1
|
Wang S, Wang P, Zhang M, Song X, Wei L, Ma X, Yao X, Zhang S, Chen Z, Zheng Y, Yu R, Zhang C. Blocking GP130 binding in interleukin-11 through site-specific PEGylation attenuates bleomycin-induced pulmonary fibrosis in mice. Int J Pharm 2024; 667:124916. [PMID: 39528144 DOI: 10.1016/j.ijpharm.2024.124916] [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: 06/26/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Recent insights have identified interleukin-11 (IL-11) as a pivotal profibrotic cytokine, with its signaling through IL-11Rα and GP130 receptors emerging as a promising therapeutic target for fibrotic diseases. Herein, we developed receptor-biased IL-11 via site-specific PEGylation at the GP130 binding interface, aiming to explore its therapeutic potential for bleomycin-induced pulmonary fibrosis in mice. By conducting single site-directed cysteine mutagenesis at site II or site III of IL-11, we refined the conjugation site, demonstrating that mutation at site III exhibits heightened sensitivity to GP130 binding and signaling. Cysteine-based PEGylation substantially attenuated the ability of GP130 to bind to IL-11 W147C, while almost entirely preserving its IL-11Rα binding ability. These PEGylated IL-11 W147C analogs showed potent inhibition of TF-1 cell proliferation and significant antagonism to TGF-β1-induced human lung fibroblasts (HLFs) differentiation into myofibroblasts. Moreover, PEGylation significantly prolonged the half-life of IL-11 W147C in healthy rats. Subcutaneous administration of PEGylated analogs, particularly PEG20/40 k-IL-11 W147C, effectively mitigated extracellular matrix deposition, preserved alveolar architecture, and attenuated the progression of pulmonary fibrosis in mice. The finding of this study not only underscores the therapeutic potential of IL-11 modulation, but also provides a general strategy for the design of cytokine-based biased antagonists and agonists targeting these multifaceted signaling pathways.
Collapse
Affiliation(s)
- Sa Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Pengyu Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Minhui Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaotong Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Long Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xinyuan Ma
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xin Yao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuwen Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zijie Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yongxiang Zheng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rong Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chun Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Biopharmaceutics, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Kabil MF, Azzazy HMES, Nasr M. Recent progress on polySarcosine as an alternative to PEGylation: Synthesis and biomedical applications. Int J Pharm 2024; 653:123871. [PMID: 38301810 DOI: 10.1016/j.ijpharm.2024.123871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Biotherapeutic PEGylation to prolong action of medications has gained popularity over the last decades. Various hydrophilic natural polymers have been developed to tackle the drawbacks of PEGylation, such as its accelerated blood clearance and non-biodegradability. Polypeptoides, such as polysarcosine (pSar), have been explored as hydrophilic substitutes for PEG. pSar has PEG-like physicochemical characteristics such as water solubility and no reported cytotoxicity and immunogenicity. This review discusses pSar derivatives, synthesis, characterization approaches, biomedical applications, in addition to the challenges and future perspectives of pSar based biomaterials as an alternative to PEG.
Collapse
Affiliation(s)
- Mohamed Fawzi Kabil
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, AUC Avenue, New Cairo 11835, Egypt
| | - Hassan Mohamed El-Said Azzazy
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, AUC Avenue, New Cairo 11835, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
3
|
Qi J, Guo Z, Zhu S, Jiang X, Wu Y, Chen Y, Hu F, Xiong J, Wu Y, Ye X, Liang X. Therapeutic effect of long-acting FGF21 with controlled site-specific modification on nonalcoholic steatohepatitis. Int J Biol Macromol 2024; 261:129797. [PMID: 38290625 DOI: 10.1016/j.ijbiomac.2024.129797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
FGF21 plays an active role in the treatment of type 2 diabetes, obesity, nonalcoholic fatty liver disease (NAFLD), and nonalcoholic steatohepatitis (NASH). However, the short half-life and poor stability of wild-type FGF21 limit its clinical application. Previous studies found that PEGylation can significantly increase the stability of FGF21. However, the uneven distribution of PEGylation sites in FGF21 makes it difficult to purify PEG-FGF21, thereby affecting its yield, purity, and activity. To obtain long-acting FGF21 with controlled site-specific modification, we mutated lysine residues in FGF21, resulting in PEGylation only at the N-terminus of FGF21 (mFGF21). In addition, we modified mFGF21 molecules with different PEG molecules and selected the PEG-mFGF21 moiety with the highest activity. The yield of PEG-mFGF21 in this study reached 1 g/L (purity >99 %), and the purification process was simple and efficient with strong quality controllability. The half-life of PEG-mFGF21 in rats reached 40.5-67.4 h. Pharmacodynamic evaluation in mice with high-fat, high-cholesterol- and methionine and choline deficiency-induced NASH illustrated that PEG-mFGF21 exhibited long-term efficacy in improving liver steatosis and reducing liver cell damage, inflammation, and fibrosis. Taken together, PEG-mFGF21 could represent a potential therapeutic drug for the treatment of NASH.
Collapse
Affiliation(s)
- Jianying Qi
- National Research Centre for Carbohydrate Synthesis, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Zhimou Guo
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, Dalian 116023, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Xuan Jiang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Yuanyuan Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Yingli Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Fei Hu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Jingjing Xiong
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - YunZhou Wu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China.
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China.
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Zhongshan Road 457, Dalian 116023, China
| |
Collapse
|
4
|
Liu Y, Xia L, Li H, Cai P, Tang S, Feng Y, Liu G, Chen Y, Liu N, Zhang W, Zhou Z. Exploring the impact of PEGylation on pharmacokinetics: a size-dependent effect of polyethylene glycol on prostate-specific membrane antigen inhibitors. EJNMMI Res 2024; 14:15. [PMID: 38324095 PMCID: PMC10850047 DOI: 10.1186/s13550-024-01071-z] [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: 10/25/2023] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Prostate cancer is the second most frequent cancer and the fifth leading cause of cancer-related deaths in men. Prostate-specific membrane antigen (PSMA) as a target has gained increasing attention. This research aims to investigate and understand how altering size of PEG impacts the in vitro and in vivo behavior and performance of PSMA inhibitors, with a specific focus on their pharmacokinetic characteristics and targeting properties. RESULTS Two 68Ga-labeled PSMA-targeted radiotracers were developed, namely [68Ga]Ga-PP4-WD and [68Ga]Ga-PP8-WD, with varying sizes of polyethylene glycol (PEG). [68Ga]Ga-PP4-WD and [68Ga]Ga-PP8-WD had excellent affinity for PSMA with IC50 being 8.06 ± 0.91, 6.13 ± 0.79 nM, respectively. Both tracers enabled clear visualization of LNCaP tumors in PET images with excellent tumor-to-background contrast. They also revealed highly efficient uptake and internalization into LNCaP cells, increasing over time. The biodistribution studies demonstrated that both radioligands exhibited significant and specific uptake into LNCaP tumors. Furthermore, they were rapidly cleared through the renal pathway, as evidenced by [68Ga]Ga-PP4-WD and [68Ga]Ga-PP8-WD showing a tenfold and a fivefold less in renal uptake, respectively, compared to [68Ga]Ga-Flu-1 in 30 min. Both in vitro and in vivo experiments demonstrated that PEG size significantly impacted tumor-targeting and pharmacokinetic properties. CONCLUSIONS These radiotracers have demonstrated their effectiveness in significantly reducing kidney uptake while maintaining the absorbed dose in tumors. Both radiotracers exhibited strong binding and internalization characteristics in vitro, displayed high specificity and affinity for PSMA in vivo.
Collapse
Affiliation(s)
- Yang Liu
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Li Xia
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Haiyang Li
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Ping Cai
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Sufan Tang
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Yue Feng
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Guangfu Liu
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
| | - Nan Liu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Sichuan, Chengdu, China.
| | - Wei Zhang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Sichuan, Chengdu, China.
| | - Zhijun Zhou
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
| |
Collapse
|
5
|
Li C, Jin K. Chemical Strategies towards the Development of Effective Anticancer Peptides. Curr Med Chem 2024; 31:1839-1873. [PMID: 37170992 DOI: 10.2174/0929867330666230426111157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/28/2023] [Accepted: 02/24/2023] [Indexed: 05/13/2023]
Abstract
Cancer is increasingly recognized as one of the primary causes of death and has become a multifaceted global health issue. Modern medical science has made significant advancements in the diagnosis and therapy of cancer over the past decade. The detrimental side effects, lack of efficacy, and multidrug resistance of conventional cancer therapies have created an urgent need for novel anticancer therapeutics or treatments with low cytotoxicity and drug resistance. The pharmaceutical groups have recognized the crucial role that peptide therapeutic agents can play in addressing unsatisfied healthcare demands and how these become great supplements or even preferable alternatives to biological therapies and small molecules. Anticancer peptides, as a vibrant therapeutic strategy against various cancer cells, have demonstrated incredible anticancer potential due to high specificity and selectivity, low toxicity, and the ability to target the surface of traditional "undruggable" proteins. This review will provide the research progression of anticancer peptides, mainly focusing on the discovery and modifications along with the optimization and application of these peptides in clinical practice.
Collapse
Affiliation(s)
- Cuicui Li
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Kang Jin
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
6
|
Jiang W, Gao X, Wang Q, Chen Y, Li D, Zhang X, Yang X. The Modified Exenatide Microspheres: PLGA-PEG-PLGA Gel and Zinc-Exenatide Complex Synergistically Reduce Burst Release and Shorten Platform Stage. AAPS PharmSciTech 2023; 24:251. [PMID: 38036924 DOI: 10.1208/s12249-023-02705-6] [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: 09/03/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023] Open
Abstract
The existing exenatide microspheres have the problem of burst release in the early stage, and minimal release in the middle stage which makes it difficult to achieve effective blood drug concentration (platform period). In this study, the modified exenatide microspheres were constructed to address the aforementioned issues. Poly(D,L-lactic-co-glycolic acid) (PLGA) and triblock copolymer with sol-gel conversion characteristics (PLGA-PEG-PLGA gel) were introduced as carriers to prepare microspheres. The hot gel characteristics and hydrophilicity of PLGA-PEG-PLGA gel were utilized to decline the burst release and shorten the platform period. Simultaneously, zinc acetate and exenatide were combined to generate an insoluble complex to further reduce the burst release. Herein, we prepared three types of exenatide microspheres using the solvent evaporation method and investigated their characterization as well as in vitro and in vivo release. According to the experimental findings, the modified exenatide microspheres, i.e., PLGA-PEG-PLGA gel and PLGA co-loaded zinc-exenatide insoluble complex microspheres (Zn-EXT-Gel-MS), had smooth and rounded surfaces, with a particle size of 24.7 μm, and the encapsulation rate reached 89.43%. And it was released for 40 days in vitro, behaving better than the other two microspheres in terms of release behavior. When this product was administered subcutaneously to rats, it produced a comparatively constant plasma exenatide concentration that lasted for 24 days and superior bioavailability than the exenatide microspheres (EXT-MS). The creation of modified exenatide microspheres may serve as a heuristic method for other long-acting medications. Schematic diagram of the synthesis process and release curves of three types of exenatide microspheres in vitro and in vivo.
Collapse
Affiliation(s)
- Wenjing Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Xiangjun Gao
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Qiuli Wang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Yang Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Dan Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Xiaoyan Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China
| | - Xinggang Yang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
7
|
Fallarini S, Cerofolini L, Salobehaj M, Rizzo D, Gheorghita GR, Licciardi G, Capialbi DE, Zullo V, Sodini A, Nativi C, Fragai M. Site-Selective Functionalized PD-1 Mutant for a Modular Immunological Activity against Cancer Cells. Biomacromolecules 2023; 24:5428-5437. [PMID: 37902625 PMCID: PMC10646970 DOI: 10.1021/acs.biomac.3c00893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/31/2023]
Abstract
Targeting immune checkpoints is a well-established strategy in cancer therapy, and antibodies blocking PD-1/PD-L1 interactions to restore the immunological activity against cancer cells have been clinically validated. High-affinity mutants of the PD-1 ectodomain have recently been proposed as an alternative to antibodies to target PD-L1 on cancer cells, shedding new light on this research area. In this dynamic scenario, the PD-1 mutant, here reported, largely expands the chemical space of nonantibody and nonsmall-molecule inhibitor therapeutics that can be used to target cancer cells overexpressing PD-L1 receptors. The polyethylene glycol moieties and the immune response-stimulating carbohydrates, used as site-selective tags, represent the proof of concept for future applications.
Collapse
Affiliation(s)
- Silvia Fallarini
- Department
of Pharmaceutical Sciences, DSF, University
of Piemonte Orientale, Largo Donegani 2, Novara (NO) 28100, Italy
| | - Linda Cerofolini
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto
Fiorentino (FI) 50019, Italy
| | - Maria Salobehaj
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto
Fiorentino (FI) 50019, Italy
| | - Domenico Rizzo
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto
Fiorentino (FI) 50019, Italy
| | - Giulia Roxana Gheorghita
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto
Fiorentino (FI) 50019, Italy
- Giotto
Biotech, S.R.L, Via Madonna
del Piano 6, Sesto Fiorentino (FI) 50019, Italy
| | - Giulia Licciardi
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto
Fiorentino (FI) 50019, Italy
| | - Daniela Eloisa Capialbi
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
| | - Valerio Zullo
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
| | - Andrea Sodini
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
| | - Cristina Nativi
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
| | - Marco Fragai
- Department
of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI) 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto
Fiorentino (FI) 50019, Italy
| |
Collapse
|
8
|
Nguyen TTK, Pham KY, Yook S. Engineered therapeutic proteins for sustained-release drug delivery systems. Acta Biomater 2023; 171:131-154. [PMID: 37717712 DOI: 10.1016/j.actbio.2023.09.018] [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: 03/01/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
Proteins play a vital role in diverse biological processes in the human body, and protein therapeutics have been applied to treat different diseases such as cancers, genetic disorders, autoimmunity, and inflammation. Protein therapeutics have demonstrated their advantages, such as specific pharmaceutical effects, low toxicity, and strong solubility. However, several disadvantages arise in clinical applications, including short half-life, immunogenicity, and low permeation, leading to reduced drug effectiveness. The structure of protein therapeutics can be modified to increase molecular size, leading to prolonged stability and increased plasma half-life. Notably, the controlled-release delivery systems for the sustained release of protein drugs and preserving the stability of cargo proteins are envisioned as a potential approach to overcome these challenges. In this review, we summarize recent research progress related to structural modifications (PEGylation, glycosylation, poly amino acid modification, and molecular biology-based strategies) and promising long-term delivery systems, such as polymer-based systems (injectable gel/implants, microparticles, nanoparticles, micro/nanogels, functional polymers), lipid-based systems (liposomes, solid lipid nanoparticles, nanostructured lipid carriers), and inorganic nanoparticles exploited for protein therapeutics. STATEMENT OF SIGNIFICANCE: In this review, we highlight recent advances concerning modifying proteins directly to enhance their stability and functionality and discuss state-of-the-art methods for the delivery and controlled long-term release of active protein therapeutics to their target site. In terms of drug modifications, four widely used strategies, including PEGylation, poly amino acid modification, glycosylation, and genetic, are discussed. As for drug delivery systems, we emphasize recent progress relating to polymer-based systems, lipid-based systems developed, and inorganic nanoparticles for protein sustained-release delivery. This review points out the areas requiring focused research attention before the full potential of protein therapeutics for human health and disease can be realized.
Collapse
Affiliation(s)
- Thoa Thi Kim Nguyen
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | - Khang-Yen Pham
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
9
|
Alamdari-Palangi V, Jaberi KR, Shahverdi M, Naeimzadeh Y, Tajbakhsh A, Khajeh S, Razban V, Fallahi J. Recent advances and applications of peptide-agent conjugates for targeting tumor cells. J Cancer Res Clin Oncol 2023; 149:15249-15273. [PMID: 37581648 DOI: 10.1007/s00432-023-05144-9] [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: 05/23/2023] [Accepted: 07/08/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Cancer, being a complex disease, presents a major challenge for the scientific and medical communities. Peptide therapeutics have played a significant role in different medical practices, including cancer treatment. METHOD This review provides an overview of the current situation and potential development prospects of anticancer peptides (ACPs), with a particular focus on peptide vaccines and peptide-drug conjugates for cancer treatment. RESULTS ACPs can be used directly as cytotoxic agents (molecularly targeted peptides) or can act as carriers (guiding missile) of chemotherapeutic agents and radionuclides by specifically targeting cancer cells. More than 60 natural and synthetic cationic peptides are approved in the USA and other major markets for the treatment of cancer and other diseases. Compared to traditional cancer treatments, peptides exhibit anticancer activity with high specificity and the ability to rapidly kill target cancer cells. ACP's target and kill cancer cells via different mechanisms, including membrane disruption, pore formation, induction of apoptosis, necrosis, autophagy, and regulation of the immune system. Modified peptides have been developed as carriers for drugs, vaccines, and peptide-drug conjugates, which have been evaluated in various phases of clinical trials for the treatment of different types of solid and leukemia cancer. CONCLUSIONS This review highlights the potential of ACPs as a promising therapeutic option for cancer treatment, particularly through the use of peptide vaccines and peptide-drug conjugates. Despite the limitations of peptides, such as poor metabolic stability and low bioavailability, modified peptides show promise in addressing these challenges. Various mechanism of action of anticancer peptides. Modes of action against cancer cells including: inducing apoptosis by cytochrome c release, direct cell membrane lysis (necrosis), inhibiting angiogenesis, inducing autophagy-mediated cell death and immune cell regulation.
Collapse
Affiliation(s)
- Vahab Alamdari-Palangi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Khojaste Rahimi Jaberi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahshid Shahverdi
- Medical Biotechnology Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| |
Collapse
|
10
|
Wang Y, Yuan H, Liu N, Tang S, Feng Y, Liu Y, Cai P, Xia L, Zheng W, Chen Y, Zhou Z. High Affinity and FAP-Targeted Radiotracers: A Potential Design Strategy to Improve the Pharmacokinetics and Tumor Uptake for FAP Inhibitors. J Med Chem 2023. [PMID: 37390480 DOI: 10.1021/acs.jmedchem.3c00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
Fibroblast activation protein (FAP) is overexpressed in cancer-associated fibroblasts, making it an attractive target for both imaging and therapy of malignancy. This study presents a range of novel FAP inhibitors derived from amino derivatives of UAMC1110, incorporating polyethylene glycol and bulky groups containing bifunctional DOTA chelators. The compounds labeled with gallium-68 were developed and characterized to study biodistribution properties and tumor-targeting performance in nude mice bearing U87MG tumor xenografts. Several tracers of interest were screened due to the advantages in imaging and tumor-specific uptake. Positron emission tomography scans revealed that polyethylene glycol-modified 68Ga-3-3 had a rapid penetration within the neoplastic tissue and excellent tumor-to-background contrast. In a comparative biodistribution study, naphthalene-modified 68Ga-6-3 exhibited more significant tumor uptake (∼50% ID/g, 1 h p.i.) than 68Ga-3-3 and 10-fold higher than 68Ga-FAPI-04 under the same conditions. Remarkably, 68Ga-8-1, combining the two structural design strategies, obtains superior imaging performance.
Collapse
Affiliation(s)
- Yinwen Wang
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hongmei Yuan
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Nan Liu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Sufan Tang
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yue Feng
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yang Liu
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ping Cai
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Li Xia
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Wenlu Zheng
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yue Chen
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhijun Zhou
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
11
|
Sauer DF, Markel U, Schiffels J, Okuda J, Schwaneberg U. FhuA: From Iron-Transporting Transmembrane Protein to Versatile Scaffolds through Protein Engineering. Acc Chem Res 2023. [PMID: 37191525 DOI: 10.1021/acs.accounts.3c00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
ConspectusProtein engineering has emerged as a powerful methodology to tailor the properties of proteins. It empowers the design of biohybrid catalysts and materials, thereby enabling the convergence of materials science, chemistry, and medicine. The choice of a protein scaffold is an important factor for performance and potential applications. In the past two decades, we utilized the ferric hydroxamate uptake protein FhuA. FhuA is, from our point of view, a versatile scaffold due to its comparably large cavity and robustness toward temperature as well as organic cosolvents. FhuA is a natural iron transporter located in the outer membrane of Escherichia coli (E. coli). Wild-type FhuA consists of 714 amino acids and has a β-barrel structure composed of 22 antiparallel β-sheets, closed by an internal globular "cork" domain (amino acids 1-160). FhuA is robust in a broad pH range and toward organic cosolvents; therefore, we envisioned FhuA to be a suitable platform for various applications in (i) biocatalysis, (ii) materials science, and (iii) the construction of artificial metalloenzymes.(i) Applications in biocatalysis were achieved by removing the globular cork domain (FhuA_Δ1-160), thereby creating a large pore for the passive transport of otherwise difficult-to-import molecules through diffusion. Introducing this FhuA variant into the outer membrane of E. coli facilitates the uptake of substrates for downstream biocatalytic conversion. Furthermore, removing the globular "cork" domain without structural collapse of the ß-barrel protein allowed the use of FhuA as a membrane filter, exhibiting a preference for d-arginine over l-arginine.(ii) FhuA is a transmembrane protein, which makes it attractive to be used for applications in non-natural polymeric membranes. Inserting FhuA into polymer vesicles yielded so-called synthosomes (i.e., catalytic synthetic vesicles in which the transmembrane protein acted as a switchable gate or filter). Our work in this direction enables polymersomes to be used in biocatalysis, DNA recovery, and the controlled (triggered) release of molecules. Furthermore, FhuA can be used as a building block to create protein-polymer conjugates to generate membranes.(iii) Artificial metalloenzymes (ArMs) are formed by incorporating a non-native metal ion or metal complex into a protein. This combines the best of two worlds: the vast reaction and substrate scope of chemocatalysis and the selectivity and evolvability of enzymes. With its large inner diameter, FhuA can harbor (bulky) metal catalysts. Among others, we covalently attached a Grubbs-Hoveyda-type catalyst for olefin metathesis to FhuA. This artificial metathease was then used in various chemical transformations, ranging from polymerizations (ring-opening metathesis polymerization) to enzymatic cascades involving cross-metathesis. Ultimately, we generated a catalytically active membrane by copolymerizing FhuA and pyrrole. The resulting biohybrid material was then equipped with the Grubbs-Hoveyda-type catalyst and used in ring-closing metathesis.The number of reports on FhuA and its various applications indicates that it is a versatile building block to generate hybrid catalysts and materials. We hope that our research will inspire future research efforts at the interface of biotechnology, catalysis, and material science in order to create biohybrid systems that offer smart solutions for current challenges in catalysis, material science, and medicine.
Collapse
Affiliation(s)
- Daniel F Sauer
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Ulrich Markel
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Johannes Schiffels
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
| | - Jun Okuda
- Institute of Inorganic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
| | - Ulrich Schwaneberg
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, D-52056, Aachen, Germany
| |
Collapse
|
12
|
Bootwala A, An HH, Franklin MW, Manning BJ, Xu LY, Panchal S, Garlick JD, Baral R, Hudson ME, Grigoryan G, Murakami MA, Hopson K, Leventhal DS. Protein re-surfacing of E. coli L-Asparaginase to evade pre-existing anti-drug antibodies and hypersensitivity responses. Front Immunol 2022; 13:1016179. [PMID: 36569945 PMCID: PMC9767956 DOI: 10.3389/fimmu.2022.1016179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
The optimal use of many biotherapeutics is restricted by Anti-drug antibodies (ADAs) and hypersensitivity responses which can affect potency and ability to administer a treatment. Here we demonstrate that Re-surfacing can be utilized as a generalizable approach to engineer proteins with extensive surface residue modifications in order to avoid binding by pre-existing ADAs. This technique was applied to E. coli Asparaginase (ASN) to produce functional mutants with up to 58 substitutions resulting in direct modification of 35% of surface residues. Re-surfaced ASNs exhibited significantly reduced binding to murine, rabbit and human polyclonal ADAs, with a negative correlation observed between binding and mutational distance from the native protein. Reductions in ADA binding correlated with diminished hypersensitivity responses in an in vivo mouse model. By using computational design approaches to traverse extended distances in mutational space while maintaining function, protein Re-surfacing may provide a means to generate novel or second line therapies for life-saving drugs with limited therapeutic alternatives.
Collapse
Affiliation(s)
- Ali Bootwala
- Generate Biomedicines, Somerville, MA, United States
| | - Hyun Hwan An
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | | | | | - Lucy Y. Xu
- Generate Biomedicines, Somerville, MA, United States
| | | | | | - Reshica Baral
- Generate Biomedicines, Somerville, MA, United States
| | | | | | - Mark A. Murakami
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | | | | |
Collapse
|
13
|
Anticancer peptides mechanisms, simple and complex. Chem Biol Interact 2022; 368:110194. [PMID: 36195187 DOI: 10.1016/j.cbi.2022.110194] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/12/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022]
Abstract
Peptide therapy has started since 1920s with the advent of insulin application, and now it has emerged as a new approach in treatment of diseases including cancer. Using anti-cancer peptides (ACPs) is a promising way of cancer therapy as ACPs are continuing to be approved and arrived at major pharmaceutical markets. Traditional cancer treatments face different problems like intensive adverse effects to patient's body, cell resistance to conventional chemical drugs and in some worse cases the occurrence of cell multidrug resistance (MDR) of cancerous tissues against chemotherapy. On the other hand, there are some benefits conceived for peptides usage in treatment of diseases specifically cancer, as these compounds present favorable characteristics such as smaller size, high activity, low immunogenicity, good biocompatibility in vivo, convenient and rapid way of synthesis, amenable to sequence modification and revision and there is no limitation for the type of cargo they carry. It is possible to achieve an optimum molecular and functional structure of peptides based on previous experience and bank of peptide motif data which may result in novel peptide design. Bioactive peptides are able to form pores in cell membrane and induce necrosis or apoptosis of abnormal cells. Moreover, recent researches have focused on the tumor recognizing peptide motifs with the ability to permeate to cancerous cells with the aim of cancer treatment at earlier stages. In this strategy the most important factors for addressing cancer are choosing peptides with easy accessibility to tumor cell without cytotoxicity effect towards normal cells. The peptides must also meet acceptable pharmacokinetic requirements. In this review, the characteristics of peptides and cancer cells are discussed. The various mechanisms of peptides' action proposed against cancer cells make the next part of discussion. It will be followed by giving information on peptides application, various methods of peptide designing along with introducing various databases. Future aspects of peptides for employing in area of cancer treatment come as conclusion at the end.
Collapse
|
14
|
IL-2K35C-moFA, a Long-Acting Engineered Cytokine with Decreased Interleukin 2 Receptor α Binding, Improved the Cellular Selectivity Profile and Antitumor Efficacy in a Mouse Tumor Model. Cancers (Basel) 2022; 14:cancers14194742. [PMID: 36230665 PMCID: PMC9563011 DOI: 10.3390/cancers14194742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary The application of IL-2 for treating cancer is limited owing to its toxicity and short half-life. Its high binding ability to IL-2 receptor α expands immunosuppressive Treg cells, which represents an undesirable toxicity in cancer immunotherapy. Moreover, its small molecular size is the reason for its short half-life. We solved these problems by using a covalent modification strategy of IL-2 variant IL-2K35C with fatty acid by maleimide chemistry, namely, IL-2K35C-moFA. The experiments performed in vitro and in vivo proved that IL-2K35C-moFA is a novel immunotherapeutic agent with the potential to selectively stimulate CD8+ T cells and NK cells. Compared to IL-2WT, IL-2K35C-moFA showed a specifically reduced potency for the stimulation of Treg cells. Our results also showed that fatty acid conjugation appears to be effective in half-life extension. The combination of selective lymphocyte expansion and its long half-life means IL-2K35C-moFA should be evaluated as a potential human immunotherapeutic in the future. Abstract Human interleukin 2 (IL-2) has shown impressive results as a therapeutic agent for cancer. However, IL-2-based cancer therapy is limited by strong Treg amplification owing to its high binding affinity to IL-2 receptor α (IL-2Rα) and its short half-life owing to its small molecular size. In this study, we solved these problems using a covalent modification strategy of the IL-2 variant, i.e., substituting cysteine (C) for lysine (K) at position 35, using octadecanedicarboxylic acid through maleimide chemistry, creating IL-2K35C-moFA. IL-2K35C-moFA was equipotent to human IL-2 wild type (IL-2WT) in activating tumor-killing CD8+ memory effector T cells (CD8+ T) and NK cells bearing the intermediate affinity IL-2 receptors, and less potent than IL-2WT on CTLL-2 cells bearing the high-affinity IL-2 receptors. Moreover, it was shown to support the preferential activation of IL-2 receptor β (IL-2Rβ) over IL-2Rα because of the mutation and fatty acid conjugation. In a B16F10 murine tumor model, IL-2K35C-moFA showed efficacy as a single dose and provided durable immunity for 1 week. Our results support the further evaluation of IL-2K35C-moFA as a novel cancer immunotherapy.
Collapse
|
15
|
PEGylated and zwitterated silica nanoparticles as doxorubicin carriers applied in a breast cancer cell line: Effects on protein corona formation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
16
|
Santos JHPM, Feitosa VA, Meneguetti GP, Carretero G, Coutinho JAP, Ventura SPM, Rangel-Yagui CO. Lysine-PEGylated Cytochrome C with Enhanced Shelf-Life Stability. BIOSENSORS 2022; 12:94. [PMID: 35200354 PMCID: PMC8869816 DOI: 10.3390/bios12020094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Cytochrome c (Cyt-c), a small mitochondrial electron transport heme protein, has been employed in bioelectrochemical and therapeutic applications. However, its potential as both a biosensor and anticancer drug is significantly impaired due to poor long-term and thermal stability. To overcome these drawbacks, we developed a site-specific PEGylation protocol for Cyt-c. The PEG derivative used was a 5 kDa mPEG-NHS, and a site-directed PEGylation at the lysine amino-acids was performed. The effects of the pH of the reaction media, molar ratio (Cyt-c:mPEG-NHS) and reaction time were evaluated. The best conditions were defined as pH 7, 1:25 Cyt-c:mPEG-NHS and 15 min reaction time, resulting in PEGylation yield of 45% for Cyt-c-PEG-4 and 34% for Cyt-c-PEG-8 (PEGylated cytochrome c with 4 and 8 PEG molecules, respectively). Circular dichroism spectra demonstrated that PEGylation did not cause significant changes to the secondary and tertiary structures of the Cyt-c. The long-term stability of native and PEGylated Cyt-c forms was also investigated in terms of peroxidative activity. The results demonstrated that both Cyt-c-PEG-4 and Cyt-c-PEG-8 were more stable, presenting higher half-life than unPEGylated protein. In particular, Cyt-c-PEG-8 presented great potential for biomedical applications, since it retained 30-40% more residual activity than Cyt-c over 60-days of storage, at both studied temperatures of 4 °C and 25 °C.
Collapse
Affiliation(s)
- João H. P. M. Santos
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, Brazil; (V.A.F.); (G.P.M.)
- Bionanomanufacturing Center, Institute for Technological Research, São Paulo 05508-901, Brazil
| | - Valker A. Feitosa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, Brazil; (V.A.F.); (G.P.M.)
- Bionanomanufacturing Center, Institute for Technological Research, São Paulo 05508-901, Brazil
| | - Giovanna P. Meneguetti
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, Brazil; (V.A.F.); (G.P.M.)
- Bionanomanufacturing Center, Institute for Technological Research, São Paulo 05508-901, Brazil
| | - Gustavo Carretero
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil;
| | - João A. P. Coutinho
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal; (J.A.P.C.); (S.P.M.V.)
| | - Sónia P. M. Ventura
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal; (J.A.P.C.); (S.P.M.V.)
| | - Carlota O. Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Science, University of São Paulo, São Paulo 05508-000, Brazil; (V.A.F.); (G.P.M.)
| |
Collapse
|
17
|
Sindhu R, Manonmani HK. L-asparaginase mediated therapy in L-asparagine auxotrophic cancers: A review. Anticancer Agents Med Chem 2022; 22:2393-2410. [PMID: 34994334 DOI: 10.2174/1871520622666220106103336] [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: 04/22/2021] [Revised: 09/28/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Microbial L-asparaginase is the most effective first-line therapeutic used in the treatment protocols of paediatric and adult leukemia. Leukemic cell's auxotrophy for L-asparagine is exploited as a therapeutic strategy to mediate cell death through metabolic blockade of L-asparagine using L-asparaginase. Escherichia coli and Erwinia chrysanthemi serve as the major enzyme deriving sources accepted in clinical practise and the enzyme has bestowed improvements in patient outcomes over the last 40 years. However, an array of side effects generated by the native enzymes due to glutamine co-catalysis and short serum stays augmenting frequent dosages, intended a therapeutic switch towards the development of biobetter alternatives for the enzyme including the formulations resulting in sustained local depletion of L-asparagine. In addition, the treatment with L-asparaginase in few cancer types has proven to elicit drug-induced cytoprotective autophagy mechanisms and therefore warrants concern. Although the off-target glutamine hydrolysis has been viewed in contributing the drug-induced secondary responses in cells deficient with asparagine synthetase machinery, the beneficial role of glutaminase-asparaginase in proliferative regulation of asparagine prototrophic cells has been looked forward. The current review provides an overview on the enzyme's clinical applications in leukemia and possible therapeutic implications in other solid tumours, recent advancements in drug formulations, and discusses the aspects of two-sided roles of glutaminase-asparaginases and drug-induced cytoprotective autophagy mechanisms.
Collapse
Affiliation(s)
- Sindhu R
- Department of Microbiology, Faculty of Life Sciences, JSS-AHER, Mysuru-570015, Karnataka, India
| | - H K Manonmani
- Food Protectants and Infestation Control Department, CSIR-Central Food Technological Research Institute, Mysuru-570020, Karnataka, India
| |
Collapse
|
18
|
Ruan Q, Feng J, Jiang Y, Zhang X, Duan X, Wang Q, Yin G, Xiao D, Zhang J. Preparation and Bioevaluation of 99mTc-Labeled FAP Inhibitors as Tumor Radiotracers to Target the Fibroblast Activation Protein. Mol Pharm 2022; 19:160-171. [PMID: 34904839 DOI: 10.1021/acs.molpharmaceut.1c00712] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fibroblast activation protein (FAP) is overexpressed in cancer-associated fibroblasts (CAFs) in a majority of human epithelial cancers. With low expression in normal organs, FAP has become a promising molecular target for tumor theranostics. To develop a lower cost and more widely available alternative to positron emission tomography (PET), two isocyanide-containing FAP inhibitors (CN-C5-FAPI and CN-PEG4-FAPI) were synthesized and radiolabeled with 99mTc to obtain [99mTc][Tc-(CN-C5-FAPI)6]+ and [99mTc][Tc-(CN-PEG4-FAPI)6]+ in high yields (>95%). They showed good stability in saline and mouse serum. The partition coefficient (log P) values of [99mTc][Tc-(CN-C5-FAPI)6]+ and [99mTc][Tc-(CN-PEG4-FAPI)6]+ were -0.86 ± 0.03 and -2.38 ± 0.07, respectively, indicating that they were good hydrophilic complexes. The low nanomolar IC50 values of CN-C5-FAPI and CN-PEG4-FAPI indicated that they had specificity to FAP. In vitro cellular uptake and blocking experiments implied a FAP-targeted uptake mechanism. The nanomolar Kd values from the saturation binding assay indicated that they had significantly high target affinity to FAP. The biodistribution and blocking study in BALB/c nude mice bearing U87MG tumors showed that both exhibited specific tumor uptake. [99mTc][Tc-(CN-PEG4-FAPI)6]+ showed a higher tumor uptake and a higher tumor/nontarget ratio than [99mTc][Tc-(CN-C5-FAPI)6]+. The results of micro-single-photon emission computed tomography (SPECT) imaging studies of [99mTc][Tc-(CN-C5-FAPI)6]+ and [99mTc][Tc-(CN-PEG4-FAPI)6]+ were in accordance with the biodistribution results, suggesting that [99mTc][Tc-(CN-PEG4-FAPI)6]+ is a promising tumor imaging agent for targeting FAP.
Collapse
Affiliation(s)
- Qing Ruan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Xuran Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Di Xiao
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), College of Chemistry, Beijing Normal University, No. 19 Xinjiekou Wai Boulevard, Haidian District, Beijing 100875, P. R. China
| |
Collapse
|
19
|
Pinheiro-Junior EL, Boldrini-França J, Takeda AAS, Costa TR, Peigneur S, Cardoso IA, Oliveira ISD, Sampaio SV, de Mattos Fontes MR, Tytgat J, Arantes EC. Towards toxin PEGylation: The example of rCollinein-1, a snake venom thrombin-like enzyme, as a PEGylated biopharmaceutical prototype. Int J Biol Macromol 2021; 190:564-573. [PMID: 34506860 DOI: 10.1016/j.ijbiomac.2021.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 12/18/2022]
Abstract
PEGylation was firstly described around 50 years ago and has been used for more than 30 years as a strategy to improve the drugability of biopharmaceuticals. However, it remains poorly employed in toxinology, even though it may be a promising strategy to empower these compounds in therapeutics. This work reports the PEGylation of rCollinein-1, a recombinant snake venom serine protease (SVSP), able to degrade fibrinogen and inhibit the hEAG1 potassium channel. We compared the functional, structural, and immunogenic properties of the non-PEGylated (rCollinein-1) and PEGylated (PEG-rCollinein-1) forms. PEG-rCollinein-1 shares similar kinetic parameters with rCollinein-1, maintaining its capability of degrading fibrinogen, but with reduced activity on hEAG1 channel. CD analysis revealed the maintenance of protein conformation after PEGylation, and thermal shift assays demonstrated similar thermostability. Both forms of the enzyme showed to be non-toxic to peripheral blood mononuclear cells (PBMC). In silico epitope prediction indicated three putative immunogenic peptides. However, immune response on mice showed PEG-rCollinein-1 was devoid of immunogenicity. PEGylation directed rCollinein-1 activity towards hemostasis control, broadening its possibilities to be employed as a defibrinogenant agent.
Collapse
Affiliation(s)
- Ernesto Lopes Pinheiro-Junior
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903 Ribeirão Preto, SP, Brazil; Toxicology and Pharmacology, KU Leuven, O&N II Herestraat 49 - PO box 922, 3000 Leuven, Belgium
| | - Johara Boldrini-França
- University of Vila Velha, Av. Comissário José Dantas de Melo, 21, Boa Vista II, 29102-920 Vila Velha, ES, Brazil
| | | | - Tássia Rafaella Costa
- Institute of Biotechnology, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - Steve Peigneur
- Toxicology and Pharmacology, KU Leuven, O&N II Herestraat 49 - PO box 922, 3000 Leuven, Belgium
| | - Iara Aimê Cardoso
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903 Ribeirão Preto, SP, Brazil
| | - Isadora Sousa de Oliveira
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903 Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903 Ribeirão Preto, SP, Brazil
| | | | - Jan Tytgat
- Toxicology and Pharmacology, KU Leuven, O&N II Herestraat 49 - PO box 922, 3000 Leuven, Belgium
| | - Eliane Candiani Arantes
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n°, 14040-903 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
20
|
Subasic CN, Ardana A, Chan LJ, Huang F, Scoble JA, Butcher NJ, Meagher L, Chiefari J, Kaminskas LM, Williams CC. Poly(HPMA-co-NIPAM) copolymer as an alternative to polyethylene glycol-based pharmacokinetic modulation of therapeutic proteins. Int J Pharm 2021; 608:121075. [PMID: 34481889 DOI: 10.1016/j.ijpharm.2021.121075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022]
Abstract
PEGylation is the standard approach for prolonging the plasma exposure of protein therapeutics but has limitations. We explored whether polymers prepared by Reversible Addition-Fragmentation chain-Transfer (RAFT) may provide better alternatives to polyethylene glycol (PEG). Four RAFT polymers were synthesised with varying compositions, molar mass (Mn), and structures, including a homopolymer of N-(2-hydroxypropyl)methacrylamide, (pHPMA) and statistical copolymers of HPMA with poly(ethylene glycol methyl ether acrylate) p(HPMA-co-PEGA); HPMA and N-acryloylmorpholine, p(HPMA-co-NAM); and HPMA and N-isopropylacrylamide, p(HPMA-co-NIPAM). The intravenous pharmacokinetics of the polymers were then evaluated in rats. The in vitro activity and in vivo pharmacokinetics of p(HPMA-co-NIPAM)-conjugated trastuzumab Fab' and full length mAb were then evaluated. p(HPMA-co-NIPAM) prolonged plasma exposure more avidly compared to the other p(HPMA) polymers or PEG, irrespective of molecular weight. When conjugated to trastuzumab-Fab', p(HPMA-co-NIPAM) prolonged plasma exposure of the Fab' similar to PEG-Fab'. The generation of anti-PEG IgM in rats 7 days after intravenous and subcutaneous dosing of p(HPMA-co-NIPAM) conjugated trastuzumab mAb was also examined and was shown to exhibit lower immunogenicity than the PEGylated construct. These data suggest that p(HPMA-co-NIPAM) has potential as a promising copolymer for use as an alternative conjugation strategy to PEG, to prolong the plasma exposure of therapeutic proteins.
Collapse
Affiliation(s)
- Christopher N Subasic
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Aditya Ardana
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Linda J Chan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Fei Huang
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Judith A Scoble
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Laurence Meagher
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia; Department of Materials Science and Engineering, Monash University, 20 Research Way, Clayton, Victoria 3168, Australia
| | - John Chiefari
- CSIRO Manufacturing, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lisa M Kaminskas
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia; Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.
| | | |
Collapse
|
21
|
Torres-Obreque KM, Meneguetti GP, Muso-Cachumba JJ, Feitosa VA, Santos JHPM, Ventura SPM, Rangel-Yagui CO. Building better biobetters: From fundamentals to industrial application. Drug Discov Today 2021; 27:65-81. [PMID: 34461236 DOI: 10.1016/j.drudis.2021.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/28/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022]
Abstract
Biological drugs or biopharmaceuticals off patent open a large market for biosimilars and biobetters, follow-on biologics. Biobetters, in particular, are new drugs designed from existing ones with improved properties such as higher selectivity, stability, half-life and/or lower toxicity/immunogenicity. Glycosylation is one of the most used strategies to improve biological drugs, nonetheless bioconjugation is an additional alternative and refers to the covalent attachment of polymers to biological drugs. Extensive research on novel polymers is underway, nonetheless PEGylation is still the best alternative with the longest clinical track record. Innovative trends based on genetic engineering techniques such as fusion proteins and PASylation are also promising. In this review, all these alternatives wereexplored as well as current market trends, legislation and future perspectives.
Collapse
Affiliation(s)
- Karin M Torres-Obreque
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Giovanna P Meneguetti
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Bionanomanufacturing Center, Institute for Technological Research (IPT), São Paulo, Brazil
| | - Jorge J Muso-Cachumba
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Valker A Feitosa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Bionanomanufacturing Center, Institute for Technological Research (IPT), São Paulo, Brazil
| | - João H P M Santos
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Sónia P M Ventura
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Carlota O Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
22
|
Preparation and Evaluation of Novel Folate Isonitrile 99mTc Complexes as Potential Tumor Imaging Agents to Target Folate Receptors. Molecules 2021; 26:molecules26154552. [PMID: 34361705 PMCID: PMC8348780 DOI: 10.3390/molecules26154552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022] Open
Abstract
In order to seek novel technetium-99m folate receptor-targeting agents, two folate derivatives (CN5FA and CNPFA) were synthesized and radiolabeled to obtain [99mTc]Tc-CN5FA and [99mTc]Tc-CNPFA complexes, which exhibited high radiochemical purity (>95%) without purification, hydrophilicity, and good stability in vitro. The KB cell competitive binding experiments indicated that [99mTc]Tc-CN5FA and [99mTc]Tc-CNPFA had specificity to folate receptor. Biodistribution studies in KB tumor-bearing mice illustrated that [99mTc]Tc-CN5FA and [99mTc]Tc-CNPFA had specific tumor uptake. Compared with [99mTc]Tc-CN5FA, the tumor/muscle ratios of [99mTc]Tc-CNPFA were higher, resulting in a better SPECT/CT imaging background. According to the results, the two 99mTc complexes have potential as tumor imaging agents to target folate receptors.
Collapse
|
23
|
Li X, Gui R, Li J, Huang R, Shang Y, Zhao Q, Liu H, Jiang H, Shang X, Wu X, Nie X. Novel Multifunctional Silver Nanocomposite Serves as a Resistance-Reversal Agent to Synergistically Combat Carbapenem-Resistant Acinetobacter baumannii. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30434-30457. [PMID: 34161080 DOI: 10.1021/acsami.1c10309] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In the face of the abundant production of various types of carbapenemases, the antibacterial efficiency of imipenem, seen as "the last line of defense", is weakening. Following, the incidence of carbapenem-resistant Acinetobacter baumannii (CRAB), which can generate antibiotic-resistant biofilms, is increasing. Based on the superior antimicrobial activity of silver nanoparticles against multifarious bacterial strains compared with common antibiotics, we constructed the IPM@AgNPs-PEG-NOTA nanocomposite (silver nanoparticles were coated with SH-PEG-NOTA as well as loaded by imipenem) whose core was a silver nanoparticle to address the current challenge, and IPM@AgNPs-PEG-NOTA was able to function as a novel smart pH-sensitive nanodrug system. Synergistic bactericidal effects of silver nanoparticles and imipenem as well as drug-resistance reversal via protection of the β-ring of carbapenem due to AgNPs-PEG-NOTA were observed; thus, this nanocomposite confers multiple advantages for efficient antibacterial activity. Additionally, IPM@AgNPs-PEG-NOTA not only offers immune regulation and accelerates tissue repair to improve therapeutic efficacy in vivo but also can prevent the interaction of pathogens and hosts. Compared with free imipenem or silver nanoparticles, this platform significantly enhanced antibacterial efficiency while increasing reactive oxygen species (ROS) production and membrane damage, as well as affecting cell wall formation and metabolic pathways. According to the results of crystal violet staining, LIVE/DEAD backlight bacterial viability staining, and real-time quantitative polymerase chain reaction (RT-qPCR), this silver nanocomposite downregulated the levels of ompA expression to prevent formation of biofilms. In summary, this research demonstrated that the IPM@AgNPs-PEG-NOTA nanocomposite is a promising antibacterial agent of security, pH sensitivity, and high efficiency in reversing resistance and synergistically combatting carbapenem-resistant A. baumannii. In the future, various embellishments and selected loads for silver nanoparticles will be the focus of research in the domains of medicine and nanotechnology.
Collapse
Affiliation(s)
- Xisheng Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Rong Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Yinghui Shang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Haiting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Haiye Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Xueling Shang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Xin Wu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
| | - Xinmin Nie
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha 410013, P. R. China
- Hunan Engineering Technology Research Center of Optoelectronic Health Detection, Changsha 410000, Hunan, China
| |
Collapse
|
24
|
Guo Q, Yaron JR, Wallen JW, Browder KF, Boyd R, Olson TL, Burgin M, Ulrich P, Aliskevich E, Schutz LN, Fromme P, Zhang L, Lucas AR. PEGylated Serp-1 Markedly Reduces Pristane-Induced Experimental Diffuse Alveolar Hemorrhage, Altering uPAR Distribution, and Macrophage Invasion. Front Cardiovasc Med 2021; 8:633212. [PMID: 33665212 PMCID: PMC7921738 DOI: 10.3389/fcvm.2021.633212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022] Open
Abstract
Diffuse alveolar hemorrhage (DAH) is one of the most serious clinical complications of systemic lupus erythematosus (SLE). The prevalence of DAH is reported to range from 1 to 5%, but while DAH is considered a rare complication there is a reported 50-80% mortality. There is at present no proven effective treatment for DAH and the therapeutics that have been tested have significant side effects. There is a clear necessity to discover new drugs to improve outcomes in DAH. Serine protease inhibitors, serpins, regulate thrombotic and thrombolytic protease cascades. We are investigating a Myxomavirus derived immune modulating serpin, Serp-1, as a new class of immune modulating therapeutics for vasculopathy and lung hemorrhage. Serp-1 has proven efficacy in models of herpes virus-induced arterial inflammation (vasculitis) and lung hemorrhage and has also proved safe in a clinical trial in patients with unstable coronary syndromes and stent implant. Here, we examine Serp-1, both as a native secreted protein expressed by CHO cells and as a polyethylene glycol modified (PEGylated) variant (Serp-1m5), for potential therapy in DAH. DAH was induced by intraperitoneal (IP) injection of pristane in C57BL/6J (B6) mice. Mice were treated with 100 ng/g bodyweight of either Serp-1 as native 55 kDa secreted glycoprotein, or as Serp-1m5, or saline controls after inducing DAH. Treatments were repeated daily for 14 days (6 mice/group). Serp-1 partially and Serp-1m5 significantly reduced pristane-induced DAH when compared with saline as assessed by gross pathology and H&E staining (Serp-1, p = 0.2172; Serp-1m5, p = 0.0252). Both Serp-1m5 and Serp-1 treatment reduced perivascular inflammation and reduced M1 macrophage (Serp-1, p = 0.0350; Serp-1m5, p = 0.0053), hemosiderin-laden macrophage (Serp-1, p = 0.0370; Serp-1m5, p = 0.0424) invasion, and complement C5b/9 staining. Extracellular urokinase-type plasminogen activator receptor positive (uPAR+) clusters were significantly reduced (Serp-1, p = 0.0172; Serp-1m5, p = 0.0025). Serp-1m5 also increased intact uPAR+ alveoli in the lung (p = 0.0091). In conclusion, Serp-1m5 significantly reduces lung damage and hemorrhage in a pristane model of SLE DAH, providing a new potential therapeutic approach.
Collapse
Affiliation(s)
- Qiuyun Guo
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States.,Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jordan R Yaron
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - John W Wallen
- Exalt Therapeutics LLC, Las Vegas, NV, United States
| | - Kyle F Browder
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Ryan Boyd
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Tien L Olson
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Michelle Burgin
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Peaches Ulrich
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Emily Aliskevich
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Lauren N Schutz
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Petra Fromme
- Center for Applied Structural Discovery, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Liqiang Zhang
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Alexandra R Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
25
|
Pegylated catalase as a potential alternative to treat vitiligo and UV induced skin damage. Bioorg Med Chem 2021; 30:115933. [PMID: 33333446 DOI: 10.1016/j.bmc.2020.115933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 11/22/2022]
Abstract
The metabolic function of catalase (CAT) is to prevent oxidative damage to tissues through the hydrolysis of hydrogen peroxide, which is a strong oxidizing agent. It has been suggested as an alternative to treat skin diseases related to oxidative stress, such as vitiligo. Owing to the instability associated to the protein nature, topical use of CAT is challenging and, in this sense, PEGylation can be an interesting alternative. Here, we conjugated CAT to methoxy-poly(ethylene oxide) (mPEG) of 10, 20 and 40 kDa, by means of a nucleophilic attack of ε-amino groups to an electron-deficient carbonyl group of the reactive PEG, resulting in site specifically PEGylated bioconjugates. PEGylation yields ranged from 31% ± 2% for CAT-PEG40 to 59% ± 4% for CAT-PEG20 and were strongly affected by the reaction pH owing to the protonation/deprotonation state of primary amines of lysine and N-terminal residues. PEGylated conjugates were purified by size-exclusion chromatography (purity > 95%) and characterized by circular dichroism. Irrespectively of MW, PEG did not affected CAT secondary and tertiary structure, but a decrease in specific activity was observed, more pronounced when PEGs of higher MWs were used. However, this loss of activity is compensated by the increased long-term stability, with a gain of >5 times in t1/2. In vitro antioxidant activity of CAT-PEG20 showed complete elimination of lipid peroxidation at the skin upper layer (stratum corneum) suitable for a topical use to treat vitiligo, as well as other skin conditions related to oxidative stress.
Collapse
|
26
|
Swierczynski MJ, Ball ZT. One-Step Protein-Polymer Conjugates from Boronic-Acid-Functionalized Polymers. Bioconjug Chem 2020; 31:2494-2498. [PMID: 33078937 DOI: 10.1021/acs.bioconjchem.0c00516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polymer-protein conjugates are hybrid materials with interesting and useful properties. Methods to prepare diverse diblock materials of this sort often struggle to deal with the complexity and size of reagents, and so polymer-protein conjugation represents a stringent testing ground for nontraditional bioconjugation methods, such as metal-catalyzed arylation. This work demonstrates a simple Ni2+-promoted arylation of cysteine residues with end-functionalized polymer-boronic acid reagents, and explores some molecular and physical properties possible in these hybrid structures.
Collapse
Affiliation(s)
- Michael J Swierczynski
- Department of Chemistry, Rice University, 6100 Main Street, Bioscience Research Collaborative, Houston, Texas 77005, United States
| | - Zachary T Ball
- Department of Chemistry, Rice University, 6100 Main Street, Bioscience Research Collaborative, Houston, Texas 77005, United States
| |
Collapse
|
27
|
Yadav D, Dewangan HK. PEGYLATION: an important approach for novel drug delivery system. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:266-280. [PMID: 32942961 DOI: 10.1080/09205063.2020.1825304] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PEGylation is the covalent addition of PEG to one more molecule. PEGylation can improve the maintenance time of the therapeutics similar to proteins, liposomes, and nanoparticle through shielding them beside different debasing mechanisms dynamic in a body that improve beneficial properties. This skill is used to get better half-life and other pharmaceutical properties of a protein, peptide, or non-peptide molecule. Polyethylene glycol is harmless, non-immunogenic, non-antigenic, and extremely soluble in water and FDA accepted polymer. It shows a significant role in drug delivery. A variety of PEG-based formulations are available in the market. This paper represents the benefits of PEGylation over non-PEGylated products. Now a day, PEGylation plays an important role in the drug delivery system. PEGylation increases the therapeutic potential of drugs.
Collapse
Affiliation(s)
- Deepa Yadav
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | | |
Collapse
|
28
|
Katsura S, Furuishi T, Ueda H, Yonemochi E. Synthesis and Characterization of Cholesteryl Conjugated Lysozyme (CHLysozyme). Molecules 2020; 25:molecules25163704. [PMID: 32823837 PMCID: PMC7465789 DOI: 10.3390/molecules25163704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/19/2022] Open
Abstract
Hydrophobic interaction is important for protein conformation. Conjugation of a hydrophobic group can introduce intermolecular hydrophobic contacts that can be contained within the molecule. It is possible that a strongly folded state can be formed in solution compared with the native state. In this study, we synthesized cholesteryl conjugated lysozyme (CHLysozyme) using lysozyme and cholesterol as the model protein and hydrophobic group, respectively. Cholesteryl conjugation to lysozyme was confirmed by nuclear-magnetic resonance. Differential-scanning calorimetry suggested that CHLysozyme was folded in solution. CHLysozyme secondary structure was similar to lysozyme, although circular dichroism spectra indicated differences to the tertiary structure. Fluorescence measurements revealed a significant increase in the hydrophobic surface of CHLysozyme compared with that of lysozyme; CHLysozyme self-associated by hydrophobic interaction of the conjugated cholesterol but the hydrophobic surface of CHLysozyme decreased with time. The results suggested that hydrophobic interaction changed from intramolecular interaction to an intermolecular interaction. Furthermore, the relative activity of CHLysozyme to lysozyme increased with time. Therefore, CHLysozyme likely forms a folded state with an extended durability of activity. Moreover, lysozyme was denatured in 100% DMSO but the local environment of tryptophan in CHLysozyme was similar to that of a native lysozyme. Thus, this study suggests that protein solution stability and resistance to organic solvents may be improved by conjugation of a hydrophobic group.
Collapse
Affiliation(s)
- Shinji Katsura
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan; (S.K.); (T.F.); (H.U.)
- Formulation research Lab., Taiho Pharmaceutical Co., Ltd., 224-2, Ebisuno, Hiraishi, Kawauchi-cho, Tokushima 771-0194, Japan
| | - Takayuki Furuishi
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan; (S.K.); (T.F.); (H.U.)
| | - Haruhisa Ueda
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan; (S.K.); (T.F.); (H.U.)
| | - Etsuo Yonemochi
- Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan; (S.K.); (T.F.); (H.U.)
- Correspondence: ; Tel.: +81-3-5498-5048
| |
Collapse
|
29
|
Xie M, Liu D, Yang Y. Anti-cancer peptides: classification, mechanism of action, reconstruction and modification. Open Biol 2020; 10:200004. [PMID: 32692959 PMCID: PMC7574553 DOI: 10.1098/rsob.200004] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Anti-cancer peptides (ACPs) are a series of short peptides composed of 10-60 amino acids that can inhibit tumour cell proliferation or migration, or suppress the formation of tumour blood vessels, and are less likely to cause drug resistance. The aforementioned merits make ACPs the most promising anti-cancer candidate. However, ACPs may be degraded by proteases, or result in cytotoxicity in many cases. To overcome these drawbacks, a plethora of research has focused on reconstruction or modification of ACPs to improve their anti-cancer activity, while reducing their cytotoxicity. The modification of ACPs mainly includes main chain reconstruction and side chain modification. After summarizing the classification and mechanism of action of ACPs, this paper focuses on recent development and progress about their reconstruction and modification. The information collected here may provide some ideas for further research on ACPs, in particular their modification.
Collapse
Affiliation(s)
- Mingfeng Xie
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China
| | - Dijia Liu
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China
| | - Yufeng Yang
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China.,Zhuhai Key Laboratory of Fundamental and Applied Research in Traditional Chinese Medicine, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong 519040, People's Republic of China
| |
Collapse
|
30
|
Porfiryeva NN, Moustafine RI, Khutoryanskiy VV. PEGylated Systems in Pharmaceutics. POLYMER SCIENCE SERIES C 2020. [DOI: 10.1134/s181123822001004x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Krzyscik MA, Zakrzewska M, Otlewski J. Site-Specific, Stoichiometric-Controlled, PEGylated Conjugates of Fibroblast Growth Factor 2 (FGF2) with Hydrophilic Auristatin Y for Highly Selective Killing of Cancer Cells Overproducing Fibroblast Growth Factor Receptor 1 (FGFR1). Mol Pharm 2020; 17:2734-2748. [PMID: 32501706 PMCID: PMC7588128 DOI: 10.1021/acs.molpharmaceut.0c00419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
In
spite of significant progress in the field of targeted anticancer
therapy, the FDA has approved only five ADC-based drugs. Hence the
search for new targeted anticancer agents is an unfulfilled necessity.
Here, we present novel types of protein–drug conjugates (PDCs)
that exhibit superior anticancer activities. Instead of a monoclonal
antibody, we used fibroblast growth factor 2 (FGF2) as a targeting
molecule. FGF2 is a natural ligand of fibroblast growth factor receptor
1 (FGFR1), a transmembrane receptor overproduced in various types
of cancers. We synthesized site-specific and stoichiometric-controlled
conjugates of FGF2 with a highly potent, hydrophilic derivative of
auristatin called auristatin Y. To increase the hydrophilicity and
hydrodynamic radius of conjugates, we employed PEG4 and PEG27 molecules
as a spacer between the targeting molecule and the cytotoxic payload.
All conjugates were selective to FGFR1-positive cell lines, effectively
internalized via the FGFR1-dependent pathway, and exhibited a highly
cytotoxic effect only on FGFR1-positive cancer cell lines.
Collapse
|
32
|
Perspectives, Tendencies, and Guidelines in Affinity-Based Strategies for the Recovery and Purification of PEGylated Proteins. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/6163904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years, the effective purification of PEGylated therapeutic proteins from reaction media has received particular attention. Although several techniques have been used, affinity-based strategies have been scarcely explored despite the fact that, after PEGylation, marked changes in the molecular affinity parameters of the modified molecules are observed. With this in mind, future contributions in the bioseparation of these polymer-protein conjugates are expected to exploit affinity in chromatographic and nonchromatographic techniques which will surely derive in the integration of different operations. However, this will only occur as novel ligands which are simultaneously found. As it will be mentioned, these novel ligands may be screened or designed. In both cases, computer-aided tools will support their identification or development. Additionally, ligand discovery by high-throughput screening (HTS) is believed to become a fast, economic, and informative technology that will aid in the mass production of ligands along with genetic engineering and related technologies. Therefore, besides analyzing the state of the art in affinity separation strategies for PEGylated molecules, this review proposes a basic guideline for the selection of adequate ligands to provide information and prospective on the future of affinity operations in solving this particular bioengineering problem.
Collapse
|
33
|
Kozma GT, Shimizu T, Ishida T, Szebeni J. Anti-PEG antibodies: Properties, formation, testing and role in adverse immune reactions to PEGylated nano-biopharmaceuticals. Adv Drug Deliv Rev 2020; 154-155:163-175. [PMID: 32745496 DOI: 10.1016/j.addr.2020.07.024] [Citation(s) in RCA: 357] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
Conjugation of polyethylene glycols (PEGs) to proteins or drug delivery nanosystems is a widely accepted method to increase the therapeutic index of complex nano-biopharmaceuticals. Nevertheless, these drugs and agents are often immunogenic, triggering the rise of anti-drug antibodies (ADAs). Among these ADAs, anti-PEG IgG and IgM were shown to account for efficacy loss due to accelerated blood clearance of the drug (ABC phenomenon) and hypersensitivity reactions (HSRs) entailing severe allergic symptoms with occasionally fatal anaphylaxis. In addition to recapitulating the basic information on PEG and its applications, this review expands on the physicochemical factors influencing its immunogenicity, the prevalence, features, mechanism of formation and detection of anti-PEG IgG and IgM and the mechanisms by which these antibodies (Abs) induce ABC and HSRs. In particular, we highlight the in vitro, animal and human data attesting to anti-PEG Ab-induced complement (C) activation as common underlying cause of both adverse effects. A main message is that correct measurement of anti-PEG Abs and individual proneness for C activation might predict the rise of adverse immune reactions to PEGylated drugs and thereby increase their efficacy and safety.
Collapse
Affiliation(s)
- Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University Medical School, Budapest, Hungary; SeroScience Ltd, Budapest, Hungary
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Janos Szebeni
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University Medical School, Budapest, Hungary; SeroScience Ltd, Budapest, Hungary; Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary.
| |
Collapse
|
34
|
Wu J, Tian X, Yin Y, He M, Cai W, He G, Xu W, Dai Y, Mai J, Gan M, Zhang F. Melphalan-monomethoxypolyethylene glycol-based pH/enzyme double-response polymer prodrug nanoparticles for enhanced drug stability and anticancer efficacy. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2019.109350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
35
|
Geraldes DC, Beraldo-de-Araújo VL, Pardo BOP, Pessoa Junior A, Stephano MA, de Oliveira-Nascimento L. Protein drug delivery: current dosage form profile and formulation strategies. J Drug Target 2019; 28:339-355. [DOI: 10.1080/1061186x.2019.1669043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Danilo Costa Geraldes
- Faculty of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
- Biochemistry and Tissue Biology Department, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| | - Viviane Lucia Beraldo-de-Araújo
- Faculty of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
- Biochemistry and Tissue Biology Department, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| | | | | | | | - Laura de Oliveira-Nascimento
- Faculty of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
- Biochemistry and Tissue Biology Department, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
36
|
Acheampong DO. Bispecific Antibody (bsAb) Construct Formats and their Application in Cancer Therapy. Protein Pept Lett 2019; 26:479-493. [DOI: 10.2174/0929866526666190311163820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/15/2022]
Abstract
Development of cancers mostly involves more than one signal pathways, because of the complicated nature of cancer cells. As such, the most effective treatment option is the one that stops the cancer cells in their tracks by targeting these signal pathways simultaneously. This explains why therapeutic monoclonal antibodies targeted at cancers exert utmost activity when two or more are used as combination therapy. This notwithstanding, studies elsewhere have proven that when bispecific antibody (bsAb) is engineered from two conventional monoclonal antibodies or their chains, it produces better activity than when used as combination therapy. This therefore presents bispecific antibody (bsAb) as the appropriate and best therapeutic agent for the treatment of such cancers. This review therefore discusses the various engineering formats for bispecific antibodies (bsAbs) and their applications.
Collapse
Affiliation(s)
- Desmond O. Acheampong
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Science, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
37
|
Eskian M, Khorasanizadeh M, Zinzani PL, Illidge TM, Rezaei N. Novel Methods to Improve the Efficiency of Radioimmunotherapy for Non-Hodgkin Lymphoma. Int Rev Immunol 2019; 38:79-91. [PMID: 30931651 DOI: 10.1080/08830185.2019.1588266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Radioimmunotherapy (RIT) is a novel strategy for treating non-Hodgkin lymphoma (NHL). Several studies have shown the promising results of using RIT in NHL, which have led to FDA approval for two RIT agents in treating low grade NHL. In spite of these favorable results in low-grade NHL, most of the aggressive or relapsed/refractory NHL subjects experience relapses following RIT. Although more aggressive treatments such as myeloablative doses of RIT followed by stem cell transplantation appear to be able to provide a longer survival for some patients these approaches are associated with significant treatment-related adverse events and challenging to deliver in most centers. Therefore, it seems reasonable to develop treatment approaches that enhance the efficiency of RIT, while reducing its toxicity. In this paper, novel methods that improve the efficiency of RIT and reduce its toxicity through various mechanisms are reviewed. Further clinical development of these methods could expand the NHL patient groups eligible for receiving RIT, and even extend the use of RIT to new indications and disease groups in future.
Collapse
Affiliation(s)
- Mahsa Eskian
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,b Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - MirHojjat Khorasanizadeh
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,b Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Pier Luigi Zinzani
- c Institute of Hematology "L. e A. Seràgnoli", University of Bologna , Bologna , Italy
| | - Tim M Illidge
- d Manchester Academic Health Sciences Centre, University of Manchester, Christie NHS Foundation Trust , Manchester , UK
| | - Nima Rezaei
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,e Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,f Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
38
|
Brumano LP, da Silva FVS, Costa-Silva TA, Apolinário AC, Santos JHPM, Kleingesinds EK, Monteiro G, Rangel-Yagui CDO, Benyahia B, Junior AP. Development of L-Asparaginase Biobetters: Current Research Status and Review of the Desirable Quality Profiles. Front Bioeng Biotechnol 2019; 6:212. [PMID: 30687702 PMCID: PMC6335324 DOI: 10.3389/fbioe.2018.00212] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/21/2018] [Indexed: 01/23/2023] Open
Abstract
L-Asparaginase (ASNase) is a vital component of the first line treatment of acute lymphoblastic leukemia (ALL), an aggressive type of blood cancer expected to afflict over 53,000 people worldwide by 2020. More recently, ASNase has also been shown to have potential for preventing metastasis from solid tumors. The ASNase treatment is, however, characterized by a plethora of potential side effects, ranging from immune reactions to severe toxicity. Consequently, in accordance with Quality-by-Design (QbD) principles, ingenious new products tailored to minimize adverse reactions while increasing patient survival have been devised. In the following pages, the reader is invited for a brief discussion on the most recent developments in this field. Firstly, the review presents an outline of the recent improvements on the manufacturing and formulation processes, which can severely influence important aspects of the product quality profile, such as contamination, aggregation and enzymatic activity. Following, the most recent advances in protein engineering applied to the development of biobetter ASNases (i.e., with reduced glutaminase activity, proteolysis resistant and less immunogenic) using techniques such as site-directed mutagenesis, molecular dynamics, PEGylation, PASylation and bioconjugation are discussed. Afterwards, the attention is shifted toward nanomedicine including technologies such as encapsulation and immobilization, which aim at improving ASNase pharmacokinetics. Besides discussing the results of the most innovative and representative academic research, the review provides an overview of the products already available on the market or in the latest stages of development. With this, the review is intended to provide a solid background for the current product development and underpin the discussions on the target quality profile of future ASNase-based pharmaceuticals.
Collapse
Affiliation(s)
- Larissa Pereira Brumano
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Francisco Vitor Santos da Silva
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tales Alexandre Costa-Silva
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexsandra Conceição Apolinário
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - João Henrique Picado Madalena Santos
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Eduardo Krebs Kleingesinds
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gisele Monteiro
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carlota de Oliveira Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Brahim Benyahia
- Department of Chemical Engineering, Loughborough University, Loughborough, United Kingdom
| | - Adalberto Pessoa Junior
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
39
|
Santos JHPM, Torres-Obreque KM, Meneguetti GP, Amaro BP, Rangel-Yagui CO. Protein PEGylation for the design of biobetters: from reaction to purification processes. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000001009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
40
|
Adler J, Scheidt HA, Lemmnitzer K, Krueger M, Huster D. N-terminal lipid conjugation of amyloid β(1-40) leads to the formation of highly ordered N-terminally extended fibrils. Phys Chem Chem Phys 2018; 19:1839-1846. [PMID: 28000812 DOI: 10.1039/c6cp05982a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fibril formation of amyloid β(1-40) (Aβ(1-40)) peptides N-terminally lipid modified with saturated octanoyl or palmitoyl lipid chains was investigated. Lipid modification of Aβ(1-40) significantly accelerates the fibrillation kinetics of the Aβ peptides as revealed by ThT fluorescence. Electron microscopy and X-ray diffraction results indicate a heterogeneous cross-β structure of the fibrils formed by the lipid-conjugated peptides. Solid-state NMR was used to investigate structural features of these fibrils. The lipid moieties form dynamic and loosely structured heterogeneous lipid assemblies as inferred from 2H NMR of the deuterated lipid chains. 13C NMR studies of selected isotopic labels reveals that in addition to Phe19 and Val39, which are part of the canonical cross-β structure, also N-terminal residues (Ala2, Phe4, Val12) are found in β-strand conformation. This suggests that the increased hydrophobicity induced by the lipid modification, alters the energy landscape rendering an N-terminal extension of the β-sheet structure favorable. Furthermore, the fibrils formed by the Aβ-lipid hybrids are much more rigid than wildtype Aβ fibrils as inferred from NMR order parameter measurements. Taken together, increasing the local hydrophobicity of the Aβ N-terminus results in highly ordered but heterogeneous amyloid fibrils with extended N-terminal β-sheet structure.
Collapse
Affiliation(s)
- Juliane Adler
- Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany.
| | - Holger A Scheidt
- Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany.
| | - Katharina Lemmnitzer
- Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany.
| | - Martin Krueger
- Institute for Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Daniel Huster
- Institute for Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany.
| |
Collapse
|
41
|
Raman A, Jaime C, Puntes VF. Domain Formation and Conformational Changes in Gold Nanoparticle Conjugates Studied Using DPD Simulations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:14502-14512. [PMID: 29199832 DOI: 10.1021/acs.langmuir.7b03318] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A gold nanoparticle (AuNP) conjugate formed with 11-mercaptoundecanoic acid (MUA) and thiolated polyethylene glycol (SH-PEG) is simulated using dissipative particle dynamics (DPD) methods, obtaining an excellent agreement with previous experimental observations. The simulations cover the isolated components (AuNP, MUA, and SH-PEG), as well as pairs of components, and finally the all three components at the same time. In this latter case, changes in the order of addition of MUA and SH-PEG over the AuNP are also considered. The AuNP is formed by independent gold beads and keeps an almost spherical shape throughout the simulation. MUA forms micelles of four to six MUA units when dispersed in water, while SH-PEG stays individually and well solvated. When exposed to AuNP, both molecules show a tendency to form patches on the surface. SH-PEG displays two different conformations (radial and tangential) depending on its relative concentration and the presence of other molecules at the NP surface. When combined at subsaturation concentrations, MUA arrives faster to the AuNP surface than SH-PEG and forms patches while SH-PEG occupies the remaining free surface. In these conditions, the order of addition of the different components partially alters these results. When SH-PEG is added over an already formed MUA/AuNP partial layer, it adopts a radial conformation over the MUA formed patches; on the contrary, if MUA is added over an already formed SH-PEG/AuNP partial layer, much less SH-PEGs adopt a radial conformation and MUA patches are significantly smaller.
Collapse
Affiliation(s)
- Asli Raman
- Department of Chemistry, Universitat Autònoma de Barcelona , 08193 Bellaterra, Spain
| | - Carlos Jaime
- Department of Chemistry, Universitat Autònoma de Barcelona , 08193 Bellaterra, Spain
| | - Victor F Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2-BIST) , Campus UAB, 08193 Bellaterra, Barcelona, Spain
- Vall d'Hebron Institut de Recerca (CIBBIM - VHIR) , 08035 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA) , P. Lluis Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
42
|
Maniar KH, Jones IA, Gopalakrishna R, Vangsness CT. Lowering side effects of NSAID usage in osteoarthritis: recent attempts at minimizing dosage. Expert Opin Pharmacother 2017; 19:93-102. [PMID: 29212381 DOI: 10.1080/14656566.2017.1414802] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Osteoarthritis is a burdensome disease that causes progressive damage to articular cartilage. Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the preferred treatments for symptomatic relief. However, NSAIDs can cause serious dose-dependent side effects, which has prompted experts to recommend the minimization of NSAID dosage. AREAS COVERED This review focuses on three broad strategies that are currently being investigated or implemented to minimize NSAID dosage: nano-formulation, encapsulation, and topical delivery. The benefits, challenges and current status of these methods are discussed. EXPERT OPINION Multiple strategies are under investigation to lower NSAID dosage. There is great potential in developing formulations that utilize more than one of these strategies together. However, there are challenges to developing these lower dose preparations. In order to maximize the clinical potential of the abundance of NSAIDs that are both available and being developed, there is a major need for additional clinical studies directly comparing safety and efficacy of different preparations.
Collapse
Affiliation(s)
- Kevin H Maniar
- a Keck School of Medicine of USC , Los Angeles , CA , USA
| | - Ian A Jones
- b Department of Orthopaedic Surgery , Keck School of Medicine of USC , Los Angeles , CA , USA
| | - Rayudu Gopalakrishna
- c Department of Integrative Anatomical Sciences , Keck School of Medicine of USC , Los Angeles , CA , USA
| | - C Thomas Vangsness
- b Department of Orthopaedic Surgery , Keck School of Medicine of USC , Los Angeles , CA , USA
| |
Collapse
|
43
|
Guo Z, You L, Shi C, Song M, Gao M, Xu D, Peng C, Zhuang R, Liu T, Su X, Du J, Zhang X. Development of a New FR-Targeting Agent 99mTc-HYNFA with Improved Imaging Contrast and Comparison of Multimerization and/or PEGylation Strategies for Radio-Folate Modification. Mol Pharm 2017; 14:3780-3788. [PMID: 28969422 DOI: 10.1021/acs.molpharmaceut.7b00536] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This study aims to develop a new folate receptor (FR)-targeting agent for SPECT imaging with improved contrast and evaluate the modification strategies of multimerization and/or PEGylation in the development of new radio-folates. A series of novel folate derivatives have been synthesized and radiolabeled with 99mTc using tricine and TPPTS as coligands. To better investigate their pharmacokinetics, cell uptake, biodistribution, and microSPECT/CT imaging were evaluated. Four radioligands displayed high KB cell uptake after incubation for 2 and 4 h. Presaturated with excess folic acid (FA) resulted in a significant blocking effect in KB cells, indicating specificity of these radioligands toward FR. Biodistribution and microSPECT imaging studies in KB tumor-bearing mice showed that the folate conjugate 99mTc-HYNFA with poly(ethylene glycol) (PEG) and triazole linkage displayed the highest tumor uptake (16.30 ± 2.01 %ID/g at 2 h p.i. and 14.9 ± 0.62 %ID/g at 4 h p.i. in mice biodistribution) and best imaging contrast, indicating promising application prospect. More interestingly, the in vivo performance of this monomeric 99mTc-HYNFA was much better than that of FA multimers and non-PEGylated monomers, suggesting that multimerization may not be a feasible method for the design of radio-folates. PEG linkage rather than FA multimerization should be taken into consideration in the development of folate-based radiopharmaceuticals in the future.
Collapse
Affiliation(s)
- Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China.,Department of Isotope, China Institute of Atomic Energy , P.O. Box 2108, Beijing 102413, China
| | - Linyi You
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Changrong Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Manli Song
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China.,The First Affiliated Hospital of Zhengzhou University , Zhengzhou 450000, China
| | - Mengna Gao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Duo Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Chenyu Peng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Rongqiang Zhuang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Ting Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Xinhui Su
- Zhongshan Hospital Affiliated of Xiamen University , Hubin South Road, Xiamen 361004, China
| | - Jin Du
- Department of Isotope, China Institute of Atomic Energy , P.O. Box 2108, Beijing 102413, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University , 4221-116 Xiang'An South Road, Xiamen 361102, China
| |
Collapse
|
44
|
Holub JM. Small Scaffolds, Big Potential: Developing Miniature Proteins as Therapeutic Agents. Drug Dev Res 2017; 78:268-282. [PMID: 28799168 DOI: 10.1002/ddr.21408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Preclinical Research Miniature proteins are a class of oligopeptide characterized by their short sequence lengths and ability to adopt well-folded, three-dimensional structures. Because of their biomimetic nature and synthetic tractability, miniature proteins have been used to study a range of biochemical processes including fast protein folding, signal transduction, catalysis and molecular transport. Recently, miniature proteins have been gaining traction as potential therapeutic agents because their small size and ability to fold into defined tertiary structures facilitates their development as protein-based drugs. This research overview discusses emerging developments involving the use of miniature proteins as scaffolds to design novel therapeutics for the treatment and study of human disease. Specifically, this review will explore strategies to: (i) stabilize miniature protein tertiary structure; (ii) optimize biomolecular recognition by grafting functional epitopes onto miniature protein scaffolds; and (iii) enhance cytosolic delivery of miniature proteins through the use of cationic motifs that facilitate endosomal escape. These objectives are discussed not only to address challenges in developing effective miniature protein-based drugs, but also to highlight the tremendous potential miniature proteins hold for combating and understanding human disease. Drug Dev Res 78 : 268-282, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Justin M Holub
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
45
|
Ronda L, Marchetti M, Piano R, Liuzzi A, Corsini R, Percudani R, Bettati S. A Trivalent Enzymatic System for Uricolytic Therapy of HPRT Deficiency and Lesch-Nyhan Disease. Pharm Res 2017; 34:1477-1490. [PMID: 28508122 PMCID: PMC5445154 DOI: 10.1007/s11095-017-2167-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/25/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE Because of the evolutionary loss of the uricolytic pathway, humans accumulate poorly soluble urate as the final product of purine catabolism. Restoration of uricolysis through enzyme therapy is a promising treatment for severe hyperuricemia caused by deficiency of hypoxanthine-guanine phosphoribosyltransferase (HPRT). To this end, we studied the effect of PEG conjugation on the activity and stability of the enzymatic complement required for conversion of urate into the more soluble (S)-allantoin. METHODS We produced in recombinant form three zebrafish enzymes required in the uricolytic pathway. We carried out a systematic study of the effect of PEGylation on the function and stability of the three enzymes by varying PEG length, chemistry and degree of conjugation. We assayed in vitro the uricolytic activity of the PEGylated enzymatic triad. RESULTS We defined conditions that allow PEGylated enzymes to retain native-like enzymatic activity even after lyophilization or prolonged storage. A combination of the three enzymes in an appropriate ratio allowed efficient conversion of urate to (S)-allantoin with no accumulation of intermediate metabolites. CONCLUSIONS Pharmaceutical restoration of the uricolytic pathway is a viable approach for the treatment of severe hyperuricemia.
Collapse
Affiliation(s)
- Luca Ronda
- Department of Medicine and Surgery,, University of Parma, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Marialaura Marchetti
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma,, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Riccardo Piano
- Department of Medicine and Surgery,, University of Parma, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Anastasia Liuzzi
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma,, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Romina Corsini
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma,, Parco Area delle Scienze 23/A, 43124, Parma, Italy
| | - Riccardo Percudani
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma,, Parco Area delle Scienze 23/A, 43124, Parma, Italy.
| | - Stefano Bettati
- Department of Medicine and Surgery,, University of Parma, Parco Area delle Scienze 23/A, 43124, Parma, Italy. .,National Institute of Biostructures and Biosystems, Viale Medaglie d'Oro 305, 00136, Rome, Italy.
| |
Collapse
|
46
|
Iwamura T, Narumi H, Suzuki T, Yanai H, Mori K, Yamashita K, Tsushima Y, Asano T, Izawa A, Momen S, Nishimura K, Tsuchiyama H, Uchida M, Yamashita Y, Okano K, Taniguchi T. Novel pegylated interferon-β as strong suppressor of the malignant ascites in a peritoneal metastasis model of human cancer. Cancer Sci 2017; 108:581-589. [PMID: 28129467 PMCID: PMC5406538 DOI: 10.1111/cas.13176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/15/2017] [Accepted: 01/21/2017] [Indexed: 12/19/2022] Open
Abstract
Malignant ascites manifests as an end‐stage event during the progression of a number of cancers and lacks a generally accepted standard therapy. Interferon‐β (IFN‐β) has been used to treat several cancer indications; however, little is known about the efficacy of IFN‐β on malignant ascites. In the present study, we report on the development of a novel, engineered form of human and murine IFN‐β, each conjugated with a polyethylene glycol molecule (PEG‐hIFN‐β and PEG‐mIFN‐β, respectively). We provide evidence that these IFN‐β molecules retain anti‐viral potency comparable to unmodified IFN‐β in vitro and manifested improved pharmacokinetics in vivo. Interestingly, PEG‐mIFN‐β significantly inhibited the accumulation of ascites fluid and vascular permeability of the peritoneal membrane in models of ovarian cancer and gastric cancer cell xenograft mice. We further show that PEG‐hIFN‐β directly suppresses VEGF165‐induced hyperpermeability in a monolayer of human vascular endothelial cells and that PEG‐mIFN‐β enhanced gene expression for a number of cell adhesion related molecules in mouse vascular endothelial cells. Taken together, these findings unveil a hitherto unrecognized potential of IFN‐β in maintaining vascular integrity, and provide proof‐of‐mechanism for a novel and long‐acting pegylated hIFN‐β for the therapeutic treatment of malignant ascites.
Collapse
Affiliation(s)
- Tomokatsu Iwamura
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Hideki Narumi
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Tomohiko Suzuki
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Hideyuki Yanai
- Department of Molecular Immunology, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo, Japan
| | - Katsuyuki Mori
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Koji Yamashita
- Pharmaceuticals Technical Development Department, Toray Industries, Kamakura, Kanagawa, Japan
| | - Yoshiaki Tsushima
- Pharmaceuticals Technical Development Department, Toray Industries, Kamakura, Kanagawa, Japan
| | - Tomomi Asano
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Akiko Izawa
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Shinobu Momen
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Kazumi Nishimura
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Hiromi Tsuchiyama
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Masashi Uchida
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Yuji Yamashita
- Pharmaceuticals Technical Development Department, Toray Industries, Kamakura, Kanagawa, Japan
| | - Kiyoshi Okano
- Pharmaceutical Research Laboratory, Toray Industries, Kamakura, Kanagawa, Japan
| | - Tadatsugu Taniguchi
- Department of Molecular Immunology, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, Tokyo, Japan
| |
Collapse
|
47
|
Surface modification of lipid-based nanocarriers for cancer cell-specific drug targeting. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0329-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
48
|
Nie Q, Jia D, Yang H, Feng Y, Fan Q, Shi Q, Wan L, Lu X. Conjugation to 10 kDa Linear PEG Extends Serum Half-Life and Preserves the Receptor-Binding Ability of mmTRAIL with Minimal Stimulation of PEG-Specific Antibodies. Mol Pharm 2017; 14:502-512. [PMID: 28029256 DOI: 10.1021/acs.molpharmaceut.6b00964] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The poor in vivo potencies of most therapeutic proteins might be attributed to their short serum half-lives. PEGylation is a well-established method and has been clinically proven to improve pharmacokinetics. mmTRAIL exhibited supercytotoxicity in a variety of tumor cells, but its serum half-life was less than 10 min in mice. Here, mmTRAIL-5K, mmTRAIL-10K, and mmTRAIL-20K were produced by N-terminus-specific PEGylation of mmTRAIL with 5, 10, or 20 kDa mPEG, respectively. The particle sizes of mmTRAIL-5K, mmTRAIL-10K, and mmTRAIL-20K were 9.09 ± 2.76, 12.62 ± 4.05, and 15.68 ± 4.95 nm, which were higher than the threshold (∼7 nm) of renal clearance. Accordingly, mmTRAIL-5K exhibited a serum half-life of 30 min only 3 times longer than that of mmTRAIL. However, both mmTRAIL-10K and mmTRAIL-20K exhibited similar serum half-lives ranging from 350 to 400 min, indicating that PEGylation with 10 or 20 kDa mPEG significantly improved the pharmacokinetics of mmTRAIL. However, death receptor binding of mmTRAIL-20K was reduced 5- to 8-fold, resulting in a 3-fold reduction of cytotoxicity. Additionally, repeated administration of mmTRAIL-20K elicited both mPEG-specific IgG and IgM antibody responses in rats. In contrast, the receptor binding and cytotoxicity of mmTRAIL-10K were similar to those of mmTRAIL. Repeated administration of mmTRAIL-10K did not obviously stimulate mPEG-specific antibody responses in rats and rhesus monkeys. Of the three PEGylated mmTRAIL analogues, mmTRAIL-10K exerted the greatest tumor suppression in mice bearing human tumor xenografts. These results demonstrated that conjugation of mmTRAIL to 10 kDa mPEG was better than that to 5 or 20 kDa mPEG for enhancing antitumor effects.
Collapse
Affiliation(s)
- Qianxue Nie
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Dianlong Jia
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Hao Yang
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Yanru Feng
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Qing Fan
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Qiuxiao Shi
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Lin Wan
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| | - Xiaofeng Lu
- Key Lab of Transplant Engineering and Immunology, MOH, West China Hospital, Sichuan University , Chengdu 610041, China
| |
Collapse
|
49
|
Bastos V, Ferreira-de-Oliveira JMP, Carrola J, Daniel-da-Silva AL, Duarte IF, Santos C, Oliveira H. Coating independent cytotoxicity of citrate- and PEG-coated silver nanoparticles on a human hepatoma cell line. J Environ Sci (China) 2017; 51:191-201. [PMID: 28115130 DOI: 10.1016/j.jes.2016.05.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/21/2016] [Accepted: 05/28/2016] [Indexed: 06/06/2023]
Abstract
The antibacterial potential of silver nanoparticles (AgNPs) resulted in their increasing incorporation into consumer, industrial and biomedical products. Therefore, human and environmental exposure to AgNPs (either as an engineered product or a contaminant) supports the emergent research on the features conferring them different toxicity profiles. In this study, 30nm AgNPs coated with citrate or poly(ethylene glycol) (PEG) were used to assess the influence of coating on the effects produced on a human hepatoma cell line (HepG2), namely in terms of viability, apoptosis, apoptotic related genes, cell cycle and cyclins gene expression. Both types of coated AgNPs decreased cell proliferation and viability with a similar toxicity profile. At the concentrations used (11 and 5μg/mL corresponding to IC50 and ~IC10 levels, respectively) the amount of cells undergoing apoptosis was not significant and the apoptotic related genes BCL2 (anti-apoptotic gene) and BAX (pro-apoptotic gene) were both downregulated. Moreover, both AgNPs affected HepG2 cell cycle progression at the higher concentration (11μg/mL) by increasing the percentage of cells in S (synthesis phase) and G2 (Gap 2 phase) phases. Considering the cell-cycle related genes, the expression of cyclin B1 and cyclin E1 genes were decreased. Thus, this work has shown that citrate- and PEG-coated AgNPs impact on HepG2 apoptotic gene expression, cell cycle dynamics and cyclin regulation in a similar way. More research is needed to determine the properties that confer AgNPs at lower toxicity, since their use has proved helpful in several industrial and biomedical contexts.
Collapse
Affiliation(s)
- Verónica Bastos
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - José M P Ferreira-de-Oliveira
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joana Carrola
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana L Daniel-da-Silva
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Iola F Duarte
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Conceição Santos
- Department of Biology, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal.
| | - Helena Oliveira
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
50
|
Roncador A, Oppici E, Talelli M, Pariente AN, Donini M, Dusi S, Voltattorni CB, Vicent MJ, Cellini B. Use of polymer conjugates for the intraperoxisomal delivery of engineered human alanine:glyoxylate aminotransferase as a protein therapy for primary hyperoxaluria type I. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:897-907. [PMID: 27993722 DOI: 10.1016/j.nano.2016.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/25/2016] [Accepted: 12/07/2016] [Indexed: 01/22/2023]
Abstract
Alanine:glyoxylate aminotransferase (AGT) is a liver peroxisomal enzyme whose deficit causes the rare disorder Primary Hyperoxaluria Type I (PH1). We now describe the conjugation of poly(ethylene glycol)-co-poly(L-glutamic acid) (PEG-PGA) block-co-polymer to AGT via the formation of disulfide bonds between the polymer and solvent-exposed cysteine residues of the enzyme. PEG-PGA conjugation did not affect AGT structural/functional properties and allowed the enzyme to be internalized in a cellular model of PH1 and to restore glyoxylate-detoxification. The insertion of the C387S/K390S amino acid substitutions, known to favor interaction with the peroxisomal import machinery, reduced conjugation efficiency, but endowed conjugates with the ability to reach the peroxisomal compartment. These results, along with the finding that conjugates are hemocompatible, stable in plasma, and non-immunogenic, hold promise for the development of polypeptide-based AGT conjugates as a therapeutic option for PH1 patients and represent the base for applications to other diseases related to deficits in peroxisomal proteins.
Collapse
Affiliation(s)
- Alessandro Roncador
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy
| | - Elisa Oppici
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy
| | - Marina Talelli
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Amaya Niño Pariente
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain
| | - Marta Donini
- Department of Medicine, Section of General Pathology, University of Verona, Verona (VR), Italy
| | - Stefano Dusi
- Department of Medicine, Section of General Pathology, University of Verona, Verona (VR), Italy
| | - Carla Borri Voltattorni
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy
| | - María J Vicent
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe (CIPF), Valencia, Spain.
| | - Barbara Cellini
- Neuroscience, Biomedicine and Movement Sciences Department, Section of Biological Chemistry, University of Verona, Verona (VR), Italy.
| |
Collapse
|