1
|
Rizvi SFA, Zhang H, Fang Q. Engineering peptide drug therapeutics through chemical conjugation and implication in clinics. Med Res Rev 2024; 44:2420-2471. [PMID: 38704826 DOI: 10.1002/med.22046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2024] [Accepted: 04/21/2024] [Indexed: 05/07/2024]
Abstract
The development of peptide drugs has made tremendous progress in the past few decades because of the advancements in modification chemistry and analytical technologies. The novel-designed peptide drugs have been modified through various biochemical methods with improved diagnostic, therapeutic, and drug-delivery strategies. Researchers found it a helping hand to overcome the inherent limitations of peptides and bring continued advancements in their applications. Furthermore, the emergence of peptide-drug conjugates (PDCs)-utilizes target-oriented peptide moieties as a vehicle for cytotoxic payloads via conjugation with cleavable chemical agents, resulting in the key foundation of the new era of targeted peptide drugs. This review summarizes the various classifications of peptide drugs, suitable chemical modification strategies to improve the ADME (adsorption, distribution, metabolism, and excretion) features of peptide drugs, and recent (2015-early 2024) progress/achievements in peptide-based drug delivery systems as well as their fruitful implication in preclinical and clinical studies. Furthermore, we also summarized the brief description of other types of PDCs, including peptide-MOF conjugates and peptide-UCNP conjugates. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development and progress toward a bright future of novel peptide drugs.
Collapse
Affiliation(s)
- Syed Faheem Askari Rizvi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Qiu M, Zou J, Yang Z, Yang D, Wang R, Guo H. Strategies for Targeting Peptide-Modified Exosomes and Their Applications in the Lungs. Int J Nanomedicine 2024; 19:8175-8188. [PMID: 39157733 PMCID: PMC11328869 DOI: 10.2147/ijn.s472038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024] Open
Abstract
Exosomes belong to a subgroup of extracellular vesicles secreted by various cells and are involved in intercellular communication and material transfer. In recent years, exosomes have been used as drug delivery carriers because of their natural origin, high stability, low immunogenicity and high engineering ability. However, achieving targeted drug delivery with exosomes remains challenging. In this paper, a phage display technology was used to screen targeted peptides, and different surface modification strategies of targeted peptide exosomes were reviewed. In addition, the application of peptide-targeted exosomes in pulmonary diseases was also summarised.
Collapse
Affiliation(s)
- Min Qiu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Inner Mongolia, People’s Republic of China
- College of Pharmacy, Baotou Medical College, Baotou, People’s Republic of China
| | - Jinru Zou
- College of Pharmacy, Baotou Medical College, Baotou, People’s Republic of China
| | - Zheng Yang
- The First Affiliated Hospital, Baotou Medical College, Baotou, People’s Republic of China
| | - Dan Yang
- College of Pharmacy, Baotou Medical College, Baotou, People’s Republic of China
| | - Rui Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Inner Mongolia, People’s Republic of China
| | - Haie Guo
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Inner Mongolia, People’s Republic of China
- Agriculture, Animal Husbandry and Science and Technology Bureau of Liangcheng County, Ulanqab, Inner Mongolia, People’s Republic of China
| |
Collapse
|
3
|
Rizvi SF, Zhang L, Zhang H, Fang Q. Peptide-Drug Conjugates: Design, Chemistry, and Drug Delivery System as a Novel Cancer Theranostic. ACS Pharmacol Transl Sci 2024; 7:309-334. [PMID: 38357281 PMCID: PMC10863443 DOI: 10.1021/acsptsci.3c00269] [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: 10/04/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 02/16/2024]
Abstract
The emergence of peptide-drug conjugates (PDCs) that utilize target-oriented peptide moieties as carriers of cytotoxic payloads, interconnected with various cleavable/noncleavable linkers, resulted in the key-foundation of the new era of targeted therapeutics. They are capable of retaining the integrity of conjugates in the blood circulatory system as well as releasing the drugs at the tumor microenvironment. Other valuable advantages are specificity and selectivity toward targeted-receptors, higher penetration ability, and drug-loading capacity, making them a suitable candidate to play their vital role as promising carrier agents. In this review, we summarized the types of cell-targeting (CTPs) and cell-penetrating peptides (CPPs) that have broad applications in the advancement of targeted drug-delivery systems (DDS). Moreover, the techniques to overcome the limitations of peptide-chemistry for their extensive implementation to construct the PDCs. Besides this, the diversified breakthrough of linker chemistry, and ample knowledge of various cytotoxic payloads used in PDCs in recent years, as well as the mechanism of action of PDCs was critically discussed. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development, also their progression toward a bright future for PDCs as novel theranostics in clinical practice.
Collapse
Affiliation(s)
- Syed Faheem
Askari Rizvi
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, and
Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
- Institute
of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, 54000, Punjab Pakistan
| | - Linjie Zhang
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| | - Haixia Zhang
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| | - Quan Fang
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, and
Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| |
Collapse
|
4
|
Mező G, Gomena J, Ranđelović I, Dókus EL, Kiss K, Pethő L, Schuster S, Vári B, Vári-Mező D, Lajkó E, Polgár L, Kőhidai L, Tóvári J, Szabó I. Oxime-Linked Peptide-Daunomycin Conjugates as Good Tools for Selection of Suitable Homing Devices in Targeted Tumor Therapy: An Overview. Int J Mol Sci 2024; 25:1864. [PMID: 38339141 PMCID: PMC10855781 DOI: 10.3390/ijms25031864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Chemotherapy is still one of the main therapeutic approaches in cancer therapy. Nevertheless, its poor selectivity causes severe toxic side effects that, together with the development of drug resistance in tumor cells, results in a limitation for its application. Tumor-targeted drug delivery is a possible choice to overcome these drawbacks. As well as monoclonal antibodies, peptides are promising targeting moieties for drug delivery. However, the development of peptide-drug conjugates (PDCs) is still a big challenge. The main reason is that the conjugates have to be stable in circulation, but the drug or its active metabolite should be released efficiently in the tumor cells. For this purpose, suitable linker systems are needed that connect the drug molecule with the homing peptide. The applied linker systems are commonly categorized as cleavable and non-cleavable linkers. Both the groups possess advantages and disadvantages that are summarized briefly in this manuscript. Moreover, in this review paper, we highlight the benefit of oxime-linked anthracycline-peptide conjugates in the development of PDCs. For instance, straightforward synthesis as well as a conjugation reaction proceed in excellent yields, and the autofluorescence of anthracyclines provides a good tool to select the appropriate homing peptides. Furthermore, we demonstrate that these conjugates can be used properly in in vivo studies. The results indicate that the oxime-linked PDCs are potential candidates for targeted tumor therapy.
Collapse
Affiliation(s)
- Gábor Mező
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Institute of Chemistry, ELTE, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Jacopo Gomena
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Institute of Chemistry, ELTE, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Ivan Ranđelović
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
| | - Endre Levente Dókus
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
| | - Krisztina Kiss
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, 1111 Budapest, Hungary
| | - Lilla Pethő
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
| | - Sabine Schuster
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Institute of Chemistry, ELTE, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Balázs Vári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
- School of Ph.D. Studies, Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Diána Vári-Mező
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
- School of Ph.D. Studies, Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Eszter Lajkó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary; (E.L.); (L.P.); (L.K.)
| | - Lívia Polgár
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary; (E.L.); (L.P.); (L.K.)
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary; (E.L.); (L.P.); (L.K.)
| | - József Tóvári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
- School of Ph.D. Studies, Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Ildikó Szabó
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
| |
Collapse
|
5
|
Wang W, Chen C, Luo J, Tang C, Zheng Y, Yan S, Yuan Y, Zhu M, Diao X, Hang T, Wang H. Metabolism investigation of the peptide-drug conjugate LN005 in rats using UHPLCHRMS. J Pharm Biomed Anal 2024; 238:115860. [PMID: 37979524 DOI: 10.1016/j.jpba.2023.115860] [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: 09/16/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
LN005, as a peptide-drug conjugate (PDC), is a conjugate of the homing peptide VAP and doxorubicin (DOX). The exceptional targeting ability of the homing peptide VAP is directed toward glucose-regulated protein (GRP78), a highly expressed protein primarily found in the endoplasmic reticulum of various solid tumors. However, there are limited reports regarding the metabolism of peptide-drug conjugates (PDCs), and the in vivo metabolism of LN005 has yet to be investigated. After intravenous injection of 18 mg/kg LN005 in SD rats, biological samples including plasma, urine, fecal, and bile samples, were collected and analyzed by ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). A total of 11 possible metabolites of LN005 were identified. Unchanged LN005 was found to be the main component in rat blood and urine, accounting for 46.46% and 63.79% of the total peak areas, respectively. M1057 was the most abundant metabolite in feces, accounting for 57.65% of the total peak area. Only one metabolite, M398, was identified in rat bile. The metabolism of LN005 is closely related to DOX, and the primary metabolic pathways involved oxidative deamination or hydrolysis, reductive glycosidic cleavage, hydrolytic glycosidic cleavage, and dehydrogenation.
Collapse
Affiliation(s)
- Weiqiang Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Chong Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jing Luo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; Shanghai Whittlong Pharmaceutical Institute Co., Ltd, Shanghai, China
| | | | - Yuandong Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shu Yan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yali Yuan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | - Xingxing Diao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Taijun Hang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China.
| | - Hao Wang
- Shanghai Whittlong Pharmaceutical Institute Co., Ltd, Shanghai, China; National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China.
| |
Collapse
|
6
|
Zhou J, Xie Z, Wang J, Zeng Z, Hu Z, Zhong L, Yang Q, Shi W, Qian H. Design, synthesis and bioactivity evaluation of novel fusion peptides and their CPT conjugates inducing effective anti-tumor responses on HER2 positive tumors. Eur J Med Chem 2024; 264:116032. [PMID: 38104378 DOI: 10.1016/j.ejmech.2023.116032] [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: 10/27/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) represents an ideal target for antibody drug development, abnormal expression of the HER2 gene is associated with multiple tumor types. Pertuzumab, as the first monoclonal antibody inhibitor of HER2 dimerization, has been FDA-approved for HER2-positive patients. In order to enhance the activity of HER2-targeted peptide-drug conjugates (PDCs) developed based on pertuzumab, a novel class of conjugates 1-9 was designed and synthesized by fusing the N-terminal peptide sequence of the second mitochondria-derived activator of caspases (SMAC) with P1, followed by conjugation with CPT molecules. Compound 4 exhibited excellent in vitro anti-tumor activity across the three HER2-positive cell lines, comparable to the activity of CPT. Apoptosis induction assays indicated that the synergistic effect of the SMAC sequence enhanced the pro-apoptotic activity of the conjugate. Western Blot analysis and Caspase activity studies validated the mechanism through which SMAC peptides, in synergy with CPT, enhance the activity of PDCs. In vivo studies demonstrated that compound 4 possesses superior anti-tumor activity compared to CPT and can effectively mitigate potential renal toxicity associated with free SMAC peptides. In conclusion, conjugate 4 exhibited excellent anti-tumor activity both in vitro and in vivo, offering potential for further development as a novel peptide-conjugated drug.
Collapse
Affiliation(s)
- Jiaqi Zhou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Zhancheng Xie
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Jialing Wang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Zeqi Zeng
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Zhipeng Hu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Li Zhong
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Qimeng Yang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
7
|
Wang Z, Li H, Gou L, Li W, Wang Y. Antibody-drug conjugates: Recent advances in payloads. Acta Pharm Sin B 2023; 13:4025-4059. [PMID: 37799390 PMCID: PMC10547921 DOI: 10.1016/j.apsb.2023.06.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/30/2023] [Accepted: 06/23/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody‒drug conjugates (ADCs), which combine the advantages of monoclonal antibodies with precise targeting and payloads with efficient killing, show great clinical therapeutic value. The ADCs' payloads play a key role in determining the efficacy of ADC drugs and thus have attracted great attention in the field. An ideal ADC payload should possess sufficient toxicity, low immunogenicity, high stability, and modifiable functional groups. Common ADC payloads include tubulin inhibitors and DNA damaging agents, with tubulin inhibitors accounting for more than half of the ADC drugs in clinical development. However, due to clinical limitations of traditional ADC payloads, such as inadequate efficacy and the development of acquired drug resistance, novel highly efficient payloads with diverse targets and reduced side effects are being developed. This perspective summarizes the recent research advances of traditional and novel ADC payloads with main focuses on the structure-activity relationship studies, co-crystal structures, and designing strategies, and further discusses the future research directions of ADC payloads. This review also aims to provide valuable references and future directions for the development of novel ADC payloads that will have high efficacy, low toxicity, adequate stability, and abilities to overcome drug resistance.
Collapse
Affiliation(s)
- Zhijia Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| | - Hanxuan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lantu Gou
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
8
|
Ma W, Yang Y, Liu Z, Zhao R, Wan Q, Chen X, Tang B, Zhou Y, Lin Y. Self-Assembled Multivalent Aptamer Drug Conjugates: Enhanced Targeting and Cytotoxicity for HER2-Positive Gastric Cancer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43359-43373. [PMID: 37670592 DOI: 10.1021/acsami.3c07344] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Antibody drug conjugates (ADCs) have shown promise to be the mainstream chemotherapeutics for advanced HER2-positive cancers, yet the issues of poor drug delivery efficiency, limited chemotherapeutic effects, severe immune responses, and drug resistance remain to be addressed before the clinical applications of ADCs. The DNA aptamer-guided drug conjugates (ApDCs) are receiving growing attention for specific tumors due to their excellent tumor affinity and low cost. Therefore, developing a multivalent ApDC nanomedicine by combining anti-HER2 aptamer (HApt), tetrahedral framework nucleic acid (tFNA), and deruxtecan (Dxd) together to form HApt-tFNA@Dxd might help to address these concerns. In this study, the HER2-targeted DNA aptamer modified DNA tetrahedron (HApt-tFNA) was employed as a system for drug delivery, and the adoption of tFNA could effectively enlarge the drug-loading rate compared to aptamer-guided ApDCs previously reported. Compared with free Dxd and tFNA@Dxd, HApt-tFNA@Dxd showed better structural stability, excellent targeted cytotoxicity to HER2-positive gastric cancer, and increased tissue aggregation ability in tumors. These features and superiorities make HApt-tFNA@Dxd a promising chemotherapeutic medicine for HER2-positive tumors. Our work developed a new targeting nanomedicine by combining DNA nanomaterials and chemotherapeutic agents, which represents a critical advance toward developing novel DNA-based nanomaterials and promoting their potential applications for HER2-positive cancer therapy.
Collapse
Affiliation(s)
- Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Yuting Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Rui Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Qianyi Wan
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xingyu Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Yong Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
9
|
Tiwari H, Rai N, Singh S, Gupta P, Verma A, Singh AK, Kajal, Salvi P, Singh SK, Gautam V. Recent Advances in Nanomaterials-Based Targeted Drug Delivery for Preclinical Cancer Diagnosis and Therapeutics. Bioengineering (Basel) 2023; 10:760. [PMID: 37508788 PMCID: PMC10376516 DOI: 10.3390/bioengineering10070760] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Nano-oncology is a branch of biomedical research and engineering that focuses on using nanotechnology in cancer diagnosis and treatment. Nanomaterials are extensively employed in the field of oncology because of their minute size and ultra-specificity. A wide range of nanocarriers, such as dendrimers, micelles, PEGylated liposomes, and polymeric nanoparticles are used to facilitate the efficient transport of anti-cancer drugs at the target tumor site. Real-time labeling and monitoring of cancer cells using quantum dots is essential for determining the level of therapy needed for treatment. The drug is targeted to the tumor site either by passive or active means. Passive targeting makes use of the tumor microenvironment and enhanced permeability and retention effect, while active targeting involves the use of ligand-coated nanoparticles. Nanotechnology is being used to diagnose the early stage of cancer by detecting cancer-specific biomarkers using tumor imaging. The implication of nanotechnology in cancer therapy employs photoinduced nanosensitizers, reverse multidrug resistance, and enabling efficient delivery of CRISPR/Cas9 and RNA molecules for therapeutic applications. However, despite recent advancements in nano-oncology, there is a need to delve deeper into the domain of designing and applying nanoparticles for improved cancer diagnostics.
Collapse
Affiliation(s)
- Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Nilesh Rai
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh Kumar Singh
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Sciences, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Kajal
- Department of Agriculture Biotechnology, National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar 140306, India
| | - Prafull Salvi
- Department of Agriculture Biotechnology, National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar 140306, India
| | - Santosh Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
10
|
Transcytosable Peptide-Paclitaxel Prodrug Nanoparticle for Targeted Treatment of Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:ijms24054646. [PMID: 36902076 PMCID: PMC10003159 DOI: 10.3390/ijms24054646] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an extremely aggressive subtype associated with a poor prognosis. At present, the treatment for TNBC mainly relies on surgery and traditional chemotherapy. As a key component in the standard treatment of TNBC, paclitaxel (PTX) effectively inhibits the growth and proliferation of tumor cells. However, the application of PTX in clinical treatment is limited due to its inherent hydrophobicity, weak penetrability, nonspecific accumulation, and side effects. To counter these problems, we constructed a novel PTX conjugate based on the peptide-drug conjugates (PDCs) strategy. In this PTX conjugate, a novel fused peptide TAR consisting of a tumor-targeting peptide, A7R, and a cell-penetrating peptide, TAT, is used to modify PTX. After modification, this conjugate is named PTX-SM-TAR, which is expected to improve the specificity and penetrability of PTX at the tumor site. Depending on hydrophilic TAR peptide and hydrophobic PTX, PTX-SM-TAR can self-assemble into nanoparticles and improve the water solubility of PTX. In terms of linkage, the acid- and esterase-sensitive ester bond was used as the linking bond, with which PTX-SM-TAR NPs could remain stable in the physiological environment, whereas PTX-SM-TAR NPs could be broken and PTX be released at the tumor site. A cell uptake assay showed that PTX-SM-TAR NPs were receptor-targeting and could mediate endocytosis by binding to NRP-1. The vascular barrier, transcellular migration, and tumor spheroids experiments showed that PTX-SM-TAR NPs exhibit great transvascular transport and tumor penetration ability. In vivo experiments, PTX-SM-TAR NPs showed higher antitumor effects than PTX. As a result, PTX-SM-TAR NPs may overcome the shortcomings of PTX and present a new transcytosable and targeted delivery system for PTX in TNBC treatment.
Collapse
|
11
|
Han G, Wang J, Li Y, Chen Z, Xu X, Liu T, Wang Y, Bai F, Liu K, Zhao Y. Novel Peptide from the Hydrolysate of Hybrid Sturgeon ( Acipenseridae) Spinal Cord: Isolation, Identification, and Anti-proliferative Effects in Human Cervix Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:770-779. [PMID: 36541899 DOI: 10.1021/acs.jafc.2c07594] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Anti-proliferative peptides have recently attracted attention for their excellent bioactivity and biocompatibility. In this paper, five novel anti-proliferative peptides were identified from the hydrolysate of hybrid sturgeon spinal cord (HSSC). In addition, the structure-activity relationship of the novel anti-proliferative peptides was explored. In vitro experiments indicated that the peptide "VDSVLDVVRK" presented the highest inhibition of HeLa cell growth in all samples (IC50 = 2.5 μM). VDSVLDVVRK showed a random coil secondary structure and nanomicelles in the tumor microenvironment. Transmission electron microscopy results confirmed that nanomicelles disassemble as the concentration of VDSVLDVVRK decreases. Furthermore, VDSVLDVVRK could induce HeLa cell apoptosis by increasing the expression of Cyt-c (98.65 ± 1.85%, p < 0.01) and caspase-9 (39.85 ± 1.81%, p < 0.01). In this study, the anti-proliferative mechanism of the HSSC peptide was discussed, which provided a theoretical basis for the research and development of anti-proliferative functional food.
Collapse
Affiliation(s)
- Guixin Han
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003 Shandong, P.R. China
| | - Jinlin Wang
- Quzhou Sturgeon Aquatic Food Science and Technology Development Co., Ltd., Quzhou 324002 Zhejiang, P.R. China
| | - Yujin Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003 Shandong, P.R. China
| | - Zefan Chen
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003 Shandong, P.R. China
| | - Xinxing Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003 Shandong, P.R. China
| | - Tianhong Liu
- Marine Science Research Institute of Shandong Province, Qingdao 266104 Shandong, P.R. China
| | - Ying Wang
- Marine Science Research Institute of Shandong Province, Qingdao 266104 Shandong, P.R. China
| | - Fan Bai
- Quzhou Sturgeon Aquatic Food Science and Technology Development Co., Ltd., Quzhou 324002 Zhejiang, P.R. China
| | - Kang Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003 Shandong, P.R. China
| | - Yuanhui Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003 Shandong, P.R. China
| |
Collapse
|