1
|
Zhang Y, Liu H, Zhen W, Jiang T, Cui J. Advancement of drugs conjugated with GalNAc in the targeted delivery to hepatocytes based on asialoglycoprotein receptor. Carbohydr Res 2025; 552:109426. [PMID: 40068307 DOI: 10.1016/j.carres.2025.109426] [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: 12/03/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/22/2025]
Abstract
The asialoglycoprotein receptor (ASGPR) is specifically expressed in hepatocytes. Sugar molecules, such as asialoglycoprotein, galactose, galactosamine, and N-acetyl galactosamine (GalNAc), have a high affinity for ASGPR. This review summarizes the structure of ASGPR, the distribution of this molecule in different cells, and the factors influencing the binding of GalNAc to ASGPR. We introduce the application of GalNAc in targeted delivery into hepatocytes by forming conjugated compounds with RNAs and small molecules, and the standard methods for synthesizing GalNAc are also briefly presented. This is to provide an overview of the current research on GalNAc and to shed light on the design of the new GalNAc.
Collapse
Affiliation(s)
- Yafang Zhang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Hongliang Liu
- Pharmaron Beijing Co., Ltd. (China), Beijing, 100176, China
| | - Weina Zhen
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Tingting Jiang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Jingxuan Cui
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
2
|
Hofmeister A, Jahn-Hofmann K, Brunner B, Helms M, Metz-Weidmann C, Poeverlein C, Zech G, Li Z, Hessler G, Schreuder H, Elshorst B, Krack A, Kurz M, Heubel C, Scheidler S. Trivalent siRNA-Conjugates with Guanosine as ASGPR-Binder Show Potent Knock-Down In Vivo. J Med Chem 2025; 68:6193-6209. [PMID: 40052708 DOI: 10.1021/acs.jmedchem.4c02275] [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: 03/28/2025]
Abstract
To increase the chemical space around the well-known GalNAc-ligand as ASGPR-binder, a high-throughput screening campaign was performed, testing approximately 550,000 compounds. After evaluation of the potential screening hits, only one compound, which showed high similarity with guanosine nucleosides, was chosen for further profiling. Crystal structure analysis revealed the coordination of the Ca2+-ion within the ASGPR-binding site by the cis-diol motif of the ribose unit as well as an additional π-π-interaction of the purine heterocycle to tryptophan-243. Based on these findings, guanosine was attached via the 5'-OH group to a recently described morpholino-based nucleotide using two different linker units. The resulting morpholino-guanosine building blocks were conjugated to the 5'-end of a literature-known transthyretin targeting small interfering RNA (siRNA), leading to trivalent siRNA-guanosine conjugates, which were tested for their TTR knockdown and exhibited similar potencies as the analogous GalNAc-conjugates in vitro and in vivo.
Collapse
Affiliation(s)
- Armin Hofmeister
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | | | - Bodo Brunner
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Mike Helms
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | | | | | - Gernot Zech
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Ziyu Li
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Gerhard Hessler
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Herman Schreuder
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Bettina Elshorst
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Arne Krack
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Michael Kurz
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Christoph Heubel
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| | - Sabine Scheidler
- Sanofi R&D, Industrial Park Hoechst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
3
|
Liu M, Wang Y, Zhang Y, Hu D, Tang L, Zhou B, Yang L. Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct Target Ther 2025; 10:73. [PMID: 40059188 PMCID: PMC11891339 DOI: 10.1038/s41392-024-02112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 03/17/2025] Open
Abstract
The ability of small nucleic acids to modulate gene expression via a range of processes has been widely explored. Compared with conventional treatments, small nucleic acid therapeutics have the potential to achieve long-lasting or even curative effects via gene editing. As a result of recent technological advances, efficient small nucleic acid delivery for therapeutic and biomedical applications has been achieved, accelerating their clinical translation. Here, we review the increasing number of small nucleic acid therapeutic classes and the most common chemical modifications and delivery platforms. We also discuss the key advances in the design, development and therapeutic application of each delivery platform. Furthermore, this review presents comprehensive profiles of currently approved small nucleic acid drugs, including 11 antisense oligonucleotides (ASOs), 2 aptamers and 6 siRNA drugs, summarizing their modifications, disease-specific mechanisms of action and delivery strategies. Other candidates whose clinical trial status has been recorded and updated are also discussed. We also consider strategic issues such as important safety considerations, novel vectors and hurdles for translating academic breakthroughs to the clinic. Small nucleic acid therapeutics have produced favorable results in clinical trials and have the potential to address previously "undruggable" targets, suggesting that they could be useful for guiding the development of additional clinical candidates.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yibing Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Sampathkumar P, Jung H, Chen H, Zhang Z, Suen N, Yang Y, Huang Z, Lopez T, Benisch R, Lee SJ, Ye J, Yeh WC, Li Y. Targeted protein degradation systems to enhance Wnt signaling. eLife 2024; 13:RP93908. [PMID: 38847394 PMCID: PMC11161174 DOI: 10.7554/elife.93908] [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: 06/09/2024] Open
Abstract
Molecules that facilitate targeted protein degradation (TPD) offer great promise as novel therapeutics. The human hepatic lectin asialoglycoprotein receptor (ASGR) is selectively expressed on hepatocytes. We have previously engineered an anti-ASGR1 antibody-mutant RSPO2 (RSPO2RA) fusion protein (called SWEETS) to drive tissue-specific degradation of ZNRF3/RNF43 E3 ubiquitin ligases, which achieved hepatocyte-specific enhanced Wnt signaling, proliferation, and restored liver function in mouse models, and an antibody-RSPO2RA fusion molecule is currently in human clinical trials. In the current study, we identified two new ASGR1- and ASGR1/2-specific antibodies, 8M24 and 8G8. High-resolution crystal structures of ASGR1:8M24 and ASGR2:8G8 complexes revealed that these antibodies bind to distinct epitopes on opposing sides of ASGR, away from the substrate-binding site. Both antibodies enhanced Wnt activity when assembled as SWEETS molecules with RSPO2RA through specific effects sequestering E3 ligases. In addition, 8M24-RSPO2RA and 8G8-RSPO2RA efficiently downregulate ASGR1 through TPD mechanisms. These results demonstrate the possibility of combining different therapeutic effects and degradation mechanisms in a single molecule.
Collapse
Affiliation(s)
| | | | - Hui Chen
- Surrozen, IncSouth San FranciscoUnited States
| | | | | | - Yiran Yang
- Surrozen, IncSouth San FranciscoUnited States
| | - Zhong Huang
- Surrozen, IncSouth San FranciscoUnited States
| | - Tom Lopez
- Surrozen, IncSouth San FranciscoUnited States
| | | | | | - Jay Ye
- Surrozen, IncSouth San FranciscoUnited States
| | | | - Yang Li
- Surrozen, IncSouth San FranciscoUnited States
| |
Collapse
|
5
|
Zierke MA, Rangger C, Samadikhah K, Panzer M, Dichtl S, Hörmann N, Wilflingseder D, Schmid AM, Haubner R. [ 68Ga]Ga-NODAGA-TriGalactan, a low molecular weight tracer for the non-invasive imaging of the functional liver reserve. EJNMMI Radiopharm Chem 2024; 9:41. [PMID: 38750246 PMCID: PMC11096148 DOI: 10.1186/s41181-024-00271-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Determination of the functional liver mass is important in a variety of clinical settings including liver surgery and transplantation. [99mTc]Tc-diethylenetriamine-pentaacetic acid galactosyl human serum albumin (99mTc-GSA) is a radiotracer targeting the asialoglycoprotein receptor (ASGR) and is routinely used in Japan for this purpose. Here we describe the development and evaluation of [68Ga]Ga-NODAGA-TriGalactan a low molecular weight PET-tracer targeting this structure. RESULTS For synthesis TRIS as branching unit and NODAGA as chelator for labelling with [68Ga]Ga are included. Three galactose moieties are conjugated via a click chemistry approach resulting in the desired labelling precursor.68Ga-labelling could be accomplished in high radiochemical yield and purity. [68Ga]Ga-NODAGA-TriGalactan is very hydrophilic and revealed high plasma stability and low plasma protein binding. Fluorescence imaging showed binding on ASGR-positive organoids and the IC50-value was in the nanomolar range. Most importantly, both biodistribution as well as animal imaging studies using normal mice demonstrated high liver uptake with rapid elimination from all other organs leading to even higher liver-to-background ratios as found for 99mTc-GSA. CONCLUSION [68Ga]Ga-NODAGA-TriGalactan shows high in vitro stability and selectively binds to the ASGR allowing imaging of the functional liver mass with high contrast. Thus, our first generation compound resulted already in an alternative to 99mTc-GSA for imaging the functional liver reserve and might allow the broader use of this imaging technique.
Collapse
Affiliation(s)
- Maximilian A Zierke
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstr. 35, Innsbruck, 6020, Austria
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstr. 35, Innsbruck, 6020, Austria
| | - Kimia Samadikhah
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 73076, Tübingen, Germany
| | - Marlene Panzer
- Department of Internal Medicine I, Medical University Innsbruck, Anichstr. 35, Innsbruck, 6020, Austria
| | - Stefanie Dichtl
- Institute of Hygiene and Medical Microbiology, Medical University Innsbruck, Schöpfstr. 41, Innsbruck, 6020, Austria
| | - Nikolas Hörmann
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, University of Innsbruck, Innrain, 80-82, Innsbruck, 6020, Austria
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University Innsbruck, Schöpfstr. 41, Innsbruck, 6020, Austria
| | - Andreas M Schmid
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Röntgenweg 13, 73076, Tübingen, Germany
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University Innsbruck, Anichstr. 35, Innsbruck, 6020, Austria.
| |
Collapse
|
6
|
Zhang Z, Leng XK, Zhai YY, Zhang X, Sun ZW, Xiao JY, Lu JF, Liu K, Xia B, Gao Q, Jia M, Xu CQ, Jiang YN, Zhang XG, Tao KS, Wu JW. Deficiency of ASGR1 promotes liver injury by increasing GP73-mediated hepatic endoplasmic reticulum stress. Nat Commun 2024; 15:1908. [PMID: 38459023 PMCID: PMC10924105 DOI: 10.1038/s41467-024-46135-9] [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: 06/21/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Liver injury is a core pathological process in the majority of liver diseases, yet the genetic factors predisposing individuals to its initiation and progression remain poorly understood. Here we show that asialoglycoprotein receptor 1 (ASGR1), a lectin specifically expressed in the liver, is downregulated in patients with liver fibrosis or cirrhosis and male mice with liver injury. ASGR1 deficiency exacerbates while its overexpression mitigates acetaminophen-induced acute and CCl4-induced chronic liver injuries in male mice. Mechanistically, ASGR1 binds to an endoplasmic reticulum stress mediator GP73 and facilitates its lysosomal degradation. ASGR1 depletion increases circulating GP73 levels and promotes the interaction between GP73 and BIP to activate endoplasmic reticulum stress, leading to liver injury. Neutralization of GP73 not only attenuates ASGR1 deficiency-induced liver injuries but also improves survival in mice received a lethal dose of acetaminophen. Collectively, these findings identify ASGR1 as a potential genetic determinant of susceptibility to liver injury and propose it as a therapeutic target for the treatment of liver injury.
Collapse
Affiliation(s)
- Zhe Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiang Kai Leng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yuan Yuan Zhai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiao Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhi Wei Sun
- Beijing Sungen Biomedical Technology Co. Ltd, Beijing, China
| | - Jun Ying Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jun Feng Lu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Kun Liu
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Air Force Medical University, Xi'an, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qi Gao
- Beijing Sungen Biomedical Technology Co. Ltd, Beijing, China
| | - Miao Jia
- Beijing Sungen Biomedical Technology Co. Ltd, Beijing, China
| | - Cheng Qi Xu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Na Jiang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Gang Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Kai Shan Tao
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Air Force Medical University, Xi'an, China.
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
7
|
Li Q, Yin K, Ma HP, Liu HH, Li S, Luo X, Hu R, Zhang WW, Lv ZS, Niu XL, Gu MH, Li CL, Liu YS, Liu YJ, Li HB, Li N, Li C, Gu WW, Li JJ. Application of improved GalNAc conjugation in development of cost-effective siRNA therapies targeting cardiovascular diseases. Mol Ther 2024; 32:637-645. [PMID: 38204163 PMCID: PMC10928129 DOI: 10.1016/j.ymthe.2024.01.008] [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: 09/22/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
N-Acetylgalactosamine (GalNAc)-conjugated small interfering RNA (siRNA) therapies have received approval for treating both orphan and prevalent diseases. To improve in vivo efficacy and streamline the chemical synthesis process for efficient and cost-effective manufacturing, we conducted this study to identify better designs of GalNAc-siRNA conjugates for therapeutic development. Here, we present data on redesigned GalNAc-based ligands conjugated with siRNAs against angiopoietin-like 3 (ANGPTL3) and lipoprotein (a) (Lp(a)), two target molecules with the potential to address large unmet medical needs in atherosclerotic cardiovascular diseases. By attaching a novel pyran-derived scaffold to serial monovalent GalNAc units before solid-phase oligonucleotide synthesis, we achieved increased GalNAc-siRNA production efficiency with fewer synthesis steps compared to the standard triantennary GalNAc construct L96. The improved GalNAc-siRNA conjugates demonstrated equivalent or superior in vivo efficacy compared to triantennary GalNAc-conjugated siRNAs.
Collapse
Affiliation(s)
- Qian Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Ke Yin
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Hai-Ping Ma
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Hui-Hui Liu
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Heart Failure Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sha Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Luo
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Rong Hu
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | | | | | | | - Mei-Hua Gu
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Cheng-Lu Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | | | | | - Hai-Bo Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Nancy Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | - Chong Li
- Genoval Therapeutics Co., Ltd, Shanghai, China
| | | | - Jian-Jun Li
- Cardiometabolic Center, State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Ye W, Tang Q, Zhou T, Zhou C, Fan C, Wang X, Wang C, Zhang K, Liao G, Zhou W. Design, synthesis and biological evaluation of the positional isomers of the galactose conjugates able to target hepatocellular carcinoma cells via ASGPR-mediated cellular uptake and cytotoxicity. Eur J Med Chem 2024; 264:115988. [PMID: 38039790 DOI: 10.1016/j.ejmech.2023.115988] [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: 09/23/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023]
Abstract
Galactose as a recognizing motif for asialoglycoprotein receptor (ASGPR) is a widely accepted vector to deliver cytotoxic agents in the therapy of hepatocellular carcinoma (HCC), however, the individual hydroxyl group of galactose (Gal) contributed to recognizing ASGPR is obscure and remains largely unanswered in the design of glycoconjugates. Herein, we designed and synthesized five positional isomers of Gal-anthocyanin Cy5.0 conjugates and three Gal-doxorubicin (Dox) isomers, respectively. The fluorescence intensity of Gal-Cy5.0 conjugates accumulated in cancer cells hinted the optimal modification sites of positions C2 and C6. Comparing to the cytotoxicity of other conjugates, C2-Gal-Dox (11) was the most potent. Moreover, Gal-Dox conjugates significantly the toxicity of Dox. A progressively lower internalization capacity and siRNA technology implied the cellular uptake and cytotoxicity directly related to the ASGPR expression level. Accordingly, position C2 of galactose may be the best substitution site via ASGPR mediation in the design of anti-HCC glycoconjugates.
Collapse
Affiliation(s)
- Wenchong Ye
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, Guangdong, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Qun Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Tiantian Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Cui Zhou
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chuangchuang Fan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China
| | - Guochao Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, E. 232, University Town, Waihuan Rd, Panyu, Guangzhou, 510006, Guangdong, China.
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 200241, Shanghai, China; Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| |
Collapse
|
9
|
Devanaboyina SC, Li P, LaGory EL, Poon-Andersen C, Cook KD, Soto M, Wang Z, Dang K, Uyeda C, Case RB, Thomas VA, Primack R, Ponce M, Di M, Ouyang B, Kaner J, Lam SK, Mostafavi M. Rapid depletion of "catch-and-release" anti-ASGR1 antibody in vivo. MAbs 2024; 16:2383013. [PMID: 39051531 PMCID: PMC11275528 DOI: 10.1080/19420862.2024.2383013] [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: 01/05/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Targeting antigens with antibodies exhibiting pH/Ca2+-dependent binding against an antigen is an attractive strategy to mitigate target-mediated disposition and antigen buffering. Studies have reported improved serum exposure of antibodies exhibiting pH/Ca2+-binding against membrane-bound receptors. Asialoglycoprotein receptor 1 (ASGR1) is a membrane-bound receptor primarily localized in hepatocytes. With a high expression level of approximately one million receptors per cell, high turnover, and rapid recycling, targeting this receptor with a conventional antibody is a challenge. In this study, we identified an antibody exhibiting pH/Ca2+-dependent binding to ASGR1 and generated antibody variants with increased binding to neonatal crystallizable fragment receptor (FcRn). Serum exposures of the generated anti-ASGR1 antibodies were analyzed in transgenic mice expressing human FcRn. Contrary to published reports of increased serum exposure of pH/Ca2+-dependent antibodies, the pH/Ca2+-dependent anti-ASGR1 antibody had rapid serum clearance in comparison to a conventional anti-ASGR1 antibody. We conducted sub-cellular trafficking studies of the anti-ASGR1 antibodies along with receptor quantification analysis for mechanistic understanding of the rapid serum clearance of pH/Ca2+-dependent anti-ASGR1 antibody. The findings from our study provide valuable insights in identifying the antigens, especially membrane bound, that may benefit from targeting with pH/Ca2+-dependent antibodies to obtain increased serum exposure.
Collapse
Affiliation(s)
- Siva Charan Devanaboyina
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Peng Li
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Edward L. LaGory
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Carrie Poon-Andersen
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Kevin D. Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Marcus Soto
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Zhe Wang
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Khue Dang
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Craig Uyeda
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ryan B. Case
- Department of Lead Discovery and Characterization, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Veena A. Thomas
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Ronya Primack
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Manuel Ponce
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Mei Di
- Department of Cardiometabolic disorders, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Brian Ouyang
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Joelle Kaner
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Sheung Kwan Lam
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| | - Mina Mostafavi
- Department of Biologics, Amgen Research, Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
10
|
Kirikovich SS, Levites EV, Proskurina AS, Ritter GS, Peltek SE, Vasilieva AR, Ruzanova VS, Dolgova EV, Oshihmina SG, Sysoev AV, Koleno DI, Danilenko ED, Taranov OS, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. The Molecular Aspects of Functional Activity of Macrophage-Activating Factor GcMAF. Int J Mol Sci 2023; 24:17396. [PMID: 38139225 PMCID: PMC10743851 DOI: 10.3390/ijms242417396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Group-specific component macrophage-activating factor (GcMAF) is the vitamin D3-binding protein (DBP) deglycosylated at Thr420. The protein is believed to exhibit a wide range of therapeutic properties associated with the activation of macrophagal immunity. An original method for GcMAF production, DBP conversion to GcMAF, and the analysis of the activating potency of GcMAF was developed in this study. Data unveiling the molecular causes of macrophage activation were obtained. GcMAF was found to interact with three CLEC10A derivatives having molecular weights of 29 kDa, 63 kDa, and 65 kDa. GcMAF interacts with high-molecular-weight derivatives via Ca2+-dependent receptor engagement. Binding to the 65 kDa or 63 kDa derivative determines the pro- and anti-inflammatory direction of cytokine mRNA expression: 65 kDa-pro-inflammatory (TNF-α, IL-1β) and 63 kDa-anti-inflammatory (TGF-β, IL-10). No Ca2+ ions are required for the interaction with the canonical 29 kDa CLEC10A. Both forms, DBP protein and GcMAF, bind to the 29 kDa CLEC10A. This interaction is characterized by the stochastic mRNA synthesis of the analyzed cytokines. Ex vivo experiments have demonstrated that when there is an excess of GcMAF ligand, CLEC10A forms aggregate, and the mRNA synthesis of analyzed cytokines is inhibited. A schematic diagram of the presumable mechanism of interaction between the CLEC10A derivatives and GcMAF is provided. The principles and elements of standardizing the GcMAF preparation are elaborated.
Collapse
Affiliation(s)
- Svetlana S. Kirikovich
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Evgeniy V. Levites
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Anastasia S. Proskurina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Genrikh S. Ritter
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Sergey E. Peltek
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Asya R. Vasilieva
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Vera S. Ruzanova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Evgeniya V. Dolgova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Sofya G. Oshihmina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Alexandr V. Sysoev
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.V.S.); (D.I.K.)
| | - Danil I. Koleno
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.V.S.); (D.I.K.)
| | - Elena D. Danilenko
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Russia; (E.D.D.); (O.S.T.)
| | - Oleg S. Taranov
- State Research Center of Virology and Biotechnology “Vector”, 630559 Koltsovo, Russia; (E.D.D.); (O.S.T.)
| | - Alexandr A. Ostanin
- Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (A.A.O.); (E.R.C.)
| | - Elena R. Chernykh
- Research Institute of Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (A.A.O.); (E.R.C.)
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| | - Sergey S. Bogachev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.L.); (A.S.P.); (G.S.R.); (S.E.P.); (A.R.V.); (V.S.R.); (E.V.D.); (S.G.O.); (N.A.K.)
| |
Collapse
|
11
|
Donahue TC, Ou C, Yang Q, Flinko R, Zhang X, Zong G, Lewis GK, Wang LX. Synthetic Site-Specific Antibody-Ligand Conjugates Promote Asialoglycoprotein Receptor-Mediated Degradation of Extracellular Human PCSK9. ACS Chem Biol 2023; 18:1611-1623. [PMID: 37368876 PMCID: PMC10530246 DOI: 10.1021/acschembio.3c00229] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Targeted degradation using cell-specific lysosome targeting receptors is emerging as a new therapeutic strategy for the elimination of disease-associated proteins. The liver-specific human asialoglycoprotein receptor (ASGPR) is a particularly attractive lysosome targeting receptor leveraged for targeted protein degradation (TPD). However, the efficiency of different glycan ligands for ASGPR-mediated lysosomal delivery remains to be further characterized. In this study, we applied a chemoenzymatic Fc glycan remodeling method to construct an array of site-specific antibody-ligand conjugates carrying natural bi- and tri-antennary N-glycans as well as synthetic tri-GalNAc ligands. Alirocumab, an anti-PCSK9 (proprotein convertase subtilisin/kexin type 9) antibody, and cetuximab (an anti-EGFR antibody) were chosen to demonstrate the ASGPR-mediated degradation of extracellular and membrane-associated proteins, respectively. It was found that the nature of the glycan ligands and the length of the spacer in the conjugates are critical for the receptor binding and the receptor-mediated degradation of PCSK9, which blocks low-density lipoprotein receptor (LDLR) function and adversely affects clearance of low-density lipoprotein cholesterol. Interestingly, the antibody-tri-GalNAc conjugates showed a clear hook effect for its binding to ASGPR, while antibody conjugates carrying the natural N-glycans did not. Both the antibody-tri-antennary N-glycan conjugate and the antibody-tri-GalNAc conjugate could significantly decrease extracellular PCSK9, as shown in the cell-based assays. However, the tri-GalNAc conjugate showed a clear hook effect in the receptor-mediated degradation of PCSK9, while the antibody conjugate carrying the natural N-glycans did not. The cetuximab-tri-GalNAc conjugates also showed a similar hook effect on degradation of the membrane-associated protein, epidermal growth factor receptor (EGFR). These results suggest that the two types of ligands may involve a distinct mode of interactions in the receptor binding and target-degradation processes. Interestingly, the alirocumab-tri-GalNAc conjugate was also found to upregulate LDLR levels in comparison with the antibody alone. This study showcases the potential of the targeted degradation strategy against PCSK9 for reducing low-density lipoprotein cholesterol, a risk factor for heart disease and stroke.
Collapse
Affiliation(s)
- Thomas C Donahue
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Chong Ou
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Qiang Yang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Robin Flinko
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Xiao Zhang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Guanghui Zong
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - George K Lewis
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
12
|
Leusmann S, Ménová P, Shanin E, Titz A, Rademacher C. Glycomimetics for the inhibition and modulation of lectins. Chem Soc Rev 2023; 52:3663-3740. [PMID: 37232696 PMCID: PMC10243309 DOI: 10.1039/d2cs00954d] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 05/27/2023]
Abstract
Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.
Collapse
Affiliation(s)
- Steffen Leusmann
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Elena Shanin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
13
|
Chia S, Tay SJ, Song Z, Yang Y, Walsh I, Pang KT. Enhancing pharmacokinetic and pharmacodynamic properties of recombinant therapeutic proteins by manipulation of sialic acid content. Biomed Pharmacother 2023; 163:114757. [PMID: 37087980 DOI: 10.1016/j.biopha.2023.114757] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023] Open
Abstract
The circulatory half-life of recombinant therapeutic proteins is an important pharmacokinetic attribute because it determines the dosing frequency of these drugs, translating directly to treatment cost. Thus, recombinant therapeutic glycoproteins such as monoclonal antibodies have been chemically modified by various means to enhance their circulatory half-life. One approach is to manipulate the N-glycan composition of these agents. Among the many glycan constituents, sialic acid (specifically, N-acetylneuraminic acid) plays a critical role in extending circulatory half-life by masking the terminal galactose that would otherwise be recognised by the hepatic asialoglycoprotein receptor (ASGPR), resulting in clearance of the biotherapeutic from the circulation. This review aims to provide an illustrative overview of various strategies to enhance the pharmacokinetic/pharmacodynamic properties of recombinant therapeutic proteins through manipulation of their sialic acid content.
Collapse
Affiliation(s)
- Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Shi Jie Tay
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Zhiwei Song
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore.
| | - Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A⁎STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore; School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, 62 Nanyang Drive, N1.2-B3, 637459, Singapore.
| |
Collapse
|
14
|
Kandasamy P, Mori S, Matsuda S, Erande N, Datta D, Willoughby JLS, Taneja N, O'Shea J, Bisbe A, Manoharan RM, Yucius K, Nguyen T, Indrakanti R, Gupta S, Gilbert JA, Racie T, Chan A, Liu J, Hutabarat R, Nair JK, Charisse K, Maier MA, Rajeev KG, Egli M, Manoharan M. Metabolically Stable Anomeric Linkages Containing GalNAc-siRNA Conjugates: An Interplay among ASGPR, Glycosidase, and RISC Pathways. J Med Chem 2023; 66:2506-2523. [PMID: 36757090 DOI: 10.1021/acs.jmedchem.2c01337] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Conjugation of synthetic triantennary N-acetyl-d-galactosamine (GalNAc) to small interfering RNA (siRNA) mediates binding to the asialoglycoprotein receptor (ASGPR) on the surface of hepatocytes, facilitating liver-specific uptake and siRNA-mediated gene silencing. The natural β-glycosidic bond of the GalNAc ligand is rapidly cleaved by glycosidases in vivo. Novel GalNAc ligands with S-, and C-glycosides with both α- and β-anomeric linkages, N-glycosides with β-anomeric linkage, and the O-glycoside with α-anomeric linkage were synthesized and conjugated to siRNA either on-column during siRNA synthesis or through a high-throughput, post-synthetic method. Unlike natural GalNAc, modified ligands were resistant to glycosidase activity. The siRNAs conjugated to newly designed ligands had similar affinities for ASGPR and similar silencing activity in mice as the parent GalNAc-siRNA conjugate. These data suggest that other factors, such as protein-nucleic acid interactions and loading of the antisense strand into the RNA-induced silencing complex (RISC), are more critical to the duration of action than the stereochemistry and stability of the anomeric linkage between the GalNAc moiety of the ligand conjugated to the sense strand of the siRNA.
Collapse
Affiliation(s)
| | - Shohei Mori
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Shigeo Matsuda
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Namrata Erande
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Dhrubajyoti Datta
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | | | - Nate Taneja
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Jonathan O'Shea
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Anna Bisbe
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Rajar M Manoharan
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Kristina Yucius
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Tuyen Nguyen
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Ramesh Indrakanti
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Swati Gupta
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Jason A Gilbert
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Tim Racie
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Amy Chan
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Ju Liu
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Renta Hutabarat
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Jayaprakash K Nair
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Klaus Charisse
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | - Martin A Maier
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| | | | - Martin Egli
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Muthiah Manoharan
- Alnylam Pharmaceuticals, Inc., Cambridge, Massachusetts 02142, United States
| |
Collapse
|
15
|
Kumar V, Turnbull WB. Targeted delivery of oligonucleotides using multivalent protein-carbohydrate interactions. Chem Soc Rev 2023; 52:1273-1287. [PMID: 36723021 PMCID: PMC9940626 DOI: 10.1039/d2cs00788f] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Indexed: 02/02/2023]
Abstract
Cell surface protein-carbohydrate interactions are essential for tissue-specific recognition and endocytosis of viruses, some bacteria and their toxins, and many glycoproteins. Often protein-carbohydrate interactions are multivalent - multiple copies of glycans bind simultaneously to multimeric receptors. Multivalency enhances both affinity and binding specificity, and is of interest for targeted delivery of drugs to specific cell types. The first such example of carbohydrate-mediated drug delivery to reach the clinic is Givosiran, a small interfering ribonucleic acid (siRNA) that is conjugated to a trivalent N-acetylgalactosamine (GalNAc) ligand. This ligand enables efficient uptake of the nucleic acid by the asialoglycoprotein receptor (ASGP-R) on hepatocytes. Synthetic multivalent ligands for ASGP-R were among the first 'cluster glycosides' developed at the birth of multivalent glycoscience around 40 years ago. In this review we trace the history of 'GalNAc targeting' from early academic studies to current pharmaceuticals and consider what other opportunities could follow the success of this delivery technology.
Collapse
Affiliation(s)
- Vajinder Kumar
- Department of Chemistry, Akal University, Talwandi Sabo, Bathinda, Punjab, India.
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
16
|
Ebenezer O, Comoglio P, Wong GKS, Tuszynski JA. Development of Novel siRNA Therapeutics: A Review with a Focus on Inclisiran for the Treatment of Hypercholesterolemia. Int J Mol Sci 2023; 24:4019. [PMID: 36835426 PMCID: PMC9966809 DOI: 10.3390/ijms24044019] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
Over the past two decades, it was discovered that introducing synthetic small interfering RNAs (siRNAs) into the cytoplasm facilitates effective gene-targeted silencing. This compromises gene expression and regulation by repressing transcription or stimulating sequence-specific RNA degradation. Substantial investments in developing RNA therapeutics for disease prevention and treatment have been made. We discuss the application to proprotein convertase subtilisin/kexin type 9 (PCSK9), which binds to and degrades the low-density lipoprotein cholesterol (LDL-C) receptor, interrupting the process of LDL-C uptake into hepatocytes. PCSK9 loss-of-function modifications show significant clinical importance by causing dominant hypocholesterolemia and lessening the risk of cardiovascular disease (CVD). Monoclonal antibodies and small interfering RNA (siRNA) drugs targeting PCSK9 are a significant new option for managing lipid disorders and improving CVD outcomes. In general, monoclonal antibodies are restricted to binding with cell surface receptors or circulating proteins. Similarly, overcoming the intracellular and extracellular defenses that prevent exogenous RNA from entering cells must be achieved for the clinical application of siRNAs. N-acetylgalactosamine (GalNAc) conjugates are a simple solution to the siRNA delivery problem that is especially suitable for treating a broad spectrum of diseases involving liver-expressed genes. Inclisiran is a GalNAc-conjugated siRNA molecule that inhibits the translation of PCSK9. The administration is only required every 3 to 6 months, which is a significant improvement over monoclonal antibodies for PCSK9. This review provides an overview of siRNA therapeutics with a focus on detailed profiles of inclisiran, mainly its delivery strategies. We discuss the mechanisms of action, its status in clinical trials, and its prospects.
Collapse
Affiliation(s)
- Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa
| | - Pietro Comoglio
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, 10129 Turin, Italy
| | - Gane Ka-Shu Wong
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jack A. Tuszynski
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, 10129 Turin, Italy
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
17
|
Gupta A, Gupta GS. Applications of mannose-binding lectins and mannan glycoconjugates in nanomedicine. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2022; 24:228. [PMID: 36373057 PMCID: PMC9638366 DOI: 10.1007/s11051-022-05594-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/12/2022] [Indexed: 06/01/2023]
Abstract
UNLABELLED Glycosylated nanoparticles (NPs) have drawn a lot of attention in the biomedical field over the past few decades, particularly in applications like targeted drug delivery. Mannosylated NPs and mannan-binding lectins/proteins (MBL/MBP) are emerging as promising tools for delivery of drugs, medicines, and enzymes to targeted tissues and cells as nanocarriers, enhancing their therapeutic benefits while avoiding the adverse effects of the drug. The occurrence of plenty of lectin receptors and their mannan ligands on cell surfaces makes them multifaceted carriers appropriate for specific delivery of bioactive drug materials to their targeted sites. Thus, the present review describes the tethering of mannose (Man) to several nanostructures, like micelles, liposomes, and other NPs, applicable for drug delivery systems. Bioadhesion through MBL-like receptors on cells has involvements applicable to additional arenas of science, for example gene delivery, tissue engineering, biomaterials, and nanotechnology. This review also focuses on the role of various aspects of drug/antigen delivery using (i) mannosylated NPs, (ii) mannosylated lectins, (iii) amphiphilic glycopolymer NPs, and (iv) natural mannan-containing polysaccharides, with most significant applications of MBL-based NPs as multivalent scaffolds, using different strategies. GRAPHICAL ABSTRACT Mannosylated NPs and/or MBL/MBP are coming up as viable and versatile tools as nanocarriers to deliver drugs and enzymes precisely to their target tissues or cells. The presence of abundant number of lectin receptors and their mannan ligands on cell surfaces makes them versatile carriers suitable for the targeted delivery of bioactive drugs.
Collapse
Affiliation(s)
- Anita Gupta
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| | - G. S. Gupta
- Department of Biophysics, Panjab University, Chandigarh, 160014 India
| |
Collapse
|
18
|
Shivatare SS, Shivatare VS, Wong CH. Glycoconjugates: Synthesis, Functional Studies, and Therapeutic Developments. Chem Rev 2022; 122:15603-15671. [PMID: 36174107 PMCID: PMC9674437 DOI: 10.1021/acs.chemrev.1c01032] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glycoconjugates are major constituents of mammalian cells that are formed via covalent conjugation of carbohydrates to other biomolecules like proteins and lipids and often expressed on the cell surfaces. Among the three major classes of glycoconjugates, proteoglycans and glycoproteins contain glycans linked to the protein backbone via amino acid residues such as Asn for N-linked glycans and Ser/Thr for O-linked glycans. In glycolipids, glycans are linked to a lipid component such as glycerol, polyisoprenyl pyrophosphate, fatty acid ester, or sphingolipid. Recently, glycoconjugates have become better structurally defined and biosynthetically understood, especially those associated with human diseases, and are accessible to new drug, diagnostic, and therapeutic developments. This review describes the status and new advances in the biological study and therapeutic applications of natural and synthetic glycoconjugates, including proteoglycans, glycoproteins, and glycolipids. The scope, limitations, and novel methodologies in the synthesis and clinical development of glycoconjugates including vaccines, glyco-remodeled antibodies, glycan-based adjuvants, glycan-specific receptor-mediated drug delivery platforms, etc., and their future prospectus are discussed.
Collapse
Affiliation(s)
- Sachin S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Vidya S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
19
|
Lu J, Swearingen E, Hardy M, Collins P, Wu B, Yuan E, Lu D, Li CM, Wang S, Ollmann M. RAB18 is a key regulator of GalNAc-conjugated siRNA-induced silencing in Hep3B cells. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:423-434. [PMID: 35505960 PMCID: PMC9035644 DOI: 10.1016/j.omtn.2022.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 04/01/2022] [Indexed: 12/19/2022]
Abstract
Small interfering RNA (siRNA) therapeutics have developed rapidly in recent years, despite the challenges associated with delivery of large, highly charged nucleic acids. Delivery of siRNA therapeutics to the liver has been established, with conjugation of siRNA to N-acetylgalactosamine (GalNAc) providing durable gene knockdown in hepatocytes following subcutaneous injection. GalNAc binds the asialoglycoprotein receptor (ASGPR) that is highly expressed on hepatocytes and exploits this scavenger receptor to deliver siRNA across the plasma membrane by endocytosis. However, siRNA needs to access the RNA-induced silencing complex (RISC) in the cytoplasm to provide effective gene knockdown, and the entire siRNA delivery process is very inefficient, likely because of steps required for endosomal escape, intracellular trafficking, and stability of siRNA. To reveal the cellular factors limiting delivery of siRNA therapeutics, we performed a genome-wide pooled knockout screen on the basis of delivery of GalNAc-conjugated siRNA targeting the HPRT1 gene in the human hepatocellular carcinoma line Hep3B. Our primary genome-wide pooled knockout screen identified candidate genes that when knocked out significantly enhanced siRNA efficacy in Hep3B cells. Follow-up studies indicate that knockout of RAB18 improved the efficacy of siRNA delivered by GalNAc, cholesterol, or antibodies, but not siRNA delivered by Lipofectamine transfection, suggesting a role for RAB18 in siRNA delivery and intracellular trafficking.
Collapse
|
20
|
Hu J, Zhao M, Shi Q, Li L, Yin J. A ligand-based ELISA for detection of soluble asialoglycoprotein receptor in human serum. J Carbohydr Chem 2021. [DOI: 10.1080/07328303.2021.2008952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jing Hu
- Wuxi School of Medicine, Jiangnan University, Wuxi, PR China
| | - Ming Zhao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, PR China
| | - Qimin Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, PR China
| | - Lingxin Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, PR China
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, PR China
| |
Collapse
|
21
|
Dhawan V, Joshi G, Sutariya B, Shah J, Ashtikar M, Nagarsekar K, Steiniger F, Lokras A, Fahr A, Krishnapriya M, Warawdekar U, Saraf M, Nagarsenker M. Polysaccharide conjugates surpass monosaccharide ligands in hepatospecific targeting - Synthesis and comparative in silico and in vitro assessment. Carbohydr Res 2021; 509:108417. [PMID: 34481155 DOI: 10.1016/j.carres.2021.108417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/22/2021] [Accepted: 07/31/2021] [Indexed: 12/18/2022]
Abstract
Ligands with the polysaccharide headgroups have been recently reported by our group to possess enhanced interaction with asialoglycoprotein receptor (ASGPR) in silico as compared to ligands having galactose moieties. This enhanced interaction is a result of the polymer's backbone support in anchoring the ligand in a specific orientation within the bilayer. In this paper, we have attempted to provide an in vitro proof of concept by performing a comparative evaluation of polysaccharide and monosaccharide-based ligands. Docking was performed to understand interaction with ASGPR in silico. Agarose and galactose conjugates with behenic acid were synthesized, purified, and characterized to yield biocompatible hepatospecific ligands which were incorporated into nanoliposomes. Cellular internalization of these targeted liposomes was studied using confocal microscopy and flow cytometry. The toxicity potential was assessed in vivo. Results indicated that the polysaccharide-based ligand increased cellular uptake due to better interaction with the receptor as compared to ligand bearing a single galactose group. In addition to developing novel liver targeting ligands, the study also established proof of concept that has been suggested by earlier in silico investigations. The approach can be used to design targeting ligands and develop formulations with improved targeting efficacy.
Collapse
Affiliation(s)
- V Dhawan
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India
| | - G Joshi
- CRI Lab 1, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
| | - B Sutariya
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India
| | - J Shah
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India
| | - M Ashtikar
- Department of Pharmaceutical Technology, Friedrich Schiller University of Jena, Lessingstraße 8, D-07743, Jena, Germany
| | - K Nagarsekar
- Department of Pharmaceutical Technology, Friedrich Schiller University of Jena, Lessingstraße 8, D-07743, Jena, Germany
| | - F Steiniger
- Centre for Electron Microscopy of the Medical Faculty, Friedrich Schiller University of Jena, Ziegelmühlenweg 1, 07743, Jena, Germany
| | - A Lokras
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2100, Denmark
| | - A Fahr
- Department of Pharmaceutical Technology, Friedrich Schiller University of Jena, Lessingstraße 8, D-07743, Jena, Germany
| | - M Krishnapriya
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India
| | - U Warawdekar
- CRI Lab 1, Advanced Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - M Saraf
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India
| | - M Nagarsenker
- Bombay College of Pharmacy, Kalina, Santacruz East, Mumbai, 400098, India.
| |
Collapse
|
22
|
Biessen EAL, Van Berkel TJC. N-Acetyl Galactosamine Targeting: Paving the Way for Clinical Application of Nucleotide Medicines in Cardiovascular Diseases. Arterioscler Thromb Vasc Biol 2021; 41:2855-2865. [PMID: 34645280 DOI: 10.1161/atvbaha.121.316290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
While the promise of oligonucleotide therapeutics, such as (chemically modified) ASO (antisense oligonucleotides) and short interfering RNAs, is undisputed from their introduction onwards, their unfavorable pharmacokinetics and intrinsic capacity to mobilize innate immune responses, were limiting widespread clinical use. However, these major setbacks have been tackled by breakthroughs in chemistry, stability and delivery. When aiming an intervention hepatic targets, such as lipid and sugar metabolism, coagulation, not to mention cancer and virus infection, introduction of N-acetylgalactosamine aided targeting technology has advanced the field profoundly and by now a dozen of N-acetylgalactosamine therapeutics for these indications have been approved for clinical use or have progressed to clinical trial stage 2 to 3 testing. This technology, in combination with major advances in oligonucleotide stability allows safe and durable intervention in targets that were previously deemed undruggable, such as Lp(a) and PCSK9 (proprotein convertase subtilisin/kexin type 9), at high efficacy and specificity, often with as little as 2 doses per year. Their successful use even the most visionary would not have predicted 2 decades ago. Here, we will review the evolution of N-acetylgalactosamine technology. We shall outline their fundamental design principles and merits, and their application for the delivery of oligonucleotide therapeutics to the liver. Finally, we will discuss the perspectives of N-acetylgalactosamine technology and propose directions for future research in receptor targeted delivery of these gene medicines.
Collapse
Affiliation(s)
- Erik A L Biessen
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany (E.A.L.B.).,Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands (E.A.L.B.)
| | - Theo J C Van Berkel
- Division of Biopharmaceutics, LACDR, Leiden University, the Netherlands (T.J.C.V.B.)
| |
Collapse
|
23
|
Susan-Resiga D, Girard E, Essalmani R, Roubtsova A, Marcinkiewicz J, Derbali RM, Evagelidis A, Byun JH, Lebeau PF, Austin RC, Seidah NG. Asialoglycoprotein receptor 1 is a novel PCSK9-independent ligand of liver LDLR cleaved by furin. J Biol Chem 2021; 297:101177. [PMID: 34508778 PMCID: PMC8479480 DOI: 10.1016/j.jbc.2021.101177] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 01/06/2023] Open
Abstract
The hepatic carbohydrate-recognizing asialoglycoprotein receptor (ASGR1) mediates the endocytosis/lysosomal degradation of desialylated glycoproteins following binding to terminal galactose/N-acetylgalactosamine. Human heterozygote carriers of ASGR1 deletions exhibit ∼34% lower risk of coronary artery disease and ∼10% to 14% reduction of non-HDL cholesterol. Since the proprotein convertase PCSK9 is a major degrader of the low-density lipoprotein receptor (LDLR), we investigated the degradation and functionality of LDLR and/or PCSK9 by endogenous/overexpressed ASGR1 using Western blot and immunofluorescence in HepG2-naïve and HepG2-PCSK9-knockout cells. ASGR1, like PCSK9, targets LDLR, and both independently interact with/enhance the degradation of the receptor. This lack of cooperativity between PCSK9 and ASGR1 was confirmed in livers of wildtype (WT) and Pcsk9−/− mice. ASGR1 knockdown in HepG2-naïve cells significantly increased total (∼1.2-fold) and cell-surface (∼4-fold) LDLR protein. In HepG2-PCSK9-knockout cells, ASGR1 silencing led to ∼2-fold higher levels of LDLR protein and DiI (1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate)-LDL uptake associated with ∼9-fold increased cell-surface LDLR. Overexpression of WT-ASGR1/2 primarily reduced levels of immature non-O-glycosylated LDLR (∼110 kDa), whereas the triple Ala-mutant of Gln240/Trp244/Glu253 (characterized by loss of carbohydrate binding) reduced expression of the mature form of LDLR (∼150 kDa), suggesting that ASGR1 binds the LDLR in both a sugar-dependent and -independent fashion. The protease furin cleaves ASGR1 at the RKMK103↓ motif into a secreted form, likely resulting in a loss of function on LDLR. Altogether, we demonstrate that LDLR is the first example of a liver-receptor ligand of ASGR1. We conclude that silencing of ASGR1 and PCSK9 may lead to higher LDL uptake by hepatocytes, thereby providing a novel approach to further reduce LDL cholesterol levels.
Collapse
Affiliation(s)
- Delia Susan-Resiga
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Emmanuelle Girard
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Rachid Essalmani
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Anna Roubtsova
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Jadwiga Marcinkiewicz
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Rabeb M Derbali
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Alexandra Evagelidis
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada
| | - Jae H Byun
- Division of Nephrology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Paul F Lebeau
- Division of Nephrology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, McMaster University, St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM), Affiliated to the University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
24
|
Ilkow VF, Davies AM, Dhaliwal B, Beavil AJ, Sutton BJ, McDonnell JM. Reviving lost binding sites: Exploring calcium-binding site transitions between human and murine CD23. FEBS Open Bio 2021; 11:1827-1840. [PMID: 34075727 PMCID: PMC8255853 DOI: 10.1002/2211-5463.13214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/21/2021] [Accepted: 05/31/2021] [Indexed: 11/27/2022] Open
Abstract
Immunoglobulin E (IgE) is a central regulatory and triggering molecule of allergic immune responses. IgE's interaction with CD23 modulates both IgE production and functional activities.CD23 is a noncanonical immunoglobulin receptor, unrelated to receptors of other antibody isotypes. Human CD23 is a calcium-dependent (C-type) lectin-like domain that has apparently lost its carbohydrate-binding capability. The calcium-binding site classically required for carbohydrate binding in C-type lectins is absent in human CD23 but is present in the murine molecule. To determine whether the absence of this calcium-binding site affects the structure and function of human CD23, CD23 mutant proteins with increasingly "murine-like" sequences were generated. Restoration of the calcium-binding site was confirmed by NMR spectroscopy, and structures of mutant human CD23 proteins were determined by X-ray crystallography, although no electron density for calcium was observed. This study offers insights into the evolutionary differences between murine and human CD23 and some of the functional differences between CD23 in different species.
Collapse
Affiliation(s)
- Veronica F. Ilkow
- Randall Centre for Cell & Molecular BiophysicsKing’s College LondonUK
- Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Anna M. Davies
- Randall Centre for Cell & Molecular BiophysicsKing’s College LondonUK
- Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Balvinder Dhaliwal
- Randall Centre for Cell & Molecular BiophysicsKing’s College LondonUK
- Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Andrew J. Beavil
- Randall Centre for Cell & Molecular BiophysicsKing’s College LondonUK
- Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Brian J. Sutton
- Randall Centre for Cell & Molecular BiophysicsKing’s College LondonUK
- Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - James M. McDonnell
- Randall Centre for Cell & Molecular BiophysicsKing’s College LondonUK
- Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| |
Collapse
|
25
|
Palit S, Banerjee S, Mahata T, Niyogi S, Das T, Sova Mandi C, Chakrabarti P, Dutta S. Interaction of a Triantennary Quinoline Glycoconjugate with the Asialoglycoprotein Receptor. ChemMedChem 2021; 16:2211-2216. [PMID: 33860988 DOI: 10.1002/cmdc.202100158] [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: 03/05/2021] [Revised: 04/07/2021] [Indexed: 11/11/2022]
Abstract
Targeted intracellular delivery is an efficient strategy for developing therapeutics against cancer and other intracellular infections. Nonspecific drug delivery shows limited clinical applications owing to high dosage, cytotoxicity, nonspecific action, high cost, etc. Therefore, targeted delivery of less cytotoxic drug candidates to hepatocytes through ASGPR-mediated endocytosis could be an efficient strategy to surmount the prevailing shortcomings. In the present work, the gene encoding ASGPR-H1-CRD was amplified from Huh7 cells, cloned into pET 11a vector, and the ASGPR-H1-CRD protein was expressed and purified from E. coli. A novel triantennary galactose-conjugated quinoline derivative 4 was synthesized that demonstrates 17-fold higher binding affinity to isolated ASGPR-H1-CRD protein receptor (Kd ∼54 μM) in comparison to D-galactose (Kd ∼900 μM). Moreover, micro-calorimetric studies for the interaction of glycoconjugate 4 with ASGPR protein on live hepatocytes showed notable thermal response in case of ASGPR-containing Huh7 cells, in comparison to non-ASGPR Chang cells. These results might serve as an approach towards targeted delivery of small glycoconjugates to hepatocytes.
Collapse
Affiliation(s)
- Subhadeep Palit
- Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sayanika Banerjee
- Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Tridib Mahata
- Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sougata Niyogi
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Tanusree Das
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Chandra Sova Mandi
- Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Partha Chakrabarti
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sanjay Dutta
- Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| |
Collapse
|
26
|
Gabba A, Bogucka A, Luz JG, Diniz A, Coelho H, Corzana F, Cañada FJ, Marcelo F, Murphy PV, Birrane G. Crystal Structure of the Carbohydrate Recognition Domain of the Human Macrophage Galactose C-Type Lectin Bound to GalNAc and the Tumor-Associated Tn Antigen. Biochemistry 2021; 60:1327-1336. [PMID: 33724805 DOI: 10.1021/acs.biochem.1c00009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human macrophage galactose lectin (MGL) is an endocytic type II transmembrane receptor expressed on immature monocyte-derived dendritic cells and activated macrophages and plays a role in modulating the immune system in response to infections and cancer. MGL contains an extracellular calcium-dependent (C-type) carbohydrate recognition domain (CRD) that specifically binds terminal N-acetylgalactosamine glycan residues such as the Tn and sialyl-Tn antigens found on tumor cells, as well as other N- and O-glycans displayed on certain viruses and parasites. Even though the glycan specificity of MGL is known and several binding glycoproteins have been identified, the molecular basis for substrate recognition has remained elusive due to the lack of high-resolution structures. Here we present crystal structures of the MGL CRD at near endosomal pH and in several complexes, which reveal details of the interactions with the natural ligand, GalNAc, the cancer-associated Tn-Ser antigen, and a synthetic GalNAc mimetic ligand. Like the asialoglycoprotein receptor, additional calcium atoms are present and contribute to stabilization of the MGL CRD fold. The structure provides the molecular basis for preferential binding of N-acetylgalactosamine over galactose and prompted the re-evaluation of the binding modes previously proposed in solution. Saturation transfer difference nuclear magnetic resonance data acquired using the MGL CRD and interpreted using the crystal structure indicate a single binding mode for GalNAc in solution. Models of MGL1 and MGL2, the mouse homologues of MGL, explain how these proteins might recognize LewisX and GalNAc, respectively.
Collapse
MESH Headings
- Antigens, Tumor-Associated, Carbohydrate/metabolism
- Antigens, Tumor-Associated, Carbohydrate/chemistry
- Antigens, Tumor-Associated, Carbohydrate/immunology
- Humans
- Lectins, C-Type/chemistry
- Lectins, C-Type/metabolism
- Acetylgalactosamine/metabolism
- Acetylgalactosamine/chemistry
- Crystallography, X-Ray
- Models, Molecular
- Protein Domains
- Binding Sites
- Protein Binding
- Animals
Collapse
Affiliation(s)
- Adele Gabba
- Division of Experimental Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
- School of Chemistry, National University of Ireland Galway, Galway H91 TK33, Ireland
| | - Agnieszka Bogucka
- Division of Experimental Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
- School of Chemistry, National University of Ireland Galway, Galway H91 TK33, Ireland
| | - John G Luz
- Division of Experimental Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Ana Diniz
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade de Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Helena Coelho
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade de Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Francisco Corzana
- Departamento de Química, Centro de Investigación en Síntesis Química Universidad de La Rioja, 26006 Logroño, Spain
| | - Francisco Javier Cañada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Avda Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Filipa Marcelo
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade de Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Paul V Murphy
- School of Chemistry, National University of Ireland Galway, Galway H91 TK33, Ireland
| | - Gabriel Birrane
- Division of Experimental Medicine, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
| |
Collapse
|
27
|
de la Fuente IF, Sawant SS, Tolentino MQ, Corrigan PM, Rouge JL. Viral Mimicry as a Design Template for Nucleic Acid Nanocarriers. Front Chem 2021; 9:613209. [PMID: 33777893 PMCID: PMC7987652 DOI: 10.3389/fchem.2021.613209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Therapeutic nucleic acids hold immense potential in combating undruggable, gene-based diseases owing to their high programmability and relative ease of synthesis. While the delivery of this class of therapeutics has successfully entered the clinical setting, extrahepatic targeting, endosomal escape efficiency, and subcellular localization. On the other hand, viruses serve as natural carriers of nucleic acids and have acquired a plethora of structures and mechanisms that confer remarkable transfection efficiency. Thus, understanding the structure and mechanism of viruses can guide the design of synthetic nucleic acid vectors. This review revisits relevant structural and mechanistic features of viruses as design considerations for efficient nucleic acid delivery systems. This article explores how viral ligand display and a metastable structure are central to the molecular mechanisms of attachment, entry, and viral genome release. For comparison, accounted for are details on the design and intracellular fate of existing nucleic acid carriers and nanostructures that share similar and essential features to viruses. The review, thus, highlights unifying themes of viruses and nucleic acid delivery systems such as genome protection, target specificity, and controlled release. Sophisticated viral mechanisms that are yet to be exploited in oligonucleotide delivery are also identified as they could further the development of next-generation nonviral nucleic acid vectors.
Collapse
Affiliation(s)
| | | | | | | | - Jessica L. Rouge
- Department of Chemistry, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
28
|
Yamansarov EY, Lopatukhina EV, Evteev SA, Skvortsov DA, Lopukhov AV, Kovalev SV, Vaneev AN, Shkil' DO, Akasov RA, Lobov AN, Naumenko VA, Pavlova EN, Ryabaya OO, Burenina OY, Ivanenkov YA, Klyachko NL, Erofeev AS, Gorelkin PV, Beloglazkina EK, Majouga AG. Discovery of Bivalent GalNAc-Conjugated Betulin as a Potent ASGPR-Directed Agent against Hepatocellular Carcinoma. Bioconjug Chem 2021; 32:763-781. [PMID: 33691403 DOI: 10.1021/acs.bioconjchem.1c00042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Herein, we describe the design, synthesis, and biological evaluation of novel betulin and N-acetyl-d-galactosamine (GalNAc) glycoconjugates and suggest them as targeted agents against hepatocellular carcinoma. We prepared six conjugates derived via the C-3 and C-28 positions of betulin with one or two saccharide ligands. These molecules demonstrate high affinity to the asialoglycoprotein receptor (ASGPR) of hepatocytes assessed by in silico modeling and surface plasmon resonance tests. Cytotoxicity studies in vitro revealed a bivalent conjugate with moderate activity, selectivity of action, and cytostatic properties against hepatocellular carcinoma cells HepG2. An additional investigation confirmed the specific engagement with HepG2 cells by the enhanced generation of reactive oxygen species. Stability tests demonstrated its lability to acidic media and to intracellular enzymes. Therefore, the selected bivalent conjugate represents a new potential agent targeted against hepatocellular carcinoma. Further extensive studies of the cellular uptake in vitro and the real-time microdistribution in the murine liver in vivo for fluorescent dye-labeled analogue showed its selective internalization into hepatocytes due to the presence of GalNAc ligand in comparison with reference compounds. The betulin and GalNAc glycoconjugates can therefore be considered as a new strategy for developing therapeutic agents based on natural triterpenoids.
Collapse
Affiliation(s)
- Emil Yu Yamansarov
- Lomonosov Moscow State University, Moscow 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow 119049, Russian Federation.,Bashkir State University, Ufa 450076, Russian Federation
| | | | - Sergei A Evteev
- Lomonosov Moscow State University, Moscow 119991, Russian Federation
| | | | - Anton V Lopukhov
- Lomonosov Moscow State University, Moscow 119991, Russian Federation
| | - Sergey V Kovalev
- Lomonosov Moscow State University, Moscow 119991, Russian Federation
| | - Alexander N Vaneev
- Lomonosov Moscow State University, Moscow 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow 119049, Russian Federation
| | - Dmitry O Shkil'
- Lomonosov Moscow State University, Moscow 119991, Russian Federation
| | - Roman A Akasov
- National University of Science and Technology MISiS, Moscow 119049, Russian Federation
| | - Alexander N Lobov
- Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russian Federation
| | - Victor A Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russian Federation
| | | | - Oxana O Ryabaya
- Department of Experimental Diagnostic and Tumor Therapy, N. N. Blokhin National Medical Research Center for Oncology, Moscow 115478, Russian Federation
| | - Olga Yu Burenina
- Skolkovo Institute of Science and Technology, Skolkovo 143026, Russian Federation
| | - Yan A Ivanenkov
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow 127055, Russian Federation.,Institute of Biochemistry and Genetics, Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Ufa 450054, Russian Federation
| | - Natalia L Klyachko
- Lomonosov Moscow State University, Moscow 119991, Russian Federation.,Skolkovo Institute of Science and Technology, Skolkovo 143026, Russian Federation
| | - Alexander S Erofeev
- Lomonosov Moscow State University, Moscow 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow 119049, Russian Federation
| | - Petr V Gorelkin
- Lomonosov Moscow State University, Moscow 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow 119049, Russian Federation
| | | | - Alexander G Majouga
- Lomonosov Moscow State University, Moscow 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow 119049, Russian Federation.,Dmitry Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russian Federation
| |
Collapse
|
29
|
Wang J, Zhang Z, Ai Y, Liu F, Chen MM, Liu D. Lactobionic acid-modified thymine-chitosan nanoparticles as potential carriers for methotrexate delivery. Carbohydr Res 2021; 501:108275. [PMID: 33657498 DOI: 10.1016/j.carres.2021.108275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 01/24/2023]
Abstract
In order to achieve efficient delivery of methotrexate (MTX), thymine-chitosan nanoparticles (Thy-Cs NPs) were prepared, and further decorated with lactobionic acid (LA) to obtain tumor-targeting nanoparticles (LA-Thy-Cs NPs). These nanoparticles possessed a regular spherical structure with the average size about 190-250 nm and narrow size distribution, which were kinetically stable in the physiological environment. Due to electrostatic interactions and multiple hydrogen-bonding interactions between MTX and carriers, MTX was loaded into Thy-Cs NPs with high drug loading content (~20%). MTX release from Thy-Cs NPs was significantly accelerated in the mildly acidic environment due to the destruction of two types of non-covalent interactions. In vitro cell experiments demonstrated that LA-Thy-Cs NPs could be efficiently internalized into hepatoma carcinoma cells, leading to higher cytotoxicity. Moreover, MTX-loaded LA-Thy-Cs NPs performed an enhanced growth inhibition in three-dimensional multicellular tumor spheroids. Thus, the LA decorated thymine-chitosan nanocarriers can be a promising candidate for efficient delivery of MTX.
Collapse
Affiliation(s)
- Jun Wang
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong, PR China
| | - Zongyong Zhang
- College of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, 315211, Zhejiang, PR China
| | - Yilong Ai
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, 528000, Guangdong, PR China
| | - Fang Liu
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong, PR China
| | - Min-Min Chen
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong, PR China
| | - Dahai Liu
- Department of Basic Medicine and Biomedical Engineering, School of Medicine, Foshan University, Foshan, 528000, Guangdong, PR China.
| |
Collapse
|
30
|
Raposo CD, Canelas AB, Barros MT. Human Lectins, Their Carbohydrate Affinities and Where to Find Them. Biomolecules 2021; 11:188. [PMID: 33572889 PMCID: PMC7911577 DOI: 10.3390/biom11020188] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/02/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
Lectins are a class of proteins responsible for several biological roles such as cell-cell interactions, signaling pathways, and several innate immune responses against pathogens. Since lectins are able to bind to carbohydrates, they can be a viable target for targeted drug delivery systems. In fact, several lectins were approved by Food and Drug Administration for that purpose. Information about specific carbohydrate recognition by lectin receptors was gathered herein, plus the specific organs where those lectins can be found within the human body.
Collapse
Affiliation(s)
- Cláudia D. Raposo
- LAQV-Requimte, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| | - André B. Canelas
- Glanbia-AgriChemWhey, Lisheen Mine, Killoran, Moyne, E41 R622 Tipperary, Ireland;
| | - M. Teresa Barros
- LAQV-Requimte, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| |
Collapse
|
31
|
Multivalent rubber-like RNA nanoparticles for targeted co-delivery of paclitaxel and MiRNA to silence the drug efflux transporter and liver cancer drug resistance. J Control Release 2020; 330:173-184. [PMID: 33316298 DOI: 10.1016/j.jconrel.2020.12.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Analogous to the border customs, liver mainly functions as a filter to detoxify chemicals and metabolite administered orally or intravenously. Besides, the liver cancer cells overexpress the drug exporters which cause high drug effluxion from liver cancer cells, leading to chemoresistance and a diminished chemotherapeutic effect on liver cancer. Recently, we found that RNA nanoparticles display rubber-like property that can rapidly deliver therapeutics to tumor site efficiently and the rest of the RNA nanoparticle were cleared by renal excretion within half hour after systemic injection. Therefore, we designed a new multivalent RNA nanoparticle harboring three copies of hepatocyte targeting-ligands, one copy of miR122, and 24 copies of Paclitaxel to overcome the drug effluxion and chemoresistance thus, synergistically treating HCC. The hepatocyte targeting ligands introduce tumor specificity to the RNA nanoparticles as they selectively bind and internalize into liver cancer cells. The rubber-like RNA nanoparticles allow for enhanced targeting ability to the HCC tumors. The RNA nanoparticles carrying miR122 and PTX were delivered to the liver cancer cells efficiently due to their rubber-like property to enhance their EPR as well as the receptor-mediated endocytosis by hepatocyte targeting-ligands. The miR122 efficiently silenced the drug exporters and the oncogenic proteins. The synergistic effect between miR122 and PTX was confirmed by HSA (Highest Single Agent) synergy model. IC50 was determined to be 460 nM. In vivo studies on mice xenografts revealed that the RNA nanoparticle predominantly accumulated in HCC tumor sites and efficiently inhibited the tumor growth after multiple IV injection. This demonstrates the potential of the rubber-like multivalent RNA nanoparticles to conquest the liver cancer, a currently incurable lethal disease.
Collapse
|
32
|
Petrov RA, Mefedova SR, Yamansarov EY, Maklakova SY, Grishin DA, Lopatukhina EV, Burenina OY, Lopukhov AV, Kovalev SV, Timchenko YV, Ondar EE, Ivanenkov YA, Evteev SA, Vaneev AN, Timoshenko RV, Klyachko NL, Erofeev AS, Gorelkin PV, Beloglazkina EK, Majouga AG. New Small-Molecule Glycoconjugates of Docetaxel and GalNAc for Targeted Delivery to Hepatocellular Carcinoma. Mol Pharm 2020; 18:461-468. [PMID: 33264010 DOI: 10.1021/acs.molpharmaceut.0c00980] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this work, we have developed covalent and low molecular weight docetaxel delivery systems based on conjugation with N-acetyl-d-galactosamine and studied their properties related to hepatocellular carcinoma cells. The resulting glycoconjugates have an excellent affinity to the asialoglycoprotein receptor (ASGPR) in the nanomolar range of concentrations and a high cytotoxicity level comparable to docetaxel. Likewise, we observed the 21-75-fold increase in water solubility in comparison with parent docetaxel and prodrug lability to intracellular conditions with half-life values from 25.5 to 42 h. We also found that the trivalent conjugate possessed selective toxicity against hepatoma cells vs control cell lines (20-35 times). The absence of such selectivity in the case of monovalent conjugates indicates the effect of ligand valency. Specific ASGPR-mediated cellular uptake of conjugates was proved in vitro using fluorescent-labeled analogues. In addition, we showed an enhanced generation of reactive oxygen species in the HepG2 cells, which could be inhibited by the natural ligand of ASGPR. Overall, the obtained results highlight the potential of ASGPR-directed cytostatic taxane drugs for selective therapy of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Rostislav A Petrov
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,Institute of Biochemistry and Genetics Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Oktyabrya Prospekt 71, Ufa 450054, Russian Federation
| | - Sofiia R Mefedova
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Emil Yu Yamansarov
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Svetlana Yu Maklakova
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Dmitrii A Grishin
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Elena V Lopatukhina
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Olga Y Burenina
- Skolkovo Institute of Science and Technology, 3 Nobel str., Skolkovo 143026, Russian Federation
| | - Anton V Lopukhov
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Sergey V Kovalev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Yury V Timchenko
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Evgenia E Ondar
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Yan A Ivanenkov
- Institute of Biochemistry and Genetics Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Oktyabrya Prospekt 71, Ufa 450054, Russian Federation.,Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow 141700, Russian Federation
| | - Sergei A Evteev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Alexander N Vaneev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Roman V Timoshenko
- National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Natalia L Klyachko
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,Skolkovo Institute of Science and Technology, 3 Nobel str., Skolkovo 143026, Russian Federation
| | - Alexander S Erofeev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Petr V Gorelkin
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Elena K Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Alexander G Majouga
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation.,Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russian Federation
| |
Collapse
|
33
|
Valverde P, Martínez JD, Cañada FJ, Ardá A, Jiménez-Barbero J. Molecular Recognition in C-Type Lectins: The Cases of DC-SIGN, Langerin, MGL, and L-Sectin. Chembiochem 2020; 21:2999-3025. [PMID: 32426893 PMCID: PMC7276794 DOI: 10.1002/cbic.202000238] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/19/2020] [Indexed: 12/16/2022]
Abstract
Carbohydrates play a pivotal role in intercellular communication processes. In particular, glycan antigens are key for sustaining homeostasis, helping leukocytes to distinguish damaged tissues and invading pathogens from healthy tissues. From a structural perspective, this cross-talk is fairly complex, and multiple membrane proteins guide these recognition processes, including lectins and Toll-like receptors. Since the beginning of this century, lectins have become potential targets for therapeutics for controlling and/or avoiding the progression of pathologies derived from an incorrect immune outcome, including infectious processes, cancer, or autoimmune diseases. Therefore, a detailed knowledge of these receptors is mandatory for the development of specific treatments. In this review, we summarize the current knowledge about four key C-type lectins whose importance has been steadily growing in recent years, focusing in particular on how glycan recognition takes place at the molecular level, but also looking at recent progresses in the quest for therapeutics.
Collapse
Affiliation(s)
- Pablo Valverde
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - J Daniel Martínez
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - F Javier Cañada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Avda Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Ana Ardá
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain
- Department of Organic Chemistry II, Faculty of Science and Technology, UPV-EHU, 48940, Leioa, Spain
| |
Collapse
|
34
|
Gauthier L, Chevallet M, Bulteau F, Thépaut M, Delangle P, Fieschi F, Vivès C, Texier I, Deniaud A, Gateau C. Lectin recognition and hepatocyte endocytosis of GalNAc-decorated nanostructured lipid carriers. J Drug Target 2020; 29:99-107. [PMID: 32936032 DOI: 10.1080/1061186x.2020.1806286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver is the main organ for metabolism but is also subject to various pathologies, from viral, genetic, cancer or metabolic origin. There is thus a crucial need to develop efficient liver-targeted drug delivery strategies. Asialoglycoprotein receptor (ASGPR) is a C-type lectin expressed in the hepatocyte plasma membrane that efficiently endocytoses glycoproteins exposing galactose (Gal) or N-acetylgalactosamine (GalNAc). Its targeting has been successfully used to drive the uptake of small molecules decorated with three or four GalNAc, thanks to an optimisation of their spatial arrangement. Herein, we assessed the biological properties of highly stable nanostructured lipid carriers (NLC) made of FDA-approved ingredients and formulated with increasing amounts of GalNAc. Cellular studies showed that a high density of GalNAc was required to favour hepatocyte internalisation via the ASGPR pathway. Interaction studies using surface plasmon resonance and the macrophage galactose-lectin as GalNAc-recognising lectin confirmed the need of high GalNAc density for specific recognition of these NLC. This work is the first step for the development of efficient nanocarriers for prolonged liver delivery of active compounds.
Collapse
Affiliation(s)
- Laura Gauthier
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, Grenoble, France, IRIG-SyMMES, University Grenoble Alpes, Grenoble, France.,CEA, LETI-DTBS, Univ. Grenoble Alpes, Grenoble, France
| | - Mireille Chevallet
- Univ. Grenoble Alpes, CEA, CNRS, IRIG - Laboratoire de Chimie et Biologie des Métaux, Grenoble, France, CEA, IRIG-Laboratoire de Chimie et Biologie des Métaux, University Grenoble Alpes, Grenoble, France
| | - Francois Bulteau
- Univ. Grenoble Alpes, CEA, CNRS, IRIG - Institut de Biologie Structurale, Grenoble, France, IRIG-Institut de Biologie Structurale, University Grenoble Alpes, Grenoble, France
| | - Michel Thépaut
- Univ. Grenoble Alpes, CEA, CNRS, IRIG - Institut de Biologie Structurale, Grenoble, France, IRIG-Institut de Biologie Structurale, University Grenoble Alpes, Grenoble, France
| | - Pascale Delangle
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, Grenoble, France, IRIG-SyMMES, University Grenoble Alpes, Grenoble, France
| | - Franck Fieschi
- Univ. Grenoble Alpes, CEA, CNRS, IRIG - Institut de Biologie Structurale, Grenoble, France, IRIG-Institut de Biologie Structurale, University Grenoble Alpes, Grenoble, France
| | - Corinne Vivès
- Univ. Grenoble Alpes, CEA, CNRS, IRIG - Institut de Biologie Structurale, Grenoble, France, IRIG-Institut de Biologie Structurale, University Grenoble Alpes, Grenoble, France
| | | | - Aurélien Deniaud
- Univ. Grenoble Alpes, CEA, CNRS, IRIG - Laboratoire de Chimie et Biologie des Métaux, Grenoble, France, CEA, IRIG-Laboratoire de Chimie et Biologie des Métaux, University Grenoble Alpes, Grenoble, France
| | - Christelle Gateau
- Univ. Grenoble Alpes, CEA, CNRS, IRIG, SyMMES, Grenoble, France, IRIG-SyMMES, University Grenoble Alpes, Grenoble, France
| |
Collapse
|
35
|
Debacker AJ, Voutila J, Catley M, Blakey D, Habib N. Delivery of Oligonucleotides to the Liver with GalNAc: From Research to Registered Therapeutic Drug. Mol Ther 2020; 28:1759-1771. [PMID: 32592692 PMCID: PMC7403466 DOI: 10.1016/j.ymthe.2020.06.015] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/08/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Targeted delivery of oligonucleotides to liver hepatocytes using N-acetylgalactosamine (GalNAc) conjugates that bind to the asialoglycoprotein receptor has become a breakthrough approach in the therapeutic oligonucleotide field. This technology has led to the approval of givosiran for the treatment of acute hepatic porphyria, and there are another seven conjugates in registrational review or phase 3 trials and at least another 21 conjugates at earlier stages of clinical development. This review highlights some of the recent chemical and preclinical advances in this space, leading to a large number of clinical candidates against a diverse range of targets in liver hepatocytes. The review focuses on the use of this delivery system for small interfering RNAs (siRNAs) and antisense molecules that cause downregulation of target mRNA and protein. A number of other approaches such as anti-microRNAs and small activating RNAs are starting to exploit the technology, broadening the potential of this approach for therapeutic oligonucleotide intervention.
Collapse
Affiliation(s)
- Alexandre J Debacker
- MiNA Therapeutics, Translation & Innovation Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Jon Voutila
- MiNA Therapeutics, Translation & Innovation Hub, 80 Wood Lane, London W12 0BZ, UK
| | - Matthew Catley
- MiNA Therapeutics, Translation & Innovation Hub, 80 Wood Lane, London W12 0BZ, UK
| | - David Blakey
- MiNA Therapeutics, Translation & Innovation Hub, 80 Wood Lane, London W12 0BZ, UK.
| | - Nagy Habib
- MiNA Therapeutics, Translation & Innovation Hub, 80 Wood Lane, London W12 0BZ, UK; Department of Surgery & Cancer, Hammersmith Hospital, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
36
|
ASGR1 and Its Enigmatic Relative, CLEC10A. Int J Mol Sci 2020; 21:ijms21144818. [PMID: 32650396 PMCID: PMC7404283 DOI: 10.3390/ijms21144818] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
The large family of C-type lectin (CLEC) receptors comprises carbohydrate-binding proteins that require Ca2+ to bind a ligand. The prototypic receptor is the asialoglycoprotein receptor-1 (ASGR1, CLEC4H1) that is expressed primarily by hepatocytes. The early work on ASGR1, which is highly specific for N-acetylgalactosamine (GalNAc), established the foundation for understanding the overall function of CLEC receptors. Cells of the immune system generally express more than one CLEC receptor that serve diverse functions such as pathogen-recognition, initiation of cellular signaling, cellular adhesion, glycoprotein turnover, inflammation and immune responses. The receptor CLEC10A (C-type lectin domain family 10 member A, CD301; also called the macrophage galactose-type lectin, MGL) contains a carbohydrate-recognition domain (CRD) that is homologous to the CRD of ASGR1, and thus, is also specific for GalNAc. CLEC10A is most highly expressed on immature DCs, monocyte-derived DCs, and alternatively activated macrophages (subtype M2a) as well as oocytes and progenitor cells at several stages of embryonic development. This receptor is involved in initiation of TH1, TH2, and TH17 immune responses and induction of tolerance in naïve T cells. Ligand-mediated endocytosis of CLEC receptors initiates a Ca2+ signal that interestingly has different outcomes depending on ligand properties, concentration, and frequency of administration. This review summarizes studies that have been carried out on these receptors.
Collapse
|
37
|
Bhingardeve P, Madhanagopal BR, Naick H, Jain P, Manoharan M, Ganesh K. Receptor-Specific Delivery of Peptide Nucleic Acids Conjugated to Three Sequentially Linked N-Acetyl Galactosamine Moieties into Hepatocytes. J Org Chem 2020; 85:8812-8824. [PMID: 32529829 DOI: 10.1021/acs.joc.0c00601] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peptide nucleic acids (PNAs) are DNA analogs that bind with high affinity to DNA and RNA in a sequence-specific manner but have poor cell permeability, limiting use as therapeutic agents. The work described here is motivated by recent reports of efficient gene silencing specifically in hepatocytes by small interfering RNAs conjugated to triantennary N-acetyl galactosamine (GalNAc), the ligand recognized by the asialoglycoprotein receptor (ASGPR). PNAs conjugated to either triantennary GalNAc at the N-terminus (the branched architecture) or monomeric GalNAc moieties anchored at Cγ of three consecutive PNA monomers of N-(2-aminoethyl)glycine (aeg) scaffolds (the sequential architecture) were synthesized on the solid phase. These formed duplexes with complementary DNA and RNA as shown by UV and circular dichroism spectroscopy. The fluorescently labeled analogs of GalNAc-conjugated PNAs were internalized by HepG2 cells that express the ASGPR but were not taken up by HEK-293 cells that lack this receptor. The sequential conjugate was internalized about 13-fold more efficiently than the branched conjugate into HepG2 cells, as demonstrated by confocal microscopy. The results presented here highlight the potential significance of the architecture of GalNAc conjugation for efficient uptake by target liver cells and indicate that GalNAc-conjugated PNAs have possible therapeutic applications.
Collapse
Affiliation(s)
- Pramod Bhingardeve
- Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| | - Bharath Raj Madhanagopal
- Indian Institute of Science Education and Research (IISER) Tirupati, Karkambadi Road, Tirupati 517507, Andhra Pradesh, India
| | - Hemanth Naick
- Indian Institute of Science Education and Research (IISER) Tirupati, Karkambadi Road, Tirupati 517507, Andhra Pradesh, India
| | - Prashant Jain
- Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| | - Muthiah Manoharan
- Alnylam Pharmaceuticals, Cambridge, Massachusetts 02142, United States
| | - Krishna Ganesh
- Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India.,Indian Institute of Science Education and Research (IISER) Tirupati, Karkambadi Road, Tirupati 517507, Andhra Pradesh, India
| |
Collapse
|
38
|
Reshitko GS, Yamansarov EY, Evteev SA, Lopatukhina EV, Shkil' DO, Saltykova IV, Lopukhov AV, Kovalev SV, Lobov AN, Kislyakov IV, Burenina OY, Klyachko NL, Garanina AS, Dontsova OA, Ivanenkov YA, Erofeev AS, Gorelkin PV, Beloglazkina EK, Majouga AG. Synthesis and Evaluation of New Trivalent Ligands for Hepatocyte Targeting via the Asialoglycoprotein Receptor. Bioconjug Chem 2020; 31:1313-1319. [PMID: 32379426 DOI: 10.1021/acs.bioconjchem.0c00202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the asialoglycoprotein receptor (also known as the "Ashwell-Morell receptor" or ASGPR) was discovered as the first cellular mammalian lectin, numerous drug delivery systems have been developed and several gene delivery systems associated with multivalent ligands for liver disease targeting are undergoing clinical trials. The success of these systems has facilitated the further study of new ligands with comparable or higher affinity and less synthetic complexity. Herein, we designed two novel trivalent ligands based on the esterification of tris(hydroxymethyl) aminomethane (TRIS) followed by the azide-alkyne Huisgen cycloaddition with azido N-acetyl-d-galactosamine. The presented triazolyl glycoconjugates exhibited good binding to ASGPR, which was predicted using in silico molecular docking and assessed by a surface plasmon resonance (SPR) technique. Moreover, we demonstrated the low level of in vitro cytotoxicity, as well as the optimal spatial geometry and the required amphiphilic balance, for new, easily accessible ligands. The conjugate of a new ligand with Cy5 dye exhibited selective penetration into HepG2 cells in contrast to the ASGPR-negative PC3 cell line.
Collapse
Affiliation(s)
- Galina S Reshitko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Emil Yu Yamansarov
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Sergei A Evteev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Elena V Lopatukhina
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Dmitry O Shkil'
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Irina V Saltykova
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Anton V Lopukhov
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Sergey V Kovalev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Alexander N Lobov
- Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, 450054, Russian Federation
| | - Ivan V Kislyakov
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Olga Yu Burenina
- Skolkovo Institute of Science and Technology, Skolkovo, 143026, Russian Federation
| | - Natalia L Klyachko
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,Skolkovo Institute of Science and Technology, Skolkovo, 143026, Russian Federation
| | - Anastasiia S Garanina
- National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Olga A Dontsova
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,Skolkovo Institute of Science and Technology, Skolkovo, 143026, Russian Federation
| | - Yan A Ivanenkov
- Moscow Institute of Physics and Technology (State University), Dolgoprudny City, Moscow Region 141700, Russian Federation.,Institute of Biochemistry and Genetics, Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Ufa, 450054, Russian Federation
| | - Alexander S Erofeev
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Peter V Gorelkin
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation
| | - Elena K Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Alexander G Majouga
- Chemistry Department, Lomonosov Moscow State University, Moscow, 119991, Russian Federation.,National University of Science and Technology MISiS, Moscow, 119049, Russian Federation.,Dmitry Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation
| |
Collapse
|
39
|
Raposo CD, Costa R, Petrova KT, Brito C, Scotti MT, Cardoso MM. Development of Novel Galactosylated PLGA Nanoparticles for Hepatocyte Targeting Using Molecular Modelling. Polymers (Basel) 2020; 12:E94. [PMID: 31947904 PMCID: PMC7023654 DOI: 10.3390/polym12010094] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/17/2019] [Accepted: 12/23/2019] [Indexed: 02/02/2023] Open
Abstract
Doxorubicin-loaded PLGA nanoparticles conjugated with a new galactose-based ligand for the specific recognition by human hepatoma cellular carcinoma cells (Hep G2) were successfully produced. The new targeting compound was selected using molecular docking combined with quantum chemical calculations for modelling and comparing molecular interactions among the H1 subunit of the asialoglycoprotein receptor containing the carbohydrate recognition domain and the ligand. The ligand, bis(1-O-ethyl-β-D-galactopyranosyl)amine, was synthetized, characterized, and subsequently linked to PLGA. Unloaded (PLGA-di-GAL NP) and doxorubicin-loaded (DOX-PLGA-di-GAL NP) nanoparticles were prepared using an emulsion method and characterized. The produced DOX-PLGA-di-GAL NP are spherical in shape with a size of 258 ± 47 nm, a zeta potential of -62.3 mV, and a drug encapsulation efficiency of 83%. The in vitro drug release results obtained show a three-phase release profile. In vitro cell studies confirmed the interaction between Hep G2 cells and PLGA-di-GAL NP. Cell cytotoxicity tests showed that unloaded NP are nontoxic and that DOX-PLGA-di-GAL NP caused a decrease of around 80% in cellular viability. The strategy used in this work to design new targeting compounds represents a promising tool to develop effective hepatocyte targeting drug delivery systems and can be applied to other tissues/organs.
Collapse
Affiliation(s)
- Cláudia D. Raposo
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Quinta da Torre, 2829-516 Caparica, Portugal
| | - Rita Costa
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Krasimira T. Petrova
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Quinta da Torre, 2829-516 Caparica, Portugal
| | - Catarina Brito
- IBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Marcus T. Scotti
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba Campus I, João Pessoa-PB 58051-900, Brazil
| | - M. Margarida Cardoso
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Quinta da Torre, 2829-516 Caparica, Portugal
| |
Collapse
|
40
|
Hoober JK, Eggink LL, Cote R. Stories From the Dendritic Cell Guardhouse. Front Immunol 2019; 10:2880. [PMID: 31921144 PMCID: PMC6919295 DOI: 10.3389/fimmu.2019.02880] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022] Open
Abstract
Phagocytic cells [dendritic cells (DCs), macrophages, monocytes, neutrophils, and mast cells] utilize C-type (Ca2+-dependent) lectin-like (CLEC) receptors to identify and internalize pathogens or danger signals. As monitors of environmental imbalances, CLEC receptors are particularly important in the function of DCs. Activation of the immune system requires, in sequence, presentation of antigen to the T cell receptor (TCR) by DCs, interaction of co-stimulatory factors such as CD40/80/86 on DCs with CD40L and CD28 on T cells, and production of IL-12 and/or IFN-α/β to amplify T cell differentiation and expansion. Without this sequence of events within an inflammatory environment, or in a different order, antigen-specific T cells become unresponsive, are deleted or become regulatory T cells. Thus, the mode by which CLEC receptors on DCs are engaged can either elicit activation of T cells to achieve an immune response or induce tolerance. This minireview illustrates these aspects with Dectin-1, DEC205, the mannose receptor and CLEC10A as examples.
Collapse
Affiliation(s)
| | | | - Robert Cote
- Susavion Biosciences, Inc., Tempe, AZ, United States
| |
Collapse
|
41
|
|
42
|
Diniz A, Coelho H, Dias JS, Vliet SJ, Jiménez‐Barbero J, Corzana F, Cabrita EJ, Marcelo F. The Plasticity of the Carbohydrate Recognition Domain Dictates the Exquisite Mechanism of Binding of Human Macrophage Galactose‐Type Lectin. Chemistry 2019; 25:13945-13955. [DOI: 10.1002/chem.201902780] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/01/2019] [Indexed: 01/07/2023]
Affiliation(s)
- Ana Diniz
- UCIBIO, REQUIMTEDepartamento de QuímicaFaculdade de Ciências e TecnologiaUniversidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Helena Coelho
- UCIBIO, REQUIMTEDepartamento de QuímicaFaculdade de Ciências e TecnologiaUniversidade NOVA de Lisboa 2829-516 Caparica Portugal
- CIC bioGUNEBizkaia Technology Park, Building 801A 48170 Derio Spain
- Departament of Organic Chemistry IIFaculty of Science & TechnologyUniversity of the Basque Country, Leioa 48940 Bizkaia Spain
| | - Jorge S. Dias
- UCIBIO, REQUIMTEDepartamento de QuímicaFaculdade de Ciências e TecnologiaUniversidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Sandra J. Vliet
- Department of Molecular Cell Biology and ImmunologyAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit Amsterdam 1007MB Amsterdam the Netherlands
| | - Jesús Jiménez‐Barbero
- CIC bioGUNEBizkaia Technology Park, Building 801A 48170 Derio Spain
- Departament of Organic Chemistry IIFaculty of Science & TechnologyUniversity of the Basque Country, Leioa 48940 Bizkaia Spain
- IkerbasqueBasque Foundation for Science Maria Diaz de Haro 13 48009 Bilbao Spain
| | - Francisco Corzana
- Departamento de QuímicaCentro de Investigación en Síntesis QuímicaUniversidad de La Rioja 26006 Logroño Spain
| | - Eurico J. Cabrita
- UCIBIO, REQUIMTEDepartamento de QuímicaFaculdade de Ciências e TecnologiaUniversidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Filipa Marcelo
- UCIBIO, REQUIMTEDepartamento de QuímicaFaculdade de Ciências e TecnologiaUniversidade NOVA de Lisboa 2829-516 Caparica Portugal
| |
Collapse
|
43
|
Shinchi H, Nakamura T, Ota H, Nishihara S, Wakao M, Suda Y. Cell Profiling Based on Sugar‐Chain–Cell Binding Interaction and Its Application to Typing and Quality Verification of Cells. Chembiochem 2019; 20:1810-1816. [DOI: 10.1002/cbic.201900028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Hiroyuki Shinchi
- Graduate School of Science and Engineering Kagoshima University 1-21-40 Kohrimoto Kagoshima 890-0065 Japan
| | - Tomoya Nakamura
- Graduate School of Science and Engineering Kagoshima University 1-21-40 Kohrimoto Kagoshima 890-0065 Japan
| | - Hayato Ota
- Graduate School of Engineering Soka University, 1-236 Tangi-machi Hachioji Tokyo 192-8577 Japan
| | - Shoko Nishihara
- Graduate School of Engineering Soka University, 1-236 Tangi-machi Hachioji Tokyo 192-8577 Japan
| | - Masahiro Wakao
- Graduate School of Science and Engineering Kagoshima University 1-21-40 Kohrimoto Kagoshima 890-0065 Japan
| | - Yasuo Suda
- Graduate School of Science and Engineering Kagoshima University 1-21-40 Kohrimoto Kagoshima 890-0065 Japan
- SUDx-Biotec Corporation 1-42-1 Shiroyama Kagoshima 890-0013 Japan
| |
Collapse
|
44
|
Andersen VL, Vinther M, Kumar R, Ries A, Wengel J, Nielsen JS, Kjems J. A self-assembled, modular nucleic acid-based nanoscaffold for multivalent theranostic medicine. Am J Cancer Res 2019; 9:2662-2677. [PMID: 31131060 PMCID: PMC6525989 DOI: 10.7150/thno.32060] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/21/2019] [Indexed: 12/20/2022] Open
Abstract
Rationale: Within the field of personalized medicine there is an increasing focus on designing flexible, multifunctional drug delivery systems that combine high efficacy with minimal side effects, by tailoring treatment to the individual. Methods: We synthesized a chemically stabilized ~4 nm nucleic acid nanoscaffold, and characterized its assembly, stability and functional properties in vitro and in vivo. We tested its flexibility towards multifunctionalization by conjugating various biomolecules to the four modules of the system. The pharmacokinetics, targeting capability and bioimaging properties of the structure were investigated in mice. The role of avidity in targeted liver cell internalization was investigated by flow cytometry, confocal microscopy and in vivo by fluorescent scanning of the blood and organs of the animals. Results: We have developed a nanoscaffold that rapidly and with high efficiency can self-assemble four chemically conjugated functionalities into a stable, in vivo-applicable system with complete control of stoichiometry and site specificity. The circulation time of the nanoscaffold could be tuned by functionalization with various numbers of polyethylene glycol polymers or with albumin-binding fatty acids. Highly effective hepatocyte-specific internalization was achieved with increasing valencies of tri-antennary galactosamine (triGalNAc) in vitro and in vivo. Conclusion: With its facile functionalization, stoichiometric control, small size and high serum- and thermostability, the nanoscaffold presented here constitutes a novel and flexible platform technology for theranostics.
Collapse
|
45
|
Seth PP, Tanowitz M, Bennett CF. Selective tissue targeting of synthetic nucleic acid drugs. J Clin Invest 2019; 129:915-925. [PMID: 30688661 DOI: 10.1172/jci125228] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Antisense oligonucleotides (ASOs) are chemically synthesized nucleic acid analogs designed to bind to RNA by Watson-Crick base pairing. Following binding to the targeted RNA, the ASO perturbs RNA function by promoting selective degradation of the targeted RNA, altering RNA intermediary metabolism, or disrupting function of the RNA. Most antisense drugs are chemically modified to enhance their pharmacological properties and for passive targeting of the tissues of therapeutic interest. Recent advances in selective tissue targeting have resulted in a newer generation of ASO drugs that are more potent and better tolerated than previous generations, spawning renewed interest in identifying selective ligands that enhance targeted delivery of ASOs to tissues.
Collapse
|
46
|
Ruvinsky AM, Aloni I, Cappel D, Higgs C, Marshall K, Rotkiewicz P, Repasky M, Feher VA, Feyfant E, Hessler G, Matter H. The Role of Bridging Water and Hydrogen Bonding as Key Determinants of Noncovalent Protein-Carbohydrate Recognition. ChemMedChem 2018; 13:2684-2693. [DOI: 10.1002/cmdc.201800437] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/21/2018] [Indexed: 11/08/2022]
Affiliation(s)
| | - Ishita Aloni
- Schrödinger, Inc.; 120 West 45th Street New York NY 10036 USA
| | | | - Chris Higgs
- Schrödinger, Inc.; 10201 Wateridge Circle, Suite 220 San Diego CA 92121 USA
| | - Kyle Marshall
- Schrödinger, Inc.; 101 SW Main Street Portland OR 97204 USA
| | - Piotr Rotkiewicz
- Schrödinger, Inc.; 222 Third Street, Suite 2230 Cambridge MA 02142 USA
| | - Matt Repasky
- Schrödinger, Inc.; 101 SW Main Street Portland OR 97204 USA
| | - Victoria A. Feher
- Schrödinger, Inc.; 10201 Wateridge Circle, Suite 220 San Diego CA 92121 USA
| | - Eric Feyfant
- Schrödinger, Inc.; 222 Third Street, Suite 2230 Cambridge MA 02142 USA
| | - Gerhard Hessler
- Sanofi-Aventis (Deutschland) GmbH; Integrated Drug Discovery (IDD), Synthetic Molecular Design, Building G838; Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Hans Matter
- Sanofi-Aventis (Deutschland) GmbH; Integrated Drug Discovery (IDD), Synthetic Molecular Design, Building G838; Industriepark Höchst 65926 Frankfurt am Main Germany
| |
Collapse
|
47
|
Yan J, Chen L, Liu Z, Chen Y, Sun Y, Han J, Feng L. The D5 region of the intelectin domain is a new type of carbohydrate recognition domain in the intelectin gene family. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 85:150-160. [PMID: 29621532 DOI: 10.1016/j.dci.2018.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/05/2018] [Accepted: 02/28/2018] [Indexed: 06/08/2023]
Abstract
Intelectin is a recently characterized soluble galactofuranose-binding lectin that exists in species ranging from amphioxus to human. Interestingly, intelectin does not contain a canonical carbohydrate-recognition domain (CRD). Therefore, we designed serial deletions of intelectin in the Chinese amphioxus (Branchiostoma belcheri tsingtauense, AmphiITLN71469) in order to identify functional regions required for carbohydrate binding. Our results revealed that Domain 5 (aa 203-302) was able to bind lipopolysaccarides (LPS) or peptidoglycan (PGN) and agglutinate bacteria as efficiently as the full-length protein. Three dimensional (3D) atomic models of Domain 5 were generated by ab initio based program QUARK and by Iterative Threading Assembly Refinement (I-TASSER) programs, in which four amino acids mediating calcium-binding (G54-G55-G56-E91) were identified by hemagglutination assay. Furthermore, a striking functional conservation of Domain 5 was detected in zebrafish intelectin 1. Taken together, our findings identified for the first time a new CRD domain in intelectin, thereby providing new knowledge leading to a better understanding of pathogen-host interactions.
Collapse
Affiliation(s)
- Jie Yan
- Marine Biotechnology Research Center, Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China
| | - Lei Chen
- Marine Biotechnology Research Center, Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Zhuang Liu
- Department of Reproductive Medicine, Affiliated Hospital of Jining Medical College Jining, Shandong, China
| | - Yonglin Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Ying Sun
- Department of Anesthesiology, Affiliated Hospital of Jining Medical College, Jining, Shandong, China
| | - Jia Han
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Street, Jinan, 250021, China.
| | - Lijun Feng
- Marine Biotechnology Research Center, Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
48
|
Springer AD, Dowdy SF. GalNAc-siRNA Conjugates: Leading the Way for Delivery of RNAi Therapeutics. Nucleic Acid Ther 2018; 28:109-118. [PMID: 29792572 PMCID: PMC5994659 DOI: 10.1089/nat.2018.0736] [Citation(s) in RCA: 457] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/15/2022] Open
Abstract
Short-interfering RNA (siRNA)-induced RNAi responses have great potential to treat a wide variety of human diseases from cancer to pandemic viral outbreaks to Parkinson's Disease. However, before siRNAs can become drugs, they must overcome a billion years of evolutionary defenses designed to keep invading RNAs on the outside cells from getting to the inside of cells. Not surprisingly, significant effort has been placed in developing a wide array of delivery technologies. Foremost of these has been the development of N-acetylgalactosamine (GalNAc) siRNA conjugates for delivery to liver. Tris-GalNAc binds to the Asialoglycoprotein receptor that is highly expressed on hepatocytes resulting in rapid endocytosis. While the exact mechanism of escape across the endosomal lipid bilayer membrane remains unknown, sufficient amounts of siRNAs enter the cytoplasm to induce robust, target selective RNAi responses in vivo. Multiple GalNAc-siRNA conjugate clinical trials, including two phase III trials, are currently underway by three biotech companies to treat a wide variety of diseases. GalNAc-siRNA conjugates are a simple solution to the siRNA delivery problem for liver hepatocytes and have shown the RNAi (and antisense oligonucleotide) field the path forward for targeting other tissue types.
Collapse
Affiliation(s)
- Aaron D Springer
- Department of Cellular and Molecular Medicine, University of California San Diego , La Jolla, California
| | - Steven F Dowdy
- Department of Cellular and Molecular Medicine, University of California San Diego , La Jolla, California
| |
Collapse
|
49
|
Eggink LL, Roby KF, Cote R, Kenneth Hoober J. An innovative immunotherapeutic strategy for ovarian cancer: CLEC10A and glycomimetic peptides. J Immunother Cancer 2018; 6:28. [PMID: 29665849 PMCID: PMC5905120 DOI: 10.1186/s40425-018-0339-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/03/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Receptors specific for the sugar N-acetylgalactosamine (GalNAc) include the human type II, C-type lectin receptor macrophage galactose-type lectin/C-type lectin receptor family member 10A (MGL/CLEC10A/CD301) that is expressed prominently by human peripheral immature dendritic cells, dendritic cells in the skin, alternatively-activated (M2a) macrophages, and to lesser extents by several other types of tissues. CLEC10A is an endocytic receptor on antigen-presenting cells and has been proposed to play an important role in maturation of dendritic cells and initiation of an immune response. In this study, we asked whether a peptide that binds in the GalNAc-binding site of CLEC10A would serve as an effective tool to activate an immune response against ovarian cancer. METHODS A 12-mer sequence emerged from a screen of a phage display library with a GalNAc-specific lectin. The peptide, designated svL4, and a shorter peptide consisting of the C-terminal 6 amino acids, designated sv6D, were synthesized as tetravalent structures based on a tri-lysine core. In silico and in vitro binding assays were developed to evaluate binding of the peptides to GalNAc-specific receptors. Endotoxin-negative peptide solutions were administered by subcutaneous injection and biological activity of the peptides was determined by secretion of cytokines and the response of peritoneal immune cells in mice. Anti-cancer activity was studied in a murine model of ovarian cancer. RESULTS The peptides bound to recombinant human CLEC10A with high avidity, with half-maximal binding in the low nanomolar range. Binding to the receptor was Ca2+-dependent. Subcutaneous injection of low doses of peptides into mice on alternate days resulted in several-fold expansion of populations of mature immune cells within the peritoneal cavity. Peptide sv6D effectively suppressed development of ascites in a murine ovarian cancer model as a monotherapy and in combination with the chemotherapeutic drug paclitaxel or the immunotherapeutic antibody against the receptor PD-1. Toxicity, including antigenicity and release of cytotoxic levels of cytokines, was not observed. CONCLUSION sv6D is a functional ligand for CLEC10A and induces maturation of immune cells in the peritoneal cavity. The peptide caused a highly significant extension of survival of mice with implanted ovarian cancer cells with a favorable toxicity and non-antigenic profile.
Collapse
Affiliation(s)
- Laura L Eggink
- Susavion Biosciences, Inc., 1615 W. University Drive, Suite 132, Tempe, AZ, 85281, USA
| | | | - Robert Cote
- Susavion Biosciences, Inc., 1615 W. University Drive, Suite 132, Tempe, AZ, 85281, USA
| | - J Kenneth Hoober
- Susavion Biosciences, Inc., 1615 W. University Drive, Suite 132, Tempe, AZ, 85281, USA.
| |
Collapse
|
50
|
Yang D, Zheng X, Wang N, Fan S, Yang Y, Lu Y, Chen Q, Liu X, Zheng J. Kukoamine B promotes TLR4-independent lipopolysaccharide uptake in murine hepatocytes. Oncotarget 2018; 7:57498-57513. [PMID: 27542278 PMCID: PMC5295368 DOI: 10.18632/oncotarget.11292] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 08/11/2016] [Indexed: 12/30/2022] Open
Abstract
Free bacterial lipopolysaccharide (LPS) is generally removed from the bloodstream through hepatic uptake via TLR4, the LPS pattern recognition receptor, but mechanisms for internalization and clearance of conjugated LPS are less clear. Kukoamine B (KB) is a novel cationic alkaloid that interferes with LPS binding to TLR4. In this study, KB accelerated blood clearance of LPS. KB also enhanced LPS distribution in the hepatic tissues of C57 BL/6 mice, along with LPS uptake in primary hepatocytes and HepG2 cells. By contrast, KB inhibited LPS internalization in Kupffer and RAW 264.7 cells. Loss of TLR4 did not affect LPS uptake into KB-treated hepatocytes. We also detected selective upregulation of the asialoglycoprotein receptor (ASGPR) upon KB treatment, and ASGPR colocalized with KB in cultured hepatocytes. Molecular docking showed that KB bound to ASGPR in a manner similar to GalNAc, a known ASGPR agonist. GalNAc dose-dependently reduced KB internalization, suggesting it competes with KB for ASGPR binding, and ASGPR knockdown also impaired LPS uptake into hepatocytes. Finally, while KB enhanced LPS uptake, it was protective against LPS-induced inflammation and hepatocyte injury. Our study provides a new mechanism for conjugated LPS hepatic uptake induced by the LPS neutralizer KB and mediated by membrane ASGPR binding.
Collapse
Affiliation(s)
- Dong Yang
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xinchuan Zheng
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ning Wang
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shijun Fan
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yongjun Yang
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yongling Lu
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qian Chen
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xin Liu
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiang Zheng
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|