1
|
Sagar B, Gupta S, Verma SK, Reddy YVM, Shukla S. Navigating cancer therapy: Harnessing the power of peptide-drug conjugates as precision delivery vehicles. Eur J Med Chem 2025; 283:117131. [PMID: 39647418 DOI: 10.1016/j.ejmech.2024.117131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Cancer treatment is a formidable challenge due to the adverse effects associated with non-selective therapies like chemotherapy and radiotherapy. This review article primarily centers on the application of Peptide-Drug Conjugates (PDCs) for delivering cancer treatment. PDCs represent a promising class of precision medicines, harnessing the unique attributes of peptides in conjunction with non-peptide components. The covalent linking of peptides and drugs through specialized connectors characterizes PDCs. These constructs play a pivotal role in delivering drugs directly to tumor sites with high precision. PDCs encompass three pivotal components: a targeting ligand, a cytotoxic ligand, and a carefully chosen linker. The selection of these elements is crucial to maximize the efficiency of PDCs. PDCs offer a multitude of advantages over conventional drug molecules, including enhanced specificity, reduced off-target effects, and an improved therapeutic profile. The peptide component within PDCs can be customized to specifically adhere to disease-specific receptors or biomarkers, facilitating targeted drug delivery and accumulation in afflicted cells or tissues. This targeted approach enables the controlled release of therapeutic payloads at the localized site, resulting in heightened effectiveness and minimized systemic toxicity. Diverse linker strategies are employed to ensure the stable connection between the peptide and non-peptide components, ensuring controlled drug release at the desired location of action. The peptides utilized in these treatments encompass cell-penetrating peptides, peptides designed to target tumor cells, and those aimed at the nucleus of cancer cells. While certain clinical trials have been conducted, and some PDCs are currently in use for cancer treatment, it's essential to acknowledge that PDCs have their limitations, such as low stability in plasma, fast elimination and limited oral bioavailability. Ongoing research endeavors seek to surmount these challenges and further establish PDCs as potent agents for cancer treatment. This review sheds light on recent advancements in the design, delivery, and applications of PDCs, while also highlighting the prevailing challenges and charting a path for future research directions.
Collapse
Affiliation(s)
- Bulbul Sagar
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarthak Gupta
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarvesh Kumar Verma
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, 302017, Rajasthan, India
| | | | - Shefali Shukla
- Sri Venkateswara College, University of Delhi, New Delhi, India.
| |
Collapse
|
2
|
Kohler A, Jülke EM, Stichel J, Beck-Sickinger AG. Comparison of Protocols to Test Peptide Stability in Blood Plasma and Cell Culture Supernatants. ACS Pharmacol Transl Sci 2024; 7:3618-3625. [PMID: 39539263 PMCID: PMC11555501 DOI: 10.1021/acsptsci.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Due to their high specificity, peptides are promising candidates in drug development, but fast degradation often limits their biological activity. Thus, a short half-life is one of the major challenges in the development of new peptide therapeutics. Moreover, the enzymatic cleavage of peptides can be a reason for misleading results in biological assays. Peptide stability assays typically consist of incubation, precipitation, and detection steps. However, the current methods differ greatly regarding these three steps, thus limiting the compatibility. Here, we systematically evaluate different parameters of peptide stability assays. First, we quantified and compared the analyte loss during the precipitation of plasma proteins. Especially, broadly used precipitation by strong acids was found to be unsuitable, while mixtures of organic solvents preserved more peptides for further analysis. Next, the stability of four fluorescently labeled model peptides was analyzed in blood plasma and two different cell culture supernatants. Strong variation in the degradation dynamics and patterns was found. Finally, we evaluated the role of fluorescent labeling on peptide stability and compared results to peptides with isotopic labels, underlining the individual advantages of both methods. Altogether, the data provide important parameters for analyzing and comparing the peptide stability.
Collapse
Affiliation(s)
- Anna Kohler
- Institute of Biochemistry,
Faculty of Life Sciences, Leipzig University, 04103 Leipzig, Germany
| | - Eva-Maria Jülke
- Institute of Biochemistry,
Faculty of Life Sciences, Leipzig University, 04103 Leipzig, Germany
| | - Jan Stichel
- Institute of Biochemistry,
Faculty of Life Sciences, Leipzig University, 04103 Leipzig, Germany
| | | |
Collapse
|
3
|
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
|
4
|
Egorov M, Goujon JY, Sicard M, Moal C, Pairel S, Le Bot R. Design, Synthesis, and Characterization of HBP-Vectorized Methotrexate Prodrug Molecule 1102-39: Evaluation of In Vitro Cytotoxicity Activity in Cell Culture Models, Preliminary In Vivo Safety and Efficacy Results in Rodents. ACS OMEGA 2024; 9:42433-42447. [PMID: 39431075 PMCID: PMC11483398 DOI: 10.1021/acsomega.4c06029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/22/2024]
Abstract
A novel bone-targeted prodrug, 1102-39, is discussed with the aim of enhancing the therapeutic effects of methotrexate (MTX) within bone tissues while minimizing systemic toxicity. Within the 1102-39 molecule, the central linker part forms a cleavable ester group, with MTX being also linked by a stable imine bond to the specially designed hydroxybisphosphonic (HBP) vector. Synthesized through a convergent approach starting from MTX, this prodrug advantageously modulates MTX's activity by selective esterification of its α-carboxyl group. In vitro tests revealed a 10-fold reduction in cytotoxicity compared to standard MTX, in alignment with prodrug behavior and correlated with gradual MTX release. In vivo in rodents, 1102-39 displayed preliminary encouraging antitumor effects on orthotopic osteosarcoma. Furthermore, various aspects of designing molecules for selective therapy in bone tissue based on bisphosphonate molecules as vectors for delivering active compounds to the bone are discussed. The 1102-39 molecule exhibits strong affinity for hydroxyapatite and a progressive release of MTX in aqueous environments, enhancing the safety and efficacy of bone-specific treatments and enabling sustained activity within bone and bone joints in the therapy of tumor and inflammation.
Collapse
Affiliation(s)
- Maxim Egorov
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| | | | - Marie Sicard
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| | | | - Samuel Pairel
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| | - Ronan Le Bot
- Atlanthera, 3 rue Aronnax, Saint-Herblain, 44821, France
| |
Collapse
|
5
|
Kistner A, Chichester JA, Wang L, Calcedo R, Greig JA, Cardwell LN, Wright MC, Couthouis J, Sethi S, McIntosh BE, McKeever K, Wadsworth S, Wilson JM, Kakkis E, Sullivan BA. Prednisolone and rapamycin reduce the plasma cell gene signature and may improve AAV gene therapy in cynomolgus macaques. Gene Ther 2024; 31:128-143. [PMID: 37833563 PMCID: PMC10940161 DOI: 10.1038/s41434-023-00423-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/07/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
Adeno-associated virus (AAV) vector gene therapy is a promising approach to treat rare genetic diseases; however, an ongoing challenge is how to best modulate host immunity to improve transduction efficiency and therapeutic outcomes. This report presents two studies characterizing multiple prophylactic immunosuppression regimens in male cynomolgus macaques receiving an AAVrh10 gene therapy vector expressing human coagulation factor VIII (hFVIII). In study 1, no immunosuppression was compared with prednisolone, rapamycin (or sirolimus), rapamycin and cyclosporin A in combination, and cyclosporin A and azathioprine in combination. Prednisolone alone demonstrated higher mean peripheral blood hFVIII expression; however, this was not sustained upon taper. Anti-capsid and anti-hFVIII antibody responses were robust, and vector genomes and transgene mRNA levels were similar to no immunosuppression at necropsy. Study 2 compared no immunosuppression with prednisolone alone or in combination with rapamycin or methotrexate. The prednisolone/rapamycin group demonstrated an increase in mean hFVIII expression and a mean delay in anti-capsid IgG development until after rapamycin taper. Additionally, a significant reduction in the plasma cell gene signature was observed with prednisolone/rapamycin, suggesting that rapamycin's tolerogenic effects may include plasma cell differentiation blockade. Immunosuppression with prednisolone and rapamycin in combination could improve therapeutic outcomes in AAV vector gene therapy.
Collapse
Affiliation(s)
| | - Jessica A Chichester
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lili Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roberto Calcedo
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Affinia Therapeutics, Waltham, MA, USA
| | - Jenny A Greig
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leah N Cardwell
- Ultragenyx Gene Therapy, Ultragenyx Pharmaceutical Inc., Cambridge, MA, USA
| | | | | | | | | | | | - Samuel Wadsworth
- Ultragenyx Gene Therapy, Ultragenyx Pharmaceutical Inc., Cambridge, MA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emil Kakkis
- Ultragenyx Pharmaceutical Inc., Novato, CA, USA
| | | |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Wu Y, Xia T, Ma X, Lei L, Du L, Xu X, Liu X, Shi Y, Li X, Lin D. Autocatalytic strategy for tunning drug release from peptide-drug supramolecular hydrogel. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
9
|
Fu C, Yu L, Miao Y, Liu X, Yu Z, Wei M. Peptide-drug conjugates (PDCs): a novel trend of research and development on targeted therapy, hype or hope? Acta Pharm Sin B 2023; 13:498-516. [PMID: 36873165 PMCID: PMC9978859 DOI: 10.1016/j.apsb.2022.07.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/16/2022] [Accepted: 07/11/2022] [Indexed: 11/01/2022] Open
Abstract
Peptide-drug conjugates (PDCs) are the next generation of targeted therapeutics drug after antibody-drug conjugates (ADCs), with the core benefits of enhanced cellular permeability and improved drug selectivity. Two drugs are now approved for market by US Food and Drug Administration (FDA), and in the last two years, the pharmaceutical companies have been developing PDCs as targeted therapeutic candidates for cancer, coronavirus disease 2019 (COVID-19), metabolic diseases, and so on. The therapeutic benefits of PDCs are significant, but poor stability, low bioactivity, long research and development time, and slow clinical development process as therapeutic agents of PDC, how can we design PDCs more effectively and what is the future direction of PDCs? This review summarises the components and functions of PDCs for therapeutic, from drug target screening and PDC design improvement strategies to clinical applications to improve the permeability, targeting, and stability of the various components of PDCs. This holds great promise for the future of PDCs, such as bicyclic peptide‒toxin coupling or supramolecular nanostructures for peptide-conjugated drugs. The mode of drug delivery is determined according to the PDC design and current clinical trials are summarised. The way is shown for future PDC development.
Collapse
Affiliation(s)
- Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China.,Liaoning Medical Diagnosis and Treatment Center, Shenyang 110000, China
| | - Xinli Liu
- Department of Digestive Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, China.,Liaoning Medical Diagnosis and Treatment Center, Shenyang 110000, China
| |
Collapse
|
10
|
Antiviral Peptide-Based Conjugates: State of the Art and Future Perspectives. Pharmaceutics 2023; 15:pharmaceutics15020357. [PMID: 36839679 PMCID: PMC9958607 DOI: 10.3390/pharmaceutics15020357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Infectious diseases caused by microbial pathogens (bacteria, virus, fungi, parasites) claim millions of deaths per year worldwide and have become a serious challenge to global human health in our century. Viral infections are particularly notable in this regard, not only because humankind is facing some of the deadliest viral pandemics in recent history, but also because the arsenal of drugs to combat the high levels of mutation, and hence the antigenic variability of (mostly RNA) viruses, is disturbingly scarce. Therefore, the search for new antivirals able to successfully fight infection with minimal or no adverse effects on the host is a pressing task. Traditionally, antiviral therapies have relied on relatively small-sized drugs acting as proteases, polymerases, integrase inhibitors, etc. In recent decades, novel approaches involving targeted delivery such as that achieved by peptide-drug conjugates (PDCs) have gained attention as alternative (pro)drugs for tackling viral diseases. Antiviral PDC therapeutics typically involve one or more small drug molecules conjugated to a cell-penetrating peptide (CPP) carrier either directly or through a linker. Such integration of two bioactive elements into a single molecular entity is primarily aimed at achieving improved bioavailability in conditions where conventional drugs are challenged, but may also turn up novel unexpected functionalities and applications. Advances in peptide medicinal chemistry have eased the way to antiviral PDCs, but challenges remain on the way to therapeutic success. In this paper, we review current antiviral CPP-drug conjugates (antiviral PDCs), with emphasis on the types of CPP and antiviral cargo. We integrate the conjugate and the chemical approaches most often applied to combine both entities. Additionally, we comment on various obstacles faced in the design of antiviral PDCs and on the future outlooks for this class of antiviral therapeutics.
Collapse
|
11
|
Novel Peptide Therapeutic Approaches for Cancer Treatment. Cells 2021; 10:cells10112908. [PMID: 34831131 PMCID: PMC8616177 DOI: 10.3390/cells10112908] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
Peptides are increasingly being developed for use as therapeutics to treat many ailments, including cancer. Therapeutic peptides have the advantages of target specificity and low toxicity. The anticancer effects of a peptide can be the direct result of the peptide binding its intended target, or the peptide may be conjugated to a chemotherapy drug or radionuclide and used to target the agent to cancer cells. Peptides can be targeted to proteins on the cell surface, where the peptide–protein interaction can initiate internalization of the complex, or the peptide can be designed to directly cross the cell membrane. Peptides can induce cell death by numerous mechanisms including membrane disruption and subsequent necrosis, apoptosis, tumor angiogenesis inhibition, immune regulation, disruption of cell signaling pathways, cell cycle regulation, DNA repair pathways, or cell death pathways. Although using peptides as therapeutics has many advantages, peptides have the disadvantage of being easily degraded by proteases once administered and, depending on the mode of administration, often have difficulty being adsorbed into the blood stream. In this review, we discuss strategies recently developed to overcome these obstacles of peptide delivery and bioavailability. In addition, we present many examples of peptides developed to fight cancer.
Collapse
|
12
|
Machulkin AE, Uspenskaya AA, Zyk NY, Nimenko EA, Ber AP, Petrov SA, Shafikov RR, Skvortsov DA, Smirnova GB, Borisova YA, Pokrovsky VS, Kolmogorov VS, Vaneev AN, Ivanenkov YA, Khudyakov AD, Kovalev SV, Erofeev AS, Gorelkin PV, Beloglazkina EK, Zyk NV, Khazanova ES, Majouga AG. PSMA-targeted small-molecule docetaxel conjugate: Synthesis and preclinical evaluation. Eur J Med Chem 2021; 227:113936. [PMID: 34717125 DOI: 10.1016/j.ejmech.2021.113936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
Prostate cancer is one of the most commonly diagnosed men's cancers and remains one of the leading causes of cancer death. The development of approaches to the treatment of this oncological disease is an ongoing process. In this work, we have carried out the selection of ligands for the creation of conjugates based on the drug docetaxel and synthesized a series of three docetaxel conjugates. In vitro cytotoxicity of these molecules was evaluated using the MTT assay. Based on the assay results, we selected the conjugate which showed cytotoxic potential close to unmodified docetaxel. At the same time, the molar solubility of the resulting compound increased up to 20 times in comparison with the drug itself. In vivo evaluation on 22Rv1 (PSMA+) xenograft model demonstrated a good potency of the synthesized conjugate to inhibit tumor growth: the inhibition turned out to be more than 80% at a dose of 30 mg/kg. Pharmacokinetic parameters of conjugate distribution were analyzed. Also, it was found that PSMA-targeted docetaxel conjugate is less toxic than docetaxel itself, the decrease of molar acute toxicity in comparison with free docetaxel was up to 20%. Obtained conjugate PSMA-DOC is a good candidate for further expanded preclinical trials because of high antitumor activity, fewer side toxic effects and better solubility.
Collapse
Affiliation(s)
- Aleksei E Machulkin
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation.
| | - Anastasia A Uspenskaya
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Nikolay Y Zyk
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Ekaterina A Nimenko
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Anton P Ber
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Stanislav A Petrov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Radik R Shafikov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, GSP-7, Ulitsa Miklukho-Maklaya, 16/10, Moscow, 117997, Russian Federation
| | - Dmitry A Skvortsov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Faculty of Biology and Biotechnologies, Higher School of Economics, Myasnitskaya 13, Moscow, 101000, Russia
| | - Galina B Smirnova
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia
| | - Yulia A Borisova
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia
| | - Vadim S Pokrovsky
- N.N. Blokhin Cancer Research Center, 24 Kashirskoye Sh., Moscow, 115478, Russia; RUDN University, Miklukho-Maklaya Str.6, Moscow, 117198, Russian Federation
| | - Vasilii S Kolmogorov
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Alexander N Vaneev
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Yan A Ivanenkov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region, 141700, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation; The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, 127055, Russia; Institute of Biochemistry and Genetics Ufa Science Centre Russian Academy of Sciences (IBG RAS), Oktyabrya Prospekt 71, Ufa, 450054, Russian Federation
| | - Alexander D Khudyakov
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Sergei V Kovalev
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Alexander S Erofeev
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Petr V Gorelkin
- National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation
| | - Elena K Beloglazkina
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Nikolay V Zyk
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation
| | - Elena S Khazanova
- LLC Izvarino-Pharma, V. Vnukovskoe, Vnukovskoe Sh., 5th Km., Building 1, Moscow, 108817, Russian Federation
| | - Alexander G Majouga
- Lomonosov Moscow State University, Chemistry Dept., Leninskie Gory, Building 1/3, GSP-1, Moscow, 119991, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy Pr, Moscow, 119049, Russian Federation; Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, Moscow, 125047, Russian Federation
| |
Collapse
|
13
|
Spiller S, Wippold T, Bellmann-Sickert K, Franz S, Saalbach A, Anderegg U, Beck-Sickinger AG. Protease-Triggered Release of Stabilized CXCL12 from Coated Scaffolds in an Ex Vivo Wound Model. Pharmaceutics 2021; 13:pharmaceutics13101597. [PMID: 34683890 PMCID: PMC8539926 DOI: 10.3390/pharmaceutics13101597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Biomaterials are designed to improve impaired healing of injured tissue. To accomplish better cell integration, we suggest to coat biomaterial surfaces with bio-functional proteins. Here, a mussel-derived surface-binding peptide is used and coupled to CXCL12 (stromal cell-derived factor 1α), a chemokine that activates CXCR4 and consequently recruits tissue-specific stem and progenitor cells. CXCL12 variants with either non-releasable or protease-mediated-release properties were designed and compared. Whereas CXCL12 was stabilized at the N-terminus for protease resistance, a C-terminal linker was designed that allowed for specific cleavage-mediated release by matrix metalloproteinase 9 and 2, since both enzymes are frequently found in wound fluid. These surface adhesive CXCL12 derivatives were produced by expressed protein ligation. Functionality of the modified chemokines was assessed by inositol phosphate accumulation and cell migration assays. Increased migration of keratinocytes and primary mesenchymal stem cells was demonstrated. Immobilization and release were studied for bioresorbable PCL-co-LC scaffolds, and accelerated wound closure was demonstrated in an ex vivo wound healing assay on porcine skin grafts. After 24 h, a significantly improved CXCL12-specific growth stimulation of the epithelial tips was already observed. The presented data display a successful application of protein-coated biomaterials for skin regeneration.
Collapse
Affiliation(s)
- Sabrina Spiller
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
| | - Tom Wippold
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
| | - Sandra Franz
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Anja Saalbach
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; (T.W.); (S.F.); (A.S.)
- Correspondence: (U.A.); (A.G.B.-S.); Tel.: +49-341-972-5881 (U.A.); +49-341-973-6900 (A.G.B.-S.); Fax: +49-341-972-5878 (U.A.); +49-341-973-6909 (A.G.B.-S.)
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany; (S.S.); (K.B.-S.)
- Correspondence: (U.A.); (A.G.B.-S.); Tel.: +49-341-972-5881 (U.A.); +49-341-973-6900 (A.G.B.-S.); Fax: +49-341-972-5878 (U.A.); +49-341-973-6909 (A.G.B.-S.)
| |
Collapse
|
14
|
Fischer TF, Czerniak AS, Weiß T, Zellmann T, Zielke L, Els-Heindl S, Beck-Sickinger AG. Cyclic Derivatives of the Chemerin C-Terminus as Metabolically Stable Agonists at the Chemokine-like Receptor 1 for Cancer Treatment. Cancers (Basel) 2021; 13:cancers13153788. [PMID: 34359687 PMCID: PMC8345219 DOI: 10.3390/cancers13153788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Chemerin is a small chemotactic protein and a modulator of the innate immune system. Its activity is mainly mediated by the chemokine-like receptor 1 (CMKLR1), a receptor expressed by natural killer cells, dendritic cells, and macrophages. Downregulation of chemerin is part of the immune evasion strategy exploited by several cancer types, including melanoma, breast cancer, and hepatocellular carcinoma. Administration of chemerin can potentially counteract these effects, but synthetically accessible, metabolically stable analogs are required. Other tumors display overexpression of CMKLR1, offering a potential entry point for targeted delivery of chemotherapeutics. Here, we present cyclic derivatives of the chemerin C-terminus (chemerin-9), the minimal activation sequence of chemerin. Chemerin-9 derivatives that were cyclized through positions four and nine retained activity while displaying full stability in blood plasma for more than 24 h. Therefore, these peptides could be used as a drug shuttle system to target cancer cells as demonstrated here by methotrexate conjugates.
Collapse
|
15
|
Ziffert I, Kaiser A, Hoppenz P, Mörl K, Beck‐Sickinger AG. Shuttling of Peptide-Drug Conjugates by G Protein-Coupled Receptors Is Significantly Improved by Pulsed Application. ChemMedChem 2021; 16:164-178. [PMID: 32700391 PMCID: PMC7818256 DOI: 10.1002/cmdc.202000490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Indexed: 01/08/2023]
Abstract
G protein-coupled receptors (GPCRs) can be used to shuttle peptide-drug conjugates into cells. But, for efficient therapy, a high concentration of cargo needs to be delivered. To explore this, we studied the pharmacologically interesting neuropeptide Y1 receptor (Y1 R) in one recombinant and three oncogenic cell systems that endogenously express the receptor. We demonstrate that recycled receptors behave identically to newly synthesized receptors with respect to ligand binding and internalization pathways. Depending on the cell system, biosynthesis, recycling efficiency, and peptide uptake differ partially, but shuttling was efficient in all systems. However, by comparing continuous application of the ligand for four hours to four cycles of internalization and recycling in between, a significantly higher amount of peptide uptake was achieved in the pulsed application (150-250 % to 300-400 %). Accordingly, in this well-suited drug shuttle system pulsed application is superior under all investigated conditions and should be considered for innovative, targeted drug delivery in general.
Collapse
Affiliation(s)
- Isabelle Ziffert
- Institute of BiochemistryFaculty of Life SciencesUniversity of LeipzigBrüderstraße 3404103LeipzigGermany
| | - Anette Kaiser
- Institute of BiochemistryFaculty of Life SciencesUniversity of LeipzigBrüderstraße 3404103LeipzigGermany
| | - Paul Hoppenz
- Institute of BiochemistryFaculty of Life SciencesUniversity of LeipzigBrüderstraße 3404103LeipzigGermany
| | - Karin Mörl
- Institute of BiochemistryFaculty of Life SciencesUniversity of LeipzigBrüderstraße 3404103LeipzigGermany
| | - Annette G. Beck‐Sickinger
- Institute of BiochemistryFaculty of Life SciencesUniversity of LeipzigBrüderstraße 3404103LeipzigGermany
| |
Collapse
|
16
|
Petrov RA, Mefedova SR, Yamansarov EY, Maklakova SY, Grishin DA, Lopatukhina EV, Burenina OY, Lopukhov AV, Kovalev SV, Timchenko YV, Ondar EE, Ivanenkov YA, Evteev SA, Vaneev AN, Timoshenko RV, Klyachko NL, Erofeev AS, Gorelkin PV, Beloglazkina EK, Majouga AG. New Small-Molecule Glycoconjugates of Docetaxel and GalNAc for Targeted Delivery to Hepatocellular Carcinoma. Mol Pharm 2020; 18:461-468. [PMID: 33264010 DOI: 10.1021/acs.molpharmaceut.0c00980] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this work, we have developed covalent and low molecular weight docetaxel delivery systems based on conjugation with N-acetyl-d-galactosamine and studied their properties related to hepatocellular carcinoma cells. The resulting glycoconjugates have an excellent affinity to the asialoglycoprotein receptor (ASGPR) in the nanomolar range of concentrations and a high cytotoxicity level comparable to docetaxel. Likewise, we observed the 21-75-fold increase in water solubility in comparison with parent docetaxel and prodrug lability to intracellular conditions with half-life values from 25.5 to 42 h. We also found that the trivalent conjugate possessed selective toxicity against hepatoma cells vs control cell lines (20-35 times). The absence of such selectivity in the case of monovalent conjugates indicates the effect of ligand valency. Specific ASGPR-mediated cellular uptake of conjugates was proved in vitro using fluorescent-labeled analogues. In addition, we showed an enhanced generation of reactive oxygen species in the HepG2 cells, which could be inhibited by the natural ligand of ASGPR. Overall, the obtained results highlight the potential of ASGPR-directed cytostatic taxane drugs for selective therapy of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Rostislav A Petrov
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,Institute of Biochemistry and Genetics Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Oktyabrya Prospekt 71, Ufa 450054, Russian Federation
| | - Sofiia R Mefedova
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Emil Yu Yamansarov
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Svetlana Yu Maklakova
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Dmitrii A Grishin
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Elena V Lopatukhina
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Olga Y Burenina
- Skolkovo Institute of Science and Technology, 3 Nobel str., Skolkovo 143026, Russian Federation
| | - Anton V Lopukhov
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Sergey V Kovalev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Yury V Timchenko
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Evgenia E Ondar
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Yan A Ivanenkov
- Institute of Biochemistry and Genetics Russian Academy of Science (IBG RAS) of the Ufa Federal Research Centre, Oktyabrya Prospekt 71, Ufa 450054, Russian Federation.,Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow 141700, Russian Federation
| | - Sergei A Evteev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Alexander N Vaneev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Roman V Timoshenko
- National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Natalia L Klyachko
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,Skolkovo Institute of Science and Technology, 3 Nobel str., Skolkovo 143026, Russian Federation
| | - Alexander S Erofeev
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Petr V Gorelkin
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation
| | - Elena K Beloglazkina
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Alexander G Majouga
- Chemistry Department, Lomonosov Moscow State University, Leninskie gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation.,National University of Science and Technology MISIS, 9 Leninskiy pr., Moscow 119049, Russian Federation.,Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, Moscow 125047, Russian Federation
| |
Collapse
|
17
|
Nam SH, Jang J, Cheon DH, Chong SE, Ahn JH, Hyun S, Yu J, Lee Y. pH-Activatable cell penetrating peptide dimers for potent delivery of anticancer drug to triple-negative breast cancer. J Control Release 2020; 330:898-906. [PMID: 33152392 DOI: 10.1016/j.jconrel.2020.10.063] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023]
Abstract
We developed a pH-activatable cell-penetrating peptide dimer LH2 with histidine residues, which can penetrate cells, specifically in weak acidic conditions, even at few tens of nanomolar concentrations. LH2 effectively delivered paclitaxel into triple-negative breast cancer cells, MDA-MB-231, via formation of non-covalent complexes (PTX-LH2(M)) or covalent conjugates (PTX-LH2(C)). Moreover, LH2 showed prolonged circulation in the body and enhanced accumulation in tumors. Both PTX-LH2(M) and PTX-LH2(C) showed strong antitumor effects in a triple-negative breast cancer grafted mouse model at an extremely low dosage.
Collapse
Affiliation(s)
- So Hee Nam
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Joomyung Jang
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dae Hee Cheon
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seung-Eun Chong
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Joon Hyung Ahn
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Soonsil Hyun
- Department of Chemistry and Education, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jaehoon Yu
- Department of Chemistry and Education, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Yan Lee
- Department of Chemistry, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
18
|
Hoppenz P, Els-Heindl S, Beck-Sickinger AG. Peptide-Drug Conjugates and Their Targets in Advanced Cancer Therapies. Front Chem 2020; 8:571. [PMID: 32733853 PMCID: PMC7359416 DOI: 10.3389/fchem.2020.00571] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer became recently the leading cause of death in industrialized countries. Even though standard treatments achieve significant effects in growth inhibition and tumor elimination, they cause severe side effects as most of the applied drugs exhibit only minor selectivity for the malignant tissue. Hence, specific addressing of tumor cells without affecting healthy tissue is currently a major desire in cancer therapy. Cell surface receptors, which bind peptides are frequently overexpressed on cancer cells and can therefore be considered as promising targets for selective tumor therapy. In this review, the benefits of peptides as tumor homing agents are presented and an overview of the most commonly addressed peptide receptors is given. A special focus was set on the bombesin receptor family and the neuropeptide Y receptor family. In the second part, the specific requirements of peptide-drug conjugates (PDC) and intelligent linker structures as an essential component of PDC are outlined. Furthermore, different drug cargos are presented including classical and recent toxic agents as well as radionuclides for diagnostic and therapeutic approaches. In the last part, boron neutron capture therapy as advanced targeted cancer therapy is introduced and past and recent developments are reviewed.
Collapse
Affiliation(s)
- Paul Hoppenz
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Sylvia Els-Heindl
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
19
|
Worm DJ, Els‐Heindl S, Beck‐Sickinger AG. Targeting of peptide‐binding receptors on cancer cells with peptide‐drug conjugates. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24171] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dennis J. Worm
- Faculty of Life Sciences, Institute of BiochemistryLeipzig University Leipzig Germany
| | - Sylvia Els‐Heindl
- Faculty of Life Sciences, Institute of BiochemistryLeipzig University Leipzig Germany
| | | |
Collapse
|
20
|
Conibear AC, Schmid A, Kamalov M, Becker CFW, Bello C. Recent Advances in Peptide-Based Approaches for Cancer Treatment. Curr Med Chem 2020; 27:1174-1205. [PMID: 29173146 DOI: 10.2174/0929867325666171123204851] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Peptide-based pharmaceuticals have recently experienced a renaissance due to their ability to fill the gap between the two main classes of available drugs, small molecules and biologics. Peptides combine the high potency and selectivity typical of large proteins with some of the characteristic advantages of small molecules such as synthetic accessibility, stability and the potential of oral bioavailability. METHODS In the present manuscript we review the recent literature on selected peptide-based approaches for cancer treatment, emphasizing recent advances, advantages and challenges of each strategy. RESULTS One of the applications in which peptide-based approaches have grown rapidly is cancer therapy, with a focus on new and established targets. We describe, with selected examples, some of the novel peptide-based methods for cancer treatment that have been developed in the last few years, ranging from naturally-occurring and modified peptides to peptidedrug conjugates, peptide nanomaterials and peptide-based vaccines. CONCLUSION This review brings out the emerging role of peptide-based strategies in oncology research, critically analyzing the advantages and limitations of these approaches and the potential for their development as effective anti-cancer therapies.
Collapse
Affiliation(s)
- Anne C Conibear
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Wahringer Straße 38, 1090 Vienna, Austria
| | - Alanca Schmid
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Wahringer Straße 38, 1090 Vienna, Austria
| | - Meder Kamalov
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Wahringer Straße 38, 1090 Vienna, Austria
| | - Christian F W Becker
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Wahringer Straße 38, 1090 Vienna, Austria
| | - Claudia Bello
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Wahringer Straße 38, 1090 Vienna, Austria.,Department of Chemistry "Ugo Schiff", University of Florence, Laboratory of Peptide and Protein Chemistry and Biolology-PeptLab, Via della Lastruccia 13, 50019 Sesto, Fiorentino, Italy
| |
Collapse
|
21
|
Chastel A, Worm DJ, Alves ID, Vimont D, Petrel M, Fernandez S, Garrigue P, Fernandez P, Hindié E, Beck-Sickinger AG, Morgat C. Design, synthesis, and biological evaluation of a multifunctional neuropeptide-Y conjugate for selective nuclear delivery of radiolanthanides. EJNMMI Res 2020; 10:16. [PMID: 32124111 PMCID: PMC7052099 DOI: 10.1186/s13550-020-0612-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Background Targeting G protein-coupled receptors on the surface of cancer cells with peptide ligands is a promising concept for the selective tumor delivery of therapeutically active cargos, including radiometals for targeted radionuclide therapy (TRT). Recently, the radiolanthanide terbium-161 (161Tb) gained significant interest for TRT application, since it decays with medium-energy β-radiation but also emits a significant amount of conversion and Auger electrons with short tissue penetration range. The therapeutic efficiency of radiometals emitting Auger electrons, like 161Tb, can therefore be highly boosted by an additional subcellular delivery into the nucleus, in order to facilitate maximum dose deposition to the DNA. In this study, we describe the design of a multifunctional, radiolabeled neuropeptide-Y (NPY) conjugate, to address radiolanthanides to the nucleus of cells naturally overexpressing the human Y1 receptor (hY1R). By using solid-phase peptide synthesis, the hY1R-preferring [F7,P34]-NPY was modified with a fatty acid, a cathepsin B-cleavable linker, followed by a nuclear localization sequence (NLS), and a DOTA chelator (compound pb12). In this proof-of-concept study, labeling was performed with either native terbium-159 (natTb), as surrogate for 161Tb, or with indium-111 (111In). Results [natTb]Tb-pb12 showed a preserved high binding affinity to endogenous hY1R on MCF-7 cells and was able to induce receptor activation and internalization similar to the hY1R-preferring [F7,P34]-NPY. Specific internalization of the 111In-labeled conjugate into MCF-7 cells was observed, and importantly, time-dependent nuclear uptake of 111In was demonstrated. Study of metabolic stability showed that the peptide is insufficiently stable in human plasma. This was confirmed by injection of [111In]In-pb12 in nude mice bearing MCF-7 xenograft which showed specific uptake only at very early time point. Conclusion The multifunctional NPY conjugate with a releasable DOTA-NLS unit represents a promising concept for enhanced TRT with Auger electron-emitting radiolanthanides. Our research is now focusing on improving the reported concept with respect to the poor plasmatic stability of this promising radiopeptide.
Collapse
Affiliation(s)
- Adrien Chastel
- Department of Nuclear Medicine, University Hospital of Bordeaux, F-33076, Bordeaux, France.,University of Bordeaux, INCIA UMR 5287, F-33400, Talence, France.,CNRS, INCIA UMR 5287, F-33400, Talence, France
| | - Dennis J Worm
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103, Leipzig, Germany
| | - Isabel D Alves
- Institute of Chemistry & Biology of Membranes & Nano-objects (CBMN), CNRS UMR 5248, University of Bordeaux, F-33600, Pessac, France
| | - Delphine Vimont
- University of Bordeaux, INCIA UMR 5287, F-33400, Talence, France.,CNRS, INCIA UMR 5287, F-33400, Talence, France
| | - Melina Petrel
- University of Bordeaux, Bordeaux Imaging Center, F-33000, Bordeaux, France
| | - Samantha Fernandez
- Aix-Marseille University, INSERM, Institut National de la Recherche Agronomique, Centre de Recherche en Cardiovasculaire et Nutrition, 13385, Marseille, France.,Aix-Marseille University, Centre Européen de Recherche en Imagerie Médicale, 13005, Marseille, France
| | - Philippe Garrigue
- Aix-Marseille University, INSERM, Institut National de la Recherche Agronomique, Centre de Recherche en Cardiovasculaire et Nutrition, 13385, Marseille, France.,Aix-Marseille University, Centre Européen de Recherche en Imagerie Médicale, 13005, Marseille, France
| | - Philippe Fernandez
- Department of Nuclear Medicine, University Hospital of Bordeaux, F-33076, Bordeaux, France.,University of Bordeaux, INCIA UMR 5287, F-33400, Talence, France.,CNRS, INCIA UMR 5287, F-33400, Talence, France
| | - Elif Hindié
- Department of Nuclear Medicine, University Hospital of Bordeaux, F-33076, Bordeaux, France.,University of Bordeaux, INCIA UMR 5287, F-33400, Talence, France.,CNRS, INCIA UMR 5287, F-33400, Talence, France
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103, Leipzig, Germany
| | - Clément Morgat
- Department of Nuclear Medicine, University Hospital of Bordeaux, F-33076, Bordeaux, France. .,University of Bordeaux, INCIA UMR 5287, F-33400, Talence, France. .,CNRS, INCIA UMR 5287, F-33400, Talence, France.
| |
Collapse
|
22
|
Liu ZY, Tang ML, Ning JF, Hao YP, Zhou L, Sun X. Novel octapeptide-DTX prodrugs targeting MMP-7 as effective agents for the treatment of colorectal cancer with lower systemic toxicity. Eur J Med Chem 2020; 193:112194. [PMID: 32203786 DOI: 10.1016/j.ejmech.2020.112194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/27/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth leading cause of cancer death around the world. The current treatments of CRC exhibited high occurrence rate of side effects. Docetaxel (DTX), an important drug widely used in cancer chemotherapy, showed serious toxicity in CRC. Reducing toxicity of DTX could be a feasible and promising way to achieve the new indication of DTX for CRC. In this study, a series of MMP-7 activated octapeptide-DTX/4FDT prodrugs (6a-10a and 6b-10b) were designed and synthesized based on the features of MMP-7 which is highly expressed in CRC and could specially recognize octapeptides with specific sequences. Among them, 9a and 9b, both possessing an octapeptide Gly-Pro-Gln-Gly-Ile-Ala-Met-Gln moiety, were the most potent prodrugs. Compounds 9a and 9b were also tested their release rate in HCT116 cell culture fluids and tumor homogenate along with in vivo anti-CRC activity and systemic toxicity. Since 9a showed better anti-CRC activity and lower systemic toxicity than 9b in CRC tumor bearing mice, it was further evaluated for its acute toxicity, pharmacokinetics and tissue distribution in comparison with its parent drug DTX. These results revealed that 9a possessed good systemic stability, rapid release rate in CRC and reduced systemic toxicity, while retaining similar anti-CRC activity to its parent drug DTX. Thus, 9a, an MMP-7 polypeptide prodrug of DTX, has been identified as a promising candidate for the treatment of CRC.
Collapse
Affiliation(s)
- Zheng-Yu Liu
- Department of Natural Medicine, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Mei-Lin Tang
- Department of Natural Medicine, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China; State Key Laboratory of Molecular Engineering and Institutes of Biomedical Sciences, Fudan University, 220 Handan Road, Shanghai, 200433, China
| | - Jin-Feng Ning
- Department of Natural Medicine, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Yun-Peng Hao
- Department of Natural Medicine, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Lu Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Xun Sun
- Department of Natural Medicine, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China; The Institutes of Integrative Medicine of Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
23
|
Varini K, Lécorché P, Sonnette R, Gassiot F, Broc B, Godard M, David M, Faucon A, Abouzid K, Ferracci G, Temsamani J, Khrestchatisky M, Jacquot G. Target engagement and intracellular delivery of mono- and bivalent LDL receptor-binding peptide-cargo conjugates: Implications for the rational design of new targeted drug therapies. J Control Release 2019; 314:141-161. [PMID: 31644939 DOI: 10.1016/j.jconrel.2019.10.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
Targeted delivery to specific tissues and subcellular compartments is of paramount importance to optimize therapeutic or diagnostic interventions while minimizing side-effects. Using recently identified LDL receptor (LDLR) -targeting small synthetic peptide-vectors conjugated to model cargos of different nature and size, we investigated in LDLR-expressing cells the impact of vector-cargo molecular engineering and coupling valency, as well as the cellular exposure duration on their target engagement and intracellular trafficking and delivery profiles. All vector-cargo conjugates evaluated were found to be delivered to late compartments together with the natural ligand LDL, although to varying extents and with different kinetics. Partial recycling together with the LDLR was also consistently observed. Under continuous cellular exposure, the extent of intracellular vector-cargo delivery primarily relies on their endosomal unloading potential. In this condition, the highest intracellular delivery potential was observed with a monovalent conjugate displaying a rather high LDLR dissociation rate. On the contrary, under transient cellular exposure followed by chase, low dissociation-rate bivalent conjugates revealed a higher intracellular delivery potential than the monovalent conjugate. This was shown to rely on their ability to undergo multiple endocytosis-recycling rounds, with limited release in the ligand-free medium. The absence of reciprocal competition with the natural ligand LDL on their respective intracellular trafficking was also demonstrated, which is essential in terms of potential safety liabilities. These results demonstrate that not only molecular engineering of new therapeutic conjugates of interest, but also the cellular exposure mode used during in vitro evaluations are critical to anticipate and optimize their delivery potential.
Collapse
Affiliation(s)
- K Varini
- VECT-HORUS SAS, Marseille, France; Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | | | | | | - B Broc
- VECT-HORUS SAS, Marseille, France
| | - M Godard
- VECT-HORUS SAS, Marseille, France
| | - M David
- VECT-HORUS SAS, Marseille, France
| | - A Faucon
- VECT-HORUS SAS, Marseille, France
| | | | - G Ferracci
- Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | | - M Khrestchatisky
- Aix-Marseille Univ., CNRS, INP, Inst. Neurophysiopathol., Marseille, France
| | | |
Collapse
|
24
|
Wittrisch S, Klöting N, Mörl K, Chakaroun R, Blüher M, Beck-Sickinger AG. NPY 1R-targeted peptide-mediated delivery of a dual PPARα/γ agonist to adipocytes enhances adipogenesis and prevents diabetes progression. Mol Metab 2019; 31:163-180. [PMID: 31918918 PMCID: PMC6931124 DOI: 10.1016/j.molmet.2019.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/31/2019] [Accepted: 11/10/2019] [Indexed: 12/12/2022] Open
Abstract
Objective PPARα/γ dual agonists have been in clinical development for the treatment of metabolic diseases including type 2 diabetes and dyslipidemia. However, severe adverse side effects led to complications in clinical trials. As most of the beneficial effects rely on the compound activity in adipocytes, the selective targeting of this cell type is a cutting-edge strategy to develop safe anti-diabetic drugs. The goal of this study was to strengthen the adipocyte-specific uptake of the PPARα/γ agonist tesaglitazar via NPY1R-mediated internalization. Methods NPY1R-preferring peptide tesaglitazar-[F7, P34]-NPY (tesa-NPY) was synthesized by a combination of automated SPPS and manual couplings. Following molecular and functional analyses for proof of concept, cell culture experiments were conducted to monitor the effects on adipogenesis. Mice treated with peptide drug conjugates or vehicle either by gavage or intraperitoneal injection were characterized phenotypically and metabolically. Histological analysis and transcriptional profiling of the adipose tissue were performed. Results In vitro studies revealed that the tesaglitazar-[F7, P34]-NPY conjugate selectively activates PPARγ in NPY1R-expressing cells and enhances adipocyte differentiation and adiponectin expression in adipocyte precursor cells. In vivo studies using db/db mice demonstrated that the anti-diabetic activity of the peptide conjugate is as efficient as that of systemically administered tesaglitazar. Additionally, tesa-NPY induces adipocyte differentiation in vivo. Conclusions The use of the tesaglitazar-[F7, P34]-NPY conjugate is a promising strategy to apply the beneficial PPARα/γ effects in adipocytes while potentially omitting adverse effects in other tissues. Tesaglitazar-NPY targets adipocytes via NPY1R receptor-mediated internalization. Peptide-drug conjugate is specifically delivered to NPY1R-expressing cells. Release of tesaglitazar in adipocytes activates PPARγ. Drug delivery enhances adipocyte differentiation and adiponectin expression. Peptide conjugate exhibits antidiabetic activity in vivo.
Collapse
Affiliation(s)
- Stefanie Wittrisch
- Universität Leipzig, Institute of Biochemistry, Brüderstraße 34, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity, and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Ph.-Rosenthal-Str. 27, 04103 Leipzig, Germany.
| | - Karin Mörl
- Universität Leipzig, Institute of Biochemistry, Brüderstraße 34, 04103 Leipzig, Germany
| | - Rima Chakaroun
- Helmholtz Institute for Metabolic, Obesity, and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Ph.-Rosenthal-Str. 27, 04103 Leipzig, Germany; Department of Medicine, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity, and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Ph.-Rosenthal-Str. 27, 04103 Leipzig, Germany; Department of Medicine, University of Leipzig, Liebigstraße 20, 04103 Leipzig, Germany.
| | | |
Collapse
|
25
|
Hofmann S, Bellmann-Sickert K, Beck-Sickinger AG. Chemical modification of neuropeptide Y for human Y1 receptor targeting in health and disease. Biol Chem 2019; 400:299-311. [PMID: 30653463 DOI: 10.1515/hsz-2018-0364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022]
Abstract
As a very abundant neuropeptide in the brain and widely distributed peptide hormone in the periphery, neuropeptide Y (NPY) appears to be a multisignaling key peptide. Together with peptide YY, pancreatic polypeptide and the four human G protein-coupled receptor subtypes hY1R, hY2R, hY4R and hY5R it forms the NPY/hYR multiligand/multireceptor system, which is involved in essential physiological processes as well as in human diseases. In particular, NPY-induced hY1R signaling plays a central role in the regulation of food intake and stress response as well as in obesity, mood disorders and cancer. Thus, several hY1R-preferring NPY analogs have been developed as versatile tools to unravel the complex NPY/hY1R signaling in health and disease. Further, these peptides provide basic lead structures for the development of innovative drugs. Here, the current research is summarized focusing on the development of differently sized hY1R-preferring NPY analogs as well as their advances with respect to hY1R profiling, potential therapeutic applications and targeted cancer imaging and therapy. Finally, major limitations and innovative strategies for next generation hY1R-preferring NPY analogs are addressed.
Collapse
Affiliation(s)
- Sven Hofmann
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| |
Collapse
|
26
|
Worm DJ, Hoppenz P, Els-Heindl S, Kellert M, Kuhnert R, Saretz S, Köbberling J, Riedl B, Hey-Hawkins E, Beck-Sickinger AG. Selective Neuropeptide Y Conjugates with Maximized Carborane Loading as Promising Boron Delivery Agents for Boron Neutron Capture Therapy. J Med Chem 2019; 63:2358-2371. [PMID: 31589041 DOI: 10.1021/acs.jmedchem.9b01136] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors like the human Y1 receptor (hY1R) are promising targets in cancer therapy due to their high overexpression on cancer cells and their ability to internalize together with the bound ligand. This mechanism was exploited to shuttle boron atoms into cancer cells for the application of boron neutron capture therapy (BNCT), a noninvasive approach to eliminate cancer cells. A maximized number of carboranes was introduced to the hY1R-preferring ligand [F7,P34]-NPY by solid phase peptide synthesis. Branched conjugates loaded with up to 80 boron atoms per peptide molecule exhibited a maintained receptor activation profile, and the selective uptake into hY1R-expressing cells was demonstrated by internalization studies. In order to ensure appropriate solubility in aqueous solution, we proved the need for eight hydroxyl groups per carborane. Thus, we suggest the utilization of bis-deoxygalactosyl-carborane building blocks in solid phase peptide synthesis to produce selective boron delivery agents for BNCT.
Collapse
Affiliation(s)
- Dennis J Worm
- Institute of Biochemistry, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Paul Hoppenz
- Institute of Biochemistry, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Martin Kellert
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | - Robert Kuhnert
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | - Stefan Saretz
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | | | - Bernd Riedl
- Bayer AG, Aprather Weg 18A, 42113 Wuppertal, Germany
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | | |
Collapse
|
27
|
Clauder F, Czerniak AS, Friebe S, Mayr SG, Scheinert D, Beck-Sickinger AG. Endothelialization of Titanium Surfaces by Bioinspired Cell Adhesion Peptide Coatings. Bioconjug Chem 2019; 30:2664-2674. [DOI: 10.1021/acs.bioconjchem.9b00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Anne Sophie Czerniak
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sabrina Friebe
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Stefan G. Mayr
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Dierk Scheinert
- Department of Angiology, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| |
Collapse
|
28
|
Peptide Conjugates with Small Molecules Designed to Enhance Efficacy and Safety. Molecules 2019; 24:molecules24101855. [PMID: 31091786 PMCID: PMC6572008 DOI: 10.3390/molecules24101855] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 12/17/2022] Open
Abstract
Peptides constitute molecular diversity with unique molecular mechanisms of action that are proven indispensable in the management of many human diseases, but of only a mere fraction relative to more traditional small molecule-based medicines. The integration of these two therapeutic modalities offers the potential to enhance and broaden pharmacology while minimizing dose-dependent toxicology. This review summarizes numerous advances in drug design, synthesis and development that provide direction for next-generation research endeavors in this field. Medicinal studies in this area have largely focused upon the application of peptides to selectively enhance small molecule cytotoxicity to more effectively treat multiple oncologic diseases. To a lesser and steadily emerging extent peptides are being therapeutically employed to complement and diversify the pharmacology of small molecule drugs in diseases other than just cancer. No matter the disease, the purpose of the molecular integration remains constant and it is to achieve superior therapeutic outcomes with diminished adverse effects. We review linker technology and conjugation chemistries that have enabled integrated and targeted pharmacology with controlled release. Finally, we offer our perspective on opportunities and obstacles in the field.
Collapse
|
29
|
Petrov RA, Maklakova SY, Ivanenkov YA, Petrov SA, Sergeeva OV, Yamansarov EY, Saltykova IV, Kireev II, Alieva IB, Deyneka EV, Sofronova AA, Aladinskaia AV, Trofimenko AV, Yamidanov RS, Kovalev SV, Kotelianski VE, Zatsepin TS, Beloglazkina EK, Majouga AG. Synthesis and biological evaluation of novel mono- and bivalent ASGP-R-targeted drug-conjugates. Bioorg Med Chem Lett 2017; 28:382-387. [PMID: 29269214 DOI: 10.1016/j.bmcl.2017.12.032] [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] [Received: 11/23/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022]
Abstract
Asialoglycoprotein receptor (ASGP-R) is a promising biological target for drug delivery into hepatoma cells. Nevertheless, there are only few examples of small-molecule conjugates of ASGP-R selective ligand equipped by a therapeutic agent for the treatment of hepatocellular carcinoma (HCC). In the present work, we describe a convenient and versatile synthetic approach to novel mono- and multivalent drug-conjugates containing N-acetyl-2-deoxy-2-aminogalactopyranose and anticancer drug - paclitaxel (PTX). Several molecules have demonstrated high affinity towards ASGP-R and good stability under physiological conditions, significant in vitro anticancer activity comparable to PTX, as well as good internalization via ASGP-R-mediated endocytosis. Therefore, the conjugates with the highest potency can be regarded as a promising therapeutic option against HCC.
Collapse
Affiliation(s)
- Rostislav A Petrov
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Svetlana Yu Maklakova
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Yan A Ivanenkov
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation; Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region 141700, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy pr, Moscow 119049, Russian Federation; Institute of Biochemistry and Genetics Ufa Science Centre Russian Academy of Sciences (IBG RAS), Prosp. Oktybrya 71, Ufa, Bashkortostan 450054, Russian Federation.
| | - Stanislav A Petrov
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Olga V Sergeeva
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation; Skolkovo Institute of Science and Technology, 100 Novaya St., 143025 Skolkovo, Russian Federation
| | - Emil Yu Yamansarov
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Irina V Saltykova
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Igor I Kireev
- Lomonosov Moscow State University, A.N. Belozersky Institute of Physico-Chemical Biology, Leninskye Gory, House 1, Building 40, Moscow 119992, Russian Federation
| | - Irina B Alieva
- Lomonosov Moscow State University, A.N. Belozersky Institute of Physico-Chemical Biology, Leninskye Gory, House 1, Building 40, Moscow 119992, Russian Federation
| | - Ekaterina V Deyneka
- Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region 141700, Russian Federation
| | - Alina A Sofronova
- Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, Russia
| | - Anastasiia V Aladinskaia
- Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region 141700, Russian Federation
| | - Alexandre V Trofimenko
- Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudny City, Moscow Region 141700, Russian Federation
| | - Renat S Yamidanov
- Institute of Biochemistry and Genetics Ufa Science Centre Russian Academy of Sciences (IBG RAS), Prosp. Oktybrya 71, Ufa, Bashkortostan 450054, Russian Federation
| | - Sergey V Kovalev
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Victor E Kotelianski
- Skolkovo Institute of Science and Technology, 100 Novaya St., 143025 Skolkovo, Russian Federation
| | - Timofey S Zatsepin
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation; Skolkovo Institute of Science and Technology, 100 Novaya St., 143025 Skolkovo, Russian Federation
| | - Elena K Beloglazkina
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation
| | - Alexander G Majouga
- Lomonosov Moscow State University, Chemistry Dept, Leninskie Gory, Building 1/3, GSP-1, Moscow 119991, Russian Federation; National University of Science and Technology MISiS, 9 Leninskiy pr, Moscow 119049, Russian Federation; Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, Moscow 125047, Russian Federation
| |
Collapse
|
30
|
Gladytz A, John T, Gladytz T, Hassert R, Pagel M, Risselada HJ, Naumov S, Beck-Sickinger AG, Abel B. Peptides@mica: from affinity to adhesion mechanism. Phys Chem Chem Phys 2016; 18:23516-27. [PMID: 27491508 DOI: 10.1039/c6cp03325c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Investigating the adsorption of peptides on inorganic surfaces, on the molecular level, is fundamental for medicinal and analytical applications. Peptides can be potent as linkers between surfaces and living cells in biochips or in implantation medicine. Here, we studied the adsorption process of the positively charged pentapeptide RTHRK, a recently identified binding sequence for surface oxidized silicon, and novel analogues thereof to negatively charged mica surfaces. Homogeneous formation of monolayers in the nano- and low micromolar peptide concentration range was observed. We propose an alternative and efficient method to both quantify binding affinity and follow adhesion behavior. This method makes use of the thermodynamic relationship between surface coverage, measured by atomic force microscopy (AFM), and the concomitant free energy of adhesion. A knowledge-based fit to the autocorrelation of the AFM images was used to correct for a biased surface coverage introduced by the finite lateral resolution of the AFM. Binding affinities and mechanisms were further explored by large scale molecular dynamics (MD) simulations. The combination of well validated MD simulations with topological data from AFM revealed a better understanding of peptide adsorption processes on the atomistic scale. We demonstrate that binding affinity is strongly determined by a peptide's ability to form salt bridges and hydrogen bonds with the surface lattice. Consequently, differences in hydrogen bond formation lead to substantial differences in binding affinity despite conservation of the peptide's overall charge. Further, MD simulations give access to relative changes in binding energy of peptide variations in comparison to a lead compound.
Collapse
Affiliation(s)
- A Gladytz
- Leibniz Institute of Surface Modification (IOM), Permoserstrasse 15, 04318 Leipzig, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Schönauer R, Els-Heindl S, Fischer JP, Köbberling J, Riedl B, Beck-Sickinger AG. Adrenomedullin 2.0: Adjusting Key Levers for Metabolic Stability. J Med Chem 2016; 59:5695-705. [PMID: 27166982 DOI: 10.1021/acs.jmedchem.6b00126] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The 52 amino acid peptide hormone adrenomedullin (ADM) plays a major role in the development and regulation of the cardiovascular and lymphatic system and has therefore gained significant interest for clinical applications. Because adrenomedullin exhibits low metabolic stability, enhancement of the plasma half-life is essential for peptide-based drug design. Fluorescently labeled ADM analogues synthesized by Fmoc/t-Bu solid phase peptide synthesis were used to analyze their enzymatic degradation and specific fragmentation pattern in human blood plasma. The determination of important cleavage sites allowed the development of selectively modified peptides in a rational approach. By combination of palmitoylation, lactam-bridging, and Nα-methylation, ADM analogues protected from enzymatic cleavage in human blood were developed and revealed an explicitly elongated half-life of 5 days in comparison to the wild-type in vitro. This triple-modification did not alter the selectivity of the analogues at the AM1 receptor, highlighting their potential for therapeutic applications.
Collapse
Affiliation(s)
- Ria Schönauer
- Institut für Biochemie, Universität Leipzig , Brüderstraße 34, 04103 Leipzig, Germany
| | - Sylvia Els-Heindl
- Institut für Biochemie, Universität Leipzig , Brüderstraße 34, 04103 Leipzig, Germany
| | - Jan-Patrick Fischer
- Institut für Biochemie, Universität Leipzig , Brüderstraße 34, 04103 Leipzig, Germany
| | | | - Bernd Riedl
- Bayer Pharma AG , Aprather Weg 18A, 42113 Wuppertal, Germany
| | | |
Collapse
|
32
|
Böhme D, Krieghoff J, Beck-Sickinger AG. Double Methotrexate-Modified Neuropeptide Y Analogues Express Increased Toxicity and Overcome Drug Resistance in Breast Cancer Cells. J Med Chem 2016; 59:3409-17. [DOI: 10.1021/acs.jmedchem.6b00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- David Böhme
- Universität Leipzig, Institute of Biochemistry, Brüderstraße 34, 04103 Leipzig, Germany
| | - Jan Krieghoff
- Universität Leipzig, Institute of Biochemistry, Brüderstraße 34, 04103 Leipzig, Germany
| | | |
Collapse
|
33
|
Li J, Tian Y, Wu A. Neuropeptide Y receptors: a promising target for cancer imaging and therapy. Regen Biomater 2015; 2:215-9. [PMID: 26816643 PMCID: PMC4669009 DOI: 10.1093/rb/rbv013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 12/12/2022] Open
Abstract
Neuropeptide Y (NPY) was first identified from porcine brain in 1982, and plays its biological functions in humans through NPY receptors (Y1, Y2, Y4 and Y5). NPY receptors are known to mediate various physiological functions and involve in a majority of human diseases, such as obesity, hypertension, epilepsy and metabolic disorders. Recently, NPY receptors have been found to be overexpressed in many cancers, so they emerged as promising target in cancer diagnosis and therapy. This review focuses on the latest research about NPY and NPY receptors, and summarizes the current knowledge on NPY receptors expression in cancers, selective ligands for NPY receptors and their application in cancer imaging and therapy.
Collapse
Affiliation(s)
- Juan Li
- Key Laboratory of Magnetic Materials and Devices & Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Yuchen Tian
- Key Laboratory of Magnetic Materials and Devices & Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices & Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo 315201, China
| |
Collapse
|
34
|
A cleavable cytolysin–neuropeptide Y bioconjugate enables specific drug delivery and demonstrates intracellular mode of action. J Control Release 2015; 209:170-8. [DOI: 10.1016/j.jconrel.2015.04.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/26/2015] [Accepted: 04/27/2015] [Indexed: 01/07/2023]
|