1
|
Wu J, Huang J, Yu J, Xu M, Liu J, Pu K. Exosome-Inhibiting Polymeric Sonosensitizer for Tumor-Specific Sonodynamic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400762. [PMID: 38445783 DOI: 10.1002/adma.202400762] [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: 01/15/2024] [Revised: 03/04/2024] [Indexed: 03/07/2024]
Abstract
Combination cancer immunotherapy based on electromagnetic energy and immunotherapy shows potent anti-cancer efficacy. However, as a factor that mediates tumor metastasis and immune suppression, the impact of tumor exosomes on therapy under electromagnetic energy stimulation remains unclear. Herein, findings indicate that sonodynamic therapy (SDT) increases serum exosome levels by inducing apoptotic exosomes and loosening the tumor extracellular matrix, promoting lung metastasis. To address this problem, an exosome-inhibiting polymeric sonosensitizer (EIPS) selectively inhibiting tumor exosome generation in response to the tumor biomarker is synthesized. EIPS consists of a semiconducting polymer backbone capable of inducing SDT and a poly(ethylene glycol) layer conjugated with a tumor-specific enzyme-responsive exosome inhibitor prodrug. After being cleaved by tumor Cathepsin B, EIPS releases active exosome inhibitors, preventing tumor exosome-mediated immune suppression and lung metastasis. As a result, EIPS elicits robust antitumor effects through the synergistic effect of SDT and tumor exosome inhibition, completely preventing lung metastasis and establishing a long-term immune memory effect. This is the first example showing that combining SDT with tumor-specific exosome inhibition can elicit a potent immune response without the help of typical immune agonists.
Collapse
Affiliation(s)
- Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jingsheng Huang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jie Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| |
Collapse
|
2
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [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] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
3
|
Schempp R, Eilts J, Schöl M, Grijalva Yépez MF, Fekete A, Wigger D, Schumacher F, Kleuser B, van Ham M, Jänsch L, Sauer M, Avota E. The Role of Neutral Sphingomyelinase-2 (NSM2) in the Control of Neutral Lipid Storage in T Cells. Int J Mol Sci 2024; 25:3247. [PMID: 38542220 PMCID: PMC10970209 DOI: 10.3390/ijms25063247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/25/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
The accumulation of lipid droplets (LDs) and ceramides (Cer) is linked to non-alcoholic fatty liver disease (NAFLD), regularly co-existing with type 2 diabetes and decreased immune function. Chronic inflammation and increased disease severity in viral infections are the hallmarks of the obesity-related immunopathology. The upregulation of neutral sphingomyelinase-2 (NSM2) has shown to be associated with the pathology of obesity in tissues. Nevertheless, the role of sphingolipids and specifically of NSM2 in the regulation of immune cell response to a fatty acid (FA) rich environment is poorly studied. Here, we identified the presence of the LD marker protein perilipin 3 (PLIN3) in the intracellular nano-environment of NSM2 using the ascorbate peroxidase APEX2-catalyzed proximity-dependent biotin labeling method. In line with this, super-resolution structured illumination microscopy (SIM) shows NSM2 and PLIN3 co-localization in LD organelles in the presence of increased extracellular concentrations of oleic acid (OA). Furthermore, the association of enzymatically active NSM2 with isolated LDs correlates with increased Cer levels in these lipid storage organelles. NSM2 enzymatic activity is not required for NSM2 association with LDs, but negatively affects the LD numbers and cellular accumulation of long-chain unsaturated triacylglycerol (TAG) species. Concurrently, NSM2 expression promotes mitochondrial respiration and fatty acid oxidation (FAO) in response to increased OA levels, thereby shifting cells to a high energetic state. Importantly, endogenous NSM2 activity is crucial for primary human CD4+ T cell survival and proliferation in a FA rich environment. To conclude, our study shows a novel NSM2 intracellular localization to LDs and the role of enzymatically active NSM2 in metabolic response to enhanced FA concentrations in T cells.
Collapse
Affiliation(s)
- Rebekka Schempp
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Wuerzburg, Germany; (R.S.); (M.S.); (M.F.G.Y.)
| | - Janna Eilts
- Department of Biotechnology and Biophysics, Biocenter, University of Wuerzburg, 97074 Wuerzburg, Germany; (J.E.); (M.S.)
| | - Marie Schöl
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Wuerzburg, Germany; (R.S.); (M.S.); (M.F.G.Y.)
| | - Maria Fernanda Grijalva Yépez
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Wuerzburg, Germany; (R.S.); (M.S.); (M.F.G.Y.)
| | - Agnes Fekete
- Pharmaceutical Biology, Julius-von-Sachs-Institute, Biocenter, University of Wuerzburg, 97082 Wuerzburg, Germany;
| | - Dominik Wigger
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (D.W.); (F.S.); (B.K.)
| | - Fabian Schumacher
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (D.W.); (F.S.); (B.K.)
| | - Burkhard Kleuser
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (D.W.); (F.S.); (B.K.)
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.v.H.); (L.J.)
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.v.H.); (L.J.)
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Wuerzburg, 97074 Wuerzburg, Germany; (J.E.); (M.S.)
| | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Wuerzburg, Germany; (R.S.); (M.S.); (M.F.G.Y.)
| |
Collapse
|
4
|
Dichiara M, Cosentino G, Giordano G, Pasquinucci L, Marrazzo A, Costanzo G, Amata E. Designing drugs optimized for both blood-brain barrier permeation and intra-cerebral partition. Expert Opin Drug Discov 2024; 19:317-329. [PMID: 38145409 DOI: 10.1080/17460441.2023.2294118] [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: 10/08/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
INTRODUCTION With the increasing incidence and prevalence of neurological disorders globally, there is a paramount need for new pharmacotherapies. BBB effectively protects the brain but raises a profound challenge to drug permeation, with less than 2% of most drugs reaching the CNS. AREAS COVERED This article reviews aspects of the most recent design strategies, providing insights into ideas and concepts in CNS drug discovery. An overview of the products available on the market is given and why clinical trials are continuously failing is discussed. EXPERT OPINION Among the available CNS drugs, small molecules account for most successful CNS therapeutics due to their ability to penetrate the BBB through passive or carrier-mediated mechanisms. The development of new CNS drugs is very difficult. To date, there is a lack of effective drugs for alleviating or even reversing the progression of brain diseases. Particularly, the use of artificial intelligence strategies, together with more appropriate animal models, may enable the design of molecules with appropriate permeation, to elicit a biological response from the neurotherapeutic target.
Collapse
Affiliation(s)
- Maria Dichiara
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Giuseppe Cosentino
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Giorgia Giordano
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Lorella Pasquinucci
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Agostino Marrazzo
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Giuliana Costanzo
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| | - Emanuele Amata
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Catania, Italy
| |
Collapse
|
5
|
Subbaiah MAM, Rautio J, Meanwell NA. Prodrugs as empowering tools in drug discovery and development: recent strategic applications of drug delivery solutions to mitigate challenges associated with lead compounds and drug candidates. Chem Soc Rev 2024; 53:2099-2210. [PMID: 38226865 DOI: 10.1039/d2cs00957a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The delivery of a drug to a specific organ or tissue at an efficacious concentration is the pharmacokinetic (PK) hallmark of promoting effective pharmacological action at a target site with an acceptable safety profile. Sub-optimal pharmaceutical or ADME profiles of drug candidates, which can often be a function of inherently poor physicochemical properties, pose significant challenges to drug discovery and development teams and may contribute to high compound attrition rates. Medicinal chemists have exploited prodrugs as an informed strategy to productively enhance the profiles of new chemical entities by optimizing the physicochemical, biopharmaceutical, and pharmacokinetic properties as well as selectively delivering a molecule to the site of action as a means of addressing a range of limitations. While discovery scientists have traditionally employed prodrugs to improve solubility and membrane permeability, the growing sophistication of prodrug technologies has enabled a significant expansion of their scope and applications as an empowering tool to mitigate a broad range of drug delivery challenges. Prodrugs have emerged as successful solutions to resolve non-linear exposure, inadequate exposure to support toxicological studies, pH-dependent absorption, high pill burden, formulation challenges, lack of feasibility of developing solid and liquid dosage forms, first-pass metabolism, high dosing frequency translating to reduced patient compliance and poor site-specific drug delivery. During the period 2012-2022, the US Food and Drug Administration (FDA) approved 50 prodrugs, which amounts to 13% of approved small molecule drugs, reflecting both the importance and success of implementing prodrug approaches in the pursuit of developing safe and effective drugs to address unmet medical needs.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon Bristol Myers Squibb R&D Centre, Biocon Park, Bommasandra Phase IV, Bangalore, PIN 560099, India.
| | - Jarkko Rautio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Nicholas A Meanwell
- The Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
- Department of Medicinal Chemistry, The College of Pharmacy, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Novotná K, Tenora L, Prchalová E, Paule J, Alt J, Veeravalli V, Lam J, Wu Y, Šnajdr I, Gori S, Mettu VS, Tsukamoto T, Majer P, Slusher BS, Rais R. Discovery of tert-Butyl Ester Based 6-Diazo-5-oxo-l-norleucine Prodrugs for Enhanced Metabolic Stability and Tumor Delivery. J Med Chem 2023; 66:15493-15510. [PMID: 37949450 PMCID: PMC10683027 DOI: 10.1021/acs.jmedchem.3c01681] [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] [Received: 09/11/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) exhibits remarkable anticancer efficacy; however, its therapeutic potential is hindered by its toxicity to gastrointestinal (GI) tissues. We recently reported the discovery of DRP-104, a tumor-targeted DON prodrug with excellent efficacy and tolerability, which is currently in clinical trials. However, DRP-104 exhibits limited aqueous solubility, and the instability of its isopropyl ester promoiety leads to the formation of an inactive M1-metabolite, reducing overall systemic prodrug exposure. Herein, we aimed to synthesize DON prodrugs with various ester and amide promoieties with improved solubility, GI stability, and DON tumor delivery. Twenty-one prodrugs were synthesized and characterized in stability and pharmacokinetics studies. Of these, P11, tert-butyl-(S)-6-diazo-2-((S)-2-(2-(dimethylamino)acetamido)-3-phenylpropanamido)-5-oxo-hexanoate, showed excellent metabolic stability in plasma and intestinal homogenate, high aqueous solubility, and high tumor DON exposures and preserved the ideal tumor-targeting profile of DRP-104. In conclusion, we report a new generation of glutamine antagonist prodrugs with improved physicochemical and pharmacokinetic attributes.
Collapse
Affiliation(s)
- Kateřina Novotná
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Prague 128 00, Czech Republic
| | - Lukáš Tenora
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - Eva Prchalová
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - James Paule
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jesse Alt
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Vijay Veeravalli
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jenny Lam
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ying Wu
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ivan Šnajdr
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - Sadakatali Gori
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Vijaya Saradhi Mettu
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Takashi Tsukamoto
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Pavel Majer
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - Barbara S. Slusher
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Rana Rais
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
7
|
Xiao T, Liu K, Gao Q, Chen M, Kim YS, Jin S, Ding Y, Huigens RW. Design, Synthesis, and Evaluation of Carbonate-Linked Halogenated Phenazine-Quinone Prodrugs with Improved Water-Solubility and Potent Antibacterial Profiles. ACS Infect Dis 2023; 9:899-915. [PMID: 36867688 PMCID: PMC10551733 DOI: 10.1021/acsinfecdis.2c00558] [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] [Indexed: 03/05/2023]
Abstract
Pathogenic bacteria have devastating impacts on human health as a result of acquired antibiotic resistance and innate tolerance. Every class of our current antibiotic arsenal was initially discovered as growth-inhibiting agents that target actively replicating (individual, free-floating) planktonic bacteria. Bacteria are notorious for utilizing a diversity of resistance mechanisms to overcome the action of conventional antibiotic therapies and forming surface-attached biofilm communities enriched in (non-replicating) persister cells. To address problems associated with pathogenic bacteria, our group is developing halogenated phenazine (HP) molecules that demonstrate potent antibacterial and biofilm-eradicating activities through a unique iron starvation mode of action. In this study, we designed, synthesized, and investigated a focused collection of carbonate-linked HP prodrugs bearing a quinone trigger to target the reductive cytoplasm of bacteria for bioactivation and subsequent HP release. The quinone moiety also contains a polyethylene glycol group, which dramatically enhances the water-solubility properties of the HP-quinone prodrugs reported herein. We found carbonate-linked HP-quinone prodrugs 11, 21-23 to demonstrate good linker stability, rapid release of the active HP warhead following dithiothreitol (reductive) treatment, and potent antibacterial activities against methicillin-resistant Staphylococcus aureus (MRSA), methicillin-resistant Staphylococcus epidermidis, and Enterococcus faecalis. In addition, HP-quinone prodrug 21 induced rapid iron starvation in MRSA and S. epidermidis biofilms, illustrating prodrug action within these surface-attached communities. Overall, we are highly encouraged by these findings and believe that HP prodrugs have the potential to address antibiotic resistant and tolerant bacterial infections.
Collapse
Affiliation(s)
- Tao Xiao
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Qiwen Gao
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Manyun Chen
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Young S Kim
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Yousong Ding
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W Huigens
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
8
|
Tallon C, Bell BJ, Sharma A, Pal A, Malvankar MM, Thomas AG, Yoo SW, Hollinger KR, Coleman K, Wilkinson EL, Kannan S, Haughey NJ, Kannan RM, Rais R, Slusher BS. Dendrimer-Conjugated nSMase2 Inhibitor Reduces Tau Propagation in Mice. Pharmaceutics 2022; 14:2066. [PMID: 36297501 PMCID: PMC9609094 DOI: 10.3390/pharmaceutics14102066] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/11/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the progressive accumulation of amyloid-β and hyperphosphorylated tau (pTau), which can spread throughout the brain via extracellular vesicles (EVs). Membrane ceramide enrichment regulated by the enzyme neutral sphingomyelinase 2 (nSMase2) is a critical component of at least one EV biogenesis pathway. Our group recently identified 2,6-Dimethoxy-4-(5-Phenyl-4-Thiophen-2-yl-1H-Imidazol-2-yl)-Phenol (DPTIP), the most potent (30 nM) and selective inhibitor of nSMase2 reported to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), modest brain penetration, and rapid clearance, limiting its clinical translation. To enhance its PK properties, we conjugated DPTIP to a hydroxyl-PAMAM dendrimer delivery system, creating dendrimer-DPTIP (D-DPTIP). In an acute brain injury model, orally administered D-DPTIP significantly reduced the intra-striatal IL-1β-induced increase in plasma EVs up to 72 h post-dose, while oral DPTIP had a limited effect. In a mouse tau propagation model, where a mutant hTau (P301L/S320F) containing adeno-associated virus was unilaterally seeded into the hippocampus, oral D-DPTIP (dosed 3× weekly) significantly inhibited brain nSMase2 activity and blocked the spread of pTau to the contralateral hippocampus. These data demonstrate that dendrimer conjugation of DPTIP improves its PK properties, resulting in significant inhibition of EV propagation of pTau in mice. Dendrimer-based delivery of DPTIP has the potential to be an exciting new therapeutic for AD.
Collapse
Affiliation(s)
- Carolyn Tallon
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin J. Bell
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Arindom Pal
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Kaleem Coleman
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth L. Wilkinson
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Moser Center for Leukodystrophies at Kennedy Krieger, Kennedy Krieger Institute, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Hugo W. Moser Research Institute at Kennedy-Krieger Inc., Baltimore, MD 21205, USA
- Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Moser Center for Leukodystrophies at Kennedy Krieger, Kennedy Krieger Institute, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|