Despite the advantages of delivering cancer drugs using nanoparticles, micelles, or other nanostructures, only a small fraction of the injected dose reaches the tumor, creating a narrow therapeutic window for an otherwise potent drug. First-pass metabolism of nanoparticles by the reticuloendothelial system (RES) has been identified as a major culprit for the depletion of nanoparticles in circulation before they reach the tumor site. To overcome this, new strategies, materials, and functionalization with stealth polymers have been developed to improve nanoparticle circulation and uptake at the tumor site. This review summarizes the strategies undertaken to evade RES uptake of nanomedicines and improve the passive and active targeting of nanoparticle drugs to solid tumors. We also outline the limitations of current strategies and the future directions we believe will be explored to yield significant benefits to patients and make nanomedicine a promising treatment modality for cancer.
Collapse
|
39
|
Kiesel BF, Guo J, Parise RA, Venkataramanan R, Clump DA, Bakkenist CJ, Beumer JH. Dose-dependent bioavailability and tissue distribution of the ATR inhibitor AZD6738 (ceralasertib) in mice. Cancer Chemother Pharmacol 2022; 89:231-242. [PMID: 35066692 PMCID: PMC8829872 DOI: 10.1007/s00280-021-04388-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/14/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE Ataxia telangiectasia and Rad3-related (ATR) initiates and regulates cellular responses to DNA damage, such as those caused by cancer treatments. Several ATR inhibitors (ATRi) are in clinical development including AZD6738. Therapeutic indices among ATRi may differ as a result of varying potencies and concentrations at both tumor and off-target sites. Additionally, AZD6738 contributes to anti-tumor immune responses necessitating evaluation of exposure at immunological sites. METHODS Using mouse models and a highly sensitive LC-MS/MS assay, the pharmacokinetics of AZD6738 were studied, including dose linearity, bioavailability, metabolism, and tissue distribution in tumor-bearing mice. RESULTS Initial studies identified dose-dependent bioavailability, with greater than proportional increases in exposure as dose increased resulting in a ~ twofold increase in bioavailability between the lowest and highest investigated doses. These behaviors were successfully captured with a compartmental PK model. Analysis of metabolite PK revealed decreasing metabolic ratios with increasing dose, indicative of saturable first-pass metabolism. Further analysis revealed that intestinal and gut metabolism contribute to metabolism and these saturable mechanisms. Studies of tumor and tissue distribution found rapid and extensive drug distribution to most tissues except brain and spinal cord. CONCLUSION The complex non-linear behavior of AZD6738 PK in mice was due to pre-systemic saturation and which appears to be recapitulated clinically at low doses. PK reported here will allow future correlation of tissue related toxicities with drug exposure as well as exposure with immunological responses. These results can also be compared with those from similar studies of other ATRi to contrast drug exposure with responses.
Collapse
Affiliation(s)
- Brian F Kiesel
- Cancer Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianxia Guo
- Cancer Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Robert A Parise
- Cancer Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Raman Venkataramanan
- Cancer Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Clump
- Department of Radiation Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher J Bakkenist
- Department of Radiation Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jan H Beumer
- Cancer Therapeutics Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Room G27e, 5117 Centre Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
40
|
Liu M, Huang Q, Zhu Y, Chen L, Li Y, Gong Z, Ai K. Harnessing reactive oxygen/nitrogen species and inflammation: Nanodrugs for liver injury. Mater Today Bio 2022; 13:100215. [PMID: 35198963 PMCID: PMC8850330 DOI: 10.1016/j.mtbio.2022.100215] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 12/11/2022] Open
Abstract
Overall, 12% of the global population (800 million) suffers from liver disease, which causes 2 million deaths every year. Liver injury involving characteristic reactive oxygen/nitrogen species (RONS) and inflammation plays a key role in progression of liver disease. As a key metabolic organ of the human body, the liver is susceptible to injury from various sources, including COVID-19 infection. Owing to unique structural features and functions of the liver, most current antioxidants and anti-inflammatory drugs are limited against liver injury. However, the characteristics of the liver could be utilized in the development of nanodrugs to achieve specific enrichment in the liver and consequently targeted treatment. Nanodrugs have shown significant potential in eliminating RONS and regulating inflammation, presenting an attractive therapeutic tool for liver disease through controlling liver injury. Therefore, the main aim of the current review is to provide a comprehensive summary of the latest developments contributing to our understanding of the mechanisms underlying nanodrugs in the treatment of liver injury via harnessing RONS and inflammation. Meanwhile, the prospects of nanodrugs for liver injury therapy are systematically discussed, which provides a sound platform for novel therapeutic insights and inspiration for design of nanodrugs to treat liver disease.
Collapse
Affiliation(s)
- Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yan Zhu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Li Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yumei Li
- Department of Assisted Reproduction, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
41
|
Damgalı Ş, Özdemir S, Kaya G, Demirkoz AB, Üner M. Development of monolithic matrix type transdermal patches containing cinnarizine: Physical characterization and permeation studies. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | | | | | - Aslı Barla Demirkoz
- Aromsa Besin Aroma ve Katkı Maddeleri Sanayi ve Ticaret Anonim Şirketi, Turkey; Halic University, Turkey
| | | |
Collapse
|
42
|
Le T, Aguilar B, Mangal JL, Acharya AP. Oral drug delivery for immunoengineering. Bioeng Transl Med 2022; 7:e10243. [PMID: 35111945 PMCID: PMC8780903 DOI: 10.1002/btm2.10243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 11/13/2022] Open
Abstract
The systemic pharmacotherapeutic efficacy of immunomodulatory drugs is heavily influenced by its route of administration. A few common routes for the systemic delivery of immunotherapeutics are intravenous, intraperitoneal, and intramuscular injections. However, the development of novel biomaterials, in adjunct to current progress in immunoengineering, is providing an exciting area of interest for oral drug delivery for systemic targeting. Oral immunotherapeutic delivery is a highly preferred route of administration due to its ease of administration, higher patient compliance, and increased ability to generate specialized immune responses. However, the harsh environment and slow systemic absorption, due to various biological barriers, reduces the immunotherapeutic bioavailability, and in turn prevents widespread use of oral delivery. Nonetheless, cutting edge biomaterials are being synthesized to combat these biological barriers within the gastrointestinal (GI) tract for the enhancement of drug bioavailability and targeting the immune system. For example, advancements in biomaterials and synthesized drug agents have provided distinctive methods to promote localized drug absorption for the modulation of local or systemic immune responses. Additionally, novel breakthroughs in the immunoengineering field show promise in the development of vaccine delivery systems for disease prevention as well as combating autoimmune diseases, inflammatory diseases, and cancer. This review will discuss current progress made within the field of biomaterials and drug delivery systems to enhance oral immunotherapeutic availability, and how these new delivery platforms can be utilized to deliver immunotherapeutics for resolution of immune-related diseases.
Collapse
Affiliation(s)
- Tien Le
- Chemical Engineering, School for the Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
| | - Brian Aguilar
- Biomedical Engineering, School of Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
| | - Joslyn L. Mangal
- Biological Design, School for Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
| | - Abhinav P. Acharya
- Chemical Engineering, School for the Engineering of Matter, Transport, and EnergyArizona State UniversityTempeArizonaUSA
- Biomedical Engineering, School of Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
- Biological Design, School for Biological and Health Systems EngineeringArizona State UniversityTempeArizonaUSA
- Materials Science and Engineering, School for the Engineering of Matter, Transport, and energyArizona State UniversityTempeArizonaUSA
- Biodesign Center for Immunotherapy, Vaccines and VirotherapyArizona State UniversityTempeArizonaUSA
| |
Collapse
|
43
|
Best practices in current models mimicking drug permeability in the gastrointestinal tract - an UNGAP review. Eur J Pharm Sci 2021; 170:106098. [PMID: 34954051 DOI: 10.1016/j.ejps.2021.106098] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/19/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022]
Abstract
The absorption of orally administered drug products is a complex, dynamic process, dependent on a range of biopharmaceutical properties; notably the aqueous solubility of a molecule, stability within the gastrointestinal tract (GIT) and permeability. From a regulatory perspective, the concept of high intestinal permeability is intrinsically linked to the fraction of the oral dose absorbed. The relationship between permeability and the extent of absorption means that experimental models of permeability have regularly been used as a surrogate measure to estimate the fraction absorbed. Accurate assessment of a molecule's intestinal permeability is of critical importance during the pharmaceutical development process of oral drug products, and the current review provides a critique of in vivo, in vitro and ex vivo approaches. The usefulness of in silico models to predict drug permeability is also discussed and an overview of solvent systems used in permeability assessments is provided. Studies of drug absorption in humans are an indirect indicator of intestinal permeability, but in vitro and ex vivo tools provide initial screening approaches are important tools for direct assessment of permeability in drug development. Continued refinement of the accuracy of in silico approaches and their validation with human in vivo data will facilitate more efficient characterisation of permeability earlier in the drug development process and will provide useful inputs for integrated, end-to-end absorption modelling.
Collapse
|
44
|
Fayzullin A, Bakulina A, Mikaelyan K, Shekhter A, Guller A. Implantable Drug Delivery Systems and Foreign Body Reaction: Traversing the Current Clinical Landscape. Bioengineering (Basel) 2021; 8:bioengineering8120205. [PMID: 34940358 PMCID: PMC8698517 DOI: 10.3390/bioengineering8120205] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/02/2023] Open
Abstract
Precise delivery of therapeutics to the target structures is essential for treatment efficiency and safety. Drug administration via conventional routes requires overcoming multiple transport barriers to achieve and maintain the local drug concentration and commonly results in unwanted off-target effects. Patients’ compliance with the treatment schedule remains another challenge. Implantable drug delivery systems (IDDSs) provide a way to solve these problems. IDDSs are bioengineering devices surgically placed inside the patient’s tissues to avoid first-pass metabolism and reduce the systemic toxicity of the drug by eluting the therapeutic payload in the vicinity of the target tissues. IDDSs present an impressive example of successful translation of the research and engineering findings to the patient’s bedside. It is envisaged that the IDDS technologies will grow exponentially in the coming years. However, to pave the way for this progress, it is essential to learn lessons from the past and present of IDDSs clinical applications. The efficiency and safety of the drug-eluting implants depend on the interactions between the device and the hosting tissues. In this review, we address this need and analyze the clinical landscape of the FDA-approved IDDSs applications in the context of the foreign body reaction, a key aspect of implant–tissue integration.
Collapse
Affiliation(s)
- Alexey Fayzullin
- Department of Experimental Morphology and Biobanking, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.F.); (A.B.); (K.M.); (A.S.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Alesia Bakulina
- Department of Experimental Morphology and Biobanking, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.F.); (A.B.); (K.M.); (A.S.)
| | - Karen Mikaelyan
- Department of Experimental Morphology and Biobanking, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.F.); (A.B.); (K.M.); (A.S.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Anatoly Shekhter
- Department of Experimental Morphology and Biobanking, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.F.); (A.B.); (K.M.); (A.S.)
| | - Anna Guller
- Department of Experimental Morphology and Biobanking, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.F.); (A.B.); (K.M.); (A.S.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Biomolecular Discovery Research Centre, Macquarie University, Sydney, NSW 2109, Australia
- Correspondence:
| |
Collapse
|
45
|
Elz AS, Trevaskis NL, Porter CJH, Bowen JM, Prestidge CA. Smart design approaches for orally administered lipophilic prodrugs to promote lymphatic transport. J Control Release 2021; 341:676-701. [PMID: 34896450 DOI: 10.1016/j.jconrel.2021.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/22/2022]
Abstract
Challenges to effective delivery of drugs following oral administration has attracted growing interest over recent decades. Small molecule drugs (<1000 Da) are generally absorbed across the gastrointestinal tract into the portal blood and further transported to the systemic circulation via the liver. This can result in a significant reduction to the oral bioavailability of drugs that are metabolically labile and ultimately lead to ineffective exposure and treatment. Targeting drug delivery to the intestinal lymphatics is attracting increased attention as an alternative route of drug transportation providing multiple benefits. These include bypassing hepatic first-pass metabolism and selectively targeting disease reservoirs residing within the lymphatic system. The particular physicochemical requirements for drugs to be able to access the lymphatics after oral delivery include high lipophilicity (logP>5) and high long-chain triglyceride solubility (> 50 mg/g), properties required to enable drug association with the lipoprotein transport pathway. The majority of small molecule drugs, however, are not this lipophilic and therefore not substantially transported via the intestinal lymph. This has contributed to a growing body of investigation into prodrug approaches to deliver drugs to the lymphatic system by chemical manipulation. Optimised lipophilic prodrugs have the potential to increase lymphatic transport thereby improving oral pharmacokinetics via a reduction in first pass metabolism and may also target of disease-specific reservoirs within the lymphatics. This may provide advantages for current pharmacotherapy approaches for a wide array of pathological conditions, e.g. immune disease, cancer and metabolic disease, and also presents a promising approach for advanced vaccination strategies. In this review, specific emphasis is placed on medicinal chemistry strategies that have been successfully employed to design lipophilic prodrugs to deliberately enable lymphatic transport. Recent progress and opportunities in medicinal chemistry and drug delivery that enable new platforms for efficacious and safe delivery of drugs are critically evaluated.
Collapse
Affiliation(s)
- Aurelia S Elz
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| | - Natalie L Trevaskis
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Christopher J H Porter
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia.
| | - Joanne M Bowen
- School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
46
|
Lee BE, Kim DK, Lee H, Yoon S, Park SH, Lee S, Yoo J. Recapitulation of First Pass Metabolism Using 3D Printed Microfluidic Chip and Organoid. Cells 2021; 10:3301. [PMID: 34943808 PMCID: PMC8699265 DOI: 10.3390/cells10123301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 01/16/2023] Open
Abstract
The low bioavailability of oral drugs due to first pass metabolism is a major obstacle in drug development. With significant developments in the field of in vitro organ modeling and microfluidic chip three-dimensional (3D) printing, the challenge is to apply these for the production and evaluation of new drug candidates. This study aimed to produce a microfluidic chip to recapitulate and assess the feasibility of the first pass metabolism. The infill condition of the polycarbonate transparent filament and layer height was optimized to visualize and maintain the organoid or spheroid on the chip. Next, the chip was fabricated using a 3D printer after a computer-aided design (CAD). The chip consisted of three wells of different heights. The small intestinal (SI) organoid and colorectal adenocarcinoma spheroids were placed on the second and third wells, respectively. No additional equipment was assembled, and the tilted tunnel was connected to each well to transport the material by gradient force. The chip was fabricated using 50% and 0.1 um thickness. Among the three different prototypes of chip (chips 1, 2, and 3), the highest distribution of plasmids in the Matrigel of the second well was observed in Chip 2 at 48 h. The effect of first pass metabolism was analyzed using docetaxel. In the chip without an SI organoid, there was a marked decrease in the viability of colorectal adenocarcinoma spheroids due to drug efficacy. However, in the chip with the SI organoid, no significant change in viability was observed because of first pass metabolism. In conclusion, we presented a simple, fast, and low-cost microfluidic chip to analyze the efficacy change of candidate drug by the first pass metabolism.
Collapse
Affiliation(s)
- Bo-Eun Lee
- CHA Organoid Research Center, Department of Microbilogy, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea;
- Organoidsciences, Ltd., Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea
| | - Do-Kyung Kim
- CHA Bundang Medical Center, Department of Orthopaedic Surgery, School of Medicine, CHA University, Seongnam 13496, Gyeonggi-do, Korea; (D.-K.K.); (S.Y.)
| | - Hyunil Lee
- Department of Orthopaedic Surgery, Ilsan Paik Hospital, Inje University, Goyang 10380, Gyeonggi-do, Korea;
| | - Siyeong Yoon
- CHA Bundang Medical Center, Department of Orthopaedic Surgery, School of Medicine, CHA University, Seongnam 13496, Gyeonggi-do, Korea; (D.-K.K.); (S.Y.)
| | - Sin-Hyung Park
- Department of Orthopaedic Surgery, Soonchunhyang University Bucheon Hospital, Bucheon 39371, Gyeonggi-do, Korea;
| | - Soonchul Lee
- CHA Bundang Medical Center, Department of Orthopaedic Surgery, School of Medicine, CHA University, Seongnam 13496, Gyeonggi-do, Korea; (D.-K.K.); (S.Y.)
| | - Jongman Yoo
- CHA Organoid Research Center, Department of Microbilogy, School of Medicine, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea;
- Organoidsciences, Ltd., Bundang-gu, Seongnam 13488, Gyeonggi-do, Korea
| |
Collapse
|
47
|
Jones AW. Comment on "Estimates of Non-Alcoholic Food-Derived Ethanol and Methanol in Human". J Anal Toxicol 2021; 46:e48-e51. [PMID: 34698859 DOI: 10.1093/jat/bkab112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Alan Wayne Jones
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
48
|
Kim J, Shim MK, Cho YJ, Jeon S, Moon Y, Choi J, Kim J, Lee J, Lee JW, Kim K. The safe and effective intraperitoneal chemotherapy with cathepsin B-specific doxorubicin prodrug nanoparticles in ovarian cancer with peritoneal carcinomatosis. Biomaterials 2021; 279:121189. [PMID: 34695659 DOI: 10.1016/j.biomaterials.2021.121189] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 10/20/2022]
Abstract
Intraperitoneal (IP) chemotherapy has shown promising efficacy in ovarian cancer with peritoneal carcinomatosis (PC), but in vivo rapid clearance and severe toxicity of free anticancer drugs hinder the effective treatment. Herein, we propose the safe and effective IP chemotherapy with cathepsin B-specific doxorubicin prodrug nanoparticles (PNPs) in ovarian cancer with PC. The PNPs are prepared by self-assembling cathepsin B-specific cleavable peptide (FRRG) and doxorubicin (DOX) conjugates, which are further formulated with pluronic F68. The PNPs exhibit stable spherical structure and cytotoxic DOX is specifically released from PNPs via sequential enzymatic degradation by cathepsin B and intracellular proteases. The PNPs induce cytotoxicity in cathepsin B-overexpressing ovarian (SKOV-3 and HeyA8) and colon (MC38 and CT26) cancer cells, but not in cathepsin B-deficient normal cells in cultured condition. With enhanced cancer-specificity and in vivo residence time, IP injected PNPs efficiently accumulate within PC through two targeting mechanisms of direct penetration (circulation independent) and systemic blood vessel-associated accumulation (circulation dependent). As a result, IP chemotherapy with PNPs efficiently inhibit tumor progression with minimal side effects in peritoneal human ovarian tumor-bearing xenograft (POX) and patient derived xenograft (PDX) models. These results demonstrate that PNPs effectively inhibit progression of ovarian cancer with peritoneal carcinomatosis with minimal local and systemic toxicities by high cancer-specificity and favorable in vivo PK/PD profiles enhancing PC accumulation.
Collapse
Affiliation(s)
- Jinseong Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Man Kyu Shim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Young-Jae Cho
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sangmin Jeon
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yujeong Moon
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Bioengineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jiwoong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongrae Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jaewan Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Kwangmeyung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
49
|
Osborn LJ, Claesen J, Brown JM. Microbial Flavonoid Metabolism: A Cardiometabolic Disease Perspective. Annu Rev Nutr 2021; 41:433-454. [PMID: 34633856 DOI: 10.1146/annurev-nutr-120420-030424] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cardiometabolic disease (CMD) is a leading cause of death worldwide and encompasses the inflammatory metabolic disorders of obesity, type 2 diabetes mellitus, nonalcoholic fatty liver disease, and cardiovascular disease. Flavonoids are polyphenolic plant metabolites that are abundantly present in fruits and vegetables and have biologically relevant protective effects in a number of cardiometabolic disorders. Several epidemiological studies underscored a negative association between dietary flavonoid consumption and the propensity to develop CMD. Recent studies elucidated the contribution of the gut microbiota in metabolizing dietary intake as it relates to CMD. Importantly, the biological efficacy of flavonoids in humans and animal models alike is linked to the gut microbial community. Herein, we discuss the opportunities and challenges of leveraging flavonoid intake as a potential strategy to prevent and treat CMD in a gut microbe-dependent manner, with special emphasis on flavonoid-derived microbial metabolites.
Collapse
Affiliation(s)
- Lucas J Osborn
- Department of Cardiovascular and Metabolic Sciences and Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio 44195, USA; , , .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, USA
| | - Jan Claesen
- Department of Cardiovascular and Metabolic Sciences and Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio 44195, USA; , , .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, USA
| | - J Mark Brown
- Department of Cardiovascular and Metabolic Sciences and Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, Ohio 44195, USA; , , .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio 44195, USA
| |
Collapse
|
50
|
Armitage JM, Hughes L, Sangion A, Arnot JA. Development and intercomparison of single and multicompartment physiologically-based toxicokinetic models: Implications for model selection and tiered modeling frameworks. ENVIRONMENT INTERNATIONAL 2021; 154:106557. [PMID: 33892222 DOI: 10.1016/j.envint.2021.106557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/05/2021] [Accepted: 04/02/2021] [Indexed: 05/21/2023]
Abstract
This study describes the development and intercomparison of generic physiologically-based toxicokinetic (PBTK) models for humans comprised of internally consistent one-compartment (1Co-) and multi-compartment (MCo-) implementations (G-PBTK). The G-PBTK models were parameterized for an adult male (70 kg) using common physiological parameters and in vitro biotransformation rate estimates and subsequently evaluated using independent concentration versus time data (n = 6) and total elimination half-lives (n = 15) for diverse organic chemicals. The model performance is acceptable considering the inherent uncertainty in the biotransformation rate data and the absence of model calibration. The G-PBTK model was then applied using hypothetical neutral organics, acidic ionizable organics and basic ionizable organics (IOCs) to identify combinations of partitioning properties and biotransformation rates leading to substantial discrepancies between 1Co- and MCo-PBTK calculations for whole body concentrations and half-lives. The 1Co- and MCo-PBTK model calculations for key toxicokinetic parameters are broadly consistent unless biotransformation is rapid (e.g., half-life less than five days). When half-lives are relatively short, discrepancies are greatest for the neutral organics and least for the acidic IOCs which follows from the estimated volumes of distribution (e.g., VDSS = 9.6-15.4 L/kg vs 0.3-1.6 L/kg for the neutral and acidic compounds respectively) and the related approach to internal chemical equilibrium. The model intercomparisons demonstrate that 1Co-PBTK models can be applied with confidence to many exposure scenarios, particularly those focused on chronic or repeat exposures and for prioritization and screening-level decision contexts. However, MCo-PBTK models may be necessary in certain contexts, particularly for intermittent, short-term and highly variable exposures. A key recommendation to guide model selection and the development of tiered PBTK modeling frameworks that emerges from this study is the need to harmonize models with respect to parameterization and process descriptions to the greatest extent possible when proceeding from the application of simpler to more complex modeling tools as part of chemical assessment activities.
Collapse
Affiliation(s)
- James M Armitage
- AES Armitage Environmental Sciences, Inc., Ottawa, Ontario K1L 8C3, Canada; Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada.
| | - Lauren Hughes
- ARC Arnot Research and Consulting, Toronto, Ontario M4M 1W4, Canada
| | - Alessandro Sangion
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada; ARC Arnot Research and Consulting, Toronto, Ontario M4M 1W4, Canada
| | - Jon A Arnot
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada; ARC Arnot Research and Consulting, Toronto, Ontario M4M 1W4, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|