1
|
Jadhav K, Abhang A, Kole EB, Gadade D, Dusane A, Iyer A, Sharma A, Rout SK, Gholap AD, Naik J, Verma RK, Rojekar S. Peptide-Drug Conjugates as Next-Generation Therapeutics: Exploring the Potential and Clinical Progress. Bioengineering (Basel) 2025; 12:481. [PMID: 40428099 PMCID: PMC12108627 DOI: 10.3390/bioengineering12050481] [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: 03/19/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Peptide-drug conjugates (PDCs) have emerged as a next-generation therapeutic platform, combining the target specificity of peptides with the pharmacological potency of small-molecule drugs. As an evolution beyond antibody-drug conjugates (ADCs), PDCs offer distinct advantages, including enhanced cellular permeability, improved drug selectivity, and versatile design flexibility. This review provides a comprehensive analysis of the fundamental components of PDCs, including homing peptide selection, linker engineering, and payload optimization, alongside strategies to address their inherent challenges, such as stability, bioactivity, and clinical translation barriers. Therapeutic applications of PDCs span oncology, infectious diseases, metabolic disorders, and emerging areas like COVID-19, with several conjugates advancing in clinical trials and achieving regulatory milestones. Innovations, including bicyclic peptides, supramolecular architectures, and novel linker technologies, are explored as promising avenues to enhance PDC design. Additionally, this review examines the clinical trajectory of PDCs, emphasizing their therapeutic potential and highlighting ongoing trials that exemplify their efficacy. By addressing limitations and leveraging emerging advancements, PDCs hold immense promise as targeted therapeutics capable of addressing complex disease states and driving progress in precision medicine.
Collapse
Affiliation(s)
- Krishna Jadhav
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India; (K.J.); (R.K.V.)
| | - Ashwin Abhang
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT 06269, USA;
| | - Eknath B. Kole
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon 425001, Maharashtra, India; (E.B.K.); (J.N.)
| | - Dipak Gadade
- Department of Pharmaceutical Sciences, Delhi Skill and Entrepreneurship University, Dwarka Campus, Sector 9 Dwarka, New Delhi 110077, Delhi, India;
| | - Apurva Dusane
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA;
| | - Aditya Iyer
- Biopharmaceutics Department, Biocon Bristol-Myers Squibb R&D Center (BBRC), Bangalore 560099, Karnataka, India;
| | | | - Saroj Kumar Rout
- Research and Development, LNK International Inc., New York, NY 11788, USA;
| | - Amol D. Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India;
| | - Jitendra Naik
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon 425001, Maharashtra, India; (E.B.K.); (J.N.)
| | - Rahul K. Verma
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India; (K.J.); (R.K.V.)
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
2
|
Chapeau D, Beekman S, Piet A, Li L, de Ridder C, Stuurman D, Seimbille Y. eSOMA-DM1, a Maytansinoid-Based Theranostic Small-Molecule Drug Conjugate for Neuroendocrine Tumors. Bioconjug Chem 2024; 35:1823-1834. [PMID: 39395152 DOI: 10.1021/acs.bioconjchem.4c00413] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Background: The main challenges of conventional chemotherapy lie in its lack of selectivity and specificity, leading to significant side effects. Using a small-molecule drug conjugate (SMDC) ensures specific delivery of a cytotoxic drug to the tumor site by coupling it to a targeting vector. This promising strategy can be applied to neuroendocrine tumors (NETs) by choosing a targeting vector that binds specifically to somatostatin receptor subtype 2 (SSTR2). Additionally, incorporation of a bifunctional chelate into the molecule enables complexation of both diagnostic and therapeutic radionuclides. Thus, it facilitates monitoring of the distribution of the SMDC in the body and allows for the implementation of combination therapy. In our study, we designed eSOMA-DM1, a SMDC combining the SSTR2-targeted octreotate peptide and the cytotoxic agent DM1 via a chelate-bridged linker (N3-Py-DOTAGA). This approach warrants conjugation of the targeting vector and the drug at opposite sites to avoid undesired steric hindrance effects. Methods: Synthesis of the DM1 moiety (4) involved a three-step synthetic route, followed by the conjugation to the cyclic peptide, N3-Py-DOTAGA-d-Phe-cyclo[Cys-Tyr-d-Trp-Lys-Thr-Cys]-Thr-OH, through a copper-free click reaction, resulting in eSOMA-DM1. Subsequent labeling with [111In]InCl3 gave a high radiochemical yield and purity. In vitro assessments of eSOMA-DM1 binding, uptake, and internalization were conducted in SSTR2-transfected U2OS cells. Ex vivo biodistribution and fluorescence imaging were performed in H69-tumor bearing mice. Results: eSOMA-DM1 exhibited an IC50 value for SSTR2 similar to the gold standard DOTA-TATE. The uptake of [111In]In-eSOMA-DM1 in U2OS.SSTR2 cells was 1.2-fold lower than that of [111In]In-DOTA-TATE. Tumor uptake in H69-xenografted mice was higher for [111In]In-eSOMA-DM1 at all-time points compared to [111In]In-DOTA-TATE. Prolonged blood circulation led to increased accumulation of [111In]In-eSOMA-DM1 in highly vascularized tissues, such as the lungs, skin, and heart. Excretion through the kidneys, liver, and spleen was also observed. Conclusion: eSOMA-DM1 is a SMDC developed for NET showing promising characteristics in vitro. However, the in vivo results obtained with [111In]In-eSOMA-DM1 suggest the need for adjustments to optimize its distribution.
Collapse
Affiliation(s)
- Dylan Chapeau
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Savanne Beekman
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Amber Piet
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Le Li
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| |
Collapse
|
3
|
Oron-Herman M, Kirmayer D, Lupp A, Schulz S, Kostenich G, Afargan M. Expression prevalence and dynamics of GPCR somatostatin receptors 2 and 3 as cancer biomarkers beyond NET: a paired immunohistochemistry approach. Sci Rep 2023; 13:20857. [PMID: 38012197 PMCID: PMC10682014 DOI: 10.1038/s41598-023-47877-0] [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: 06/07/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023] Open
Abstract
Somatostatin receptors are clinically validated GPCR biomarkers for diagnosis and treatment of various neuroendocrine tumors (NET). Among the five somatostatin receptors, SST2 and SST3 are associated with apoptosis and cell cycle arrest, making these receptor subtypes better differentiated targets in precision oncology. In this study we performed immunohistochemistry of paired tissue microarrays containing 1125 cores, representing 43 tumor types, each stained for SST2 and SST3. A 12-point immunoreactive scoring (IRS) range was used for interpretation of the staining results. We analyzed the results twice, using the conventional positivity IRS cutoffs ≥ 3 and more stringent ≥ 6. Evaluation of receptors expression dynamics was performed for tumor-nodes-metastases (TNM) defined subgroups (ovarian and hepatocellular adenocarcinomas) as a function of their tumor stage. Our results indicate that two-thirds of tested cores exhibit clinically significant expression of at least SST2 or SST3 (IRS ≥ 6). The expression prevalence of both receptors tends to decline with tumor progression. However, an unexpected upregulation of both SST2 and SST3 reemerged in metastases suggesting conserved receptors genetic potential during tumor life cycle. We suggest that SST2 and SST3 should be further explored as potential biomarkers and therapeutic tools for maximizing the efficiency of somatostatin-based precision oncology of solid tumors beyond NET.
Collapse
Affiliation(s)
- Mor Oron-Herman
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel.
| | - David Kirmayer
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, 07747, Jena, Germany
| | - Genady Kostenich
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel
- The Advanced Technology Center, Sheba Medical Center, Tel Hashomer, 5262000, Ramat Gan, Israel
| | - Michel Afargan
- Starget Pharma, 26 Snir st., 4704086, Ramat Hasharon, Israel
| |
Collapse
|
4
|
Gong L, Zhao H, Liu Y, Wu H, Liu C, Chang S, Chen L, Jin M, Wang Q, Gao Z, Huang W. Research advances in peptide‒drug conjugates. Acta Pharm Sin B 2023; 13:3659-3677. [PMID: 37719380 PMCID: PMC10501876 DOI: 10.1016/j.apsb.2023.02.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
Peptide‒drug conjugates (PDCs) are drug delivery systems consisting of a drug covalently coupled to a multifunctional peptide via a cleavable linker. As an emerging prodrug strategy, PDCs not only preserve the function and bioactivity of the peptides but also release the drugs responsively with the cleavable property of the linkers. Given the ability to significantly improve the circulation stability and targeting of drugs in vivo and reduce the toxic side effects of drugs, PDCs have already been extensively applied in drug delivery. Herein, we review the types and mechanisms of peptides, linkers and drugs used to construct PDCs, and summarize the clinical applications and challenges of PDC drugs.
Collapse
Affiliation(s)
- Liming Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Heming Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Wu
- Department of Pharmacy, Yanbian University, Yanji 133000, China
| | - Chao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuangyan Chang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qiming Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
5
|
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: 92] [Impact Index Per Article: 46.0] [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
|
6
|
Guo K, Ma X, Li J, Zhang C, Wu L. Recent advances in combretastatin A-4 codrugs for cancer therapy. Eur J Med Chem 2022; 241:114660. [PMID: 35964428 DOI: 10.1016/j.ejmech.2022.114660] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
CA4 is a potent microtubule polymerization inhibitor and vascular disrupting agent. However, the in vivo efficiency of CA4 is limited owing to its poor pharmacokinetics resulting from its high lipophilicity and low water solubility. To improve the water solubility, CA4 phosphate (CA4P) has been developed and shows potent antivascular and antitumor effects. CA4P had been evaluated as a vascular disrupting agent in previousc linical trials. However, it had been discontinued due to the lack of a meaningful improvement in progression-free survival and unfavorable partial response data. Codrug is a drug design approach to chemically bind two or more drugs to improve therapeutic efficiency or decrease adverse effects. This review describes the progress made over the last twenty years in developing CA4-based codrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues. It also discusses the existing problems and the developmental prospects of CA4 codrugs.
Collapse
Affiliation(s)
- Kerong Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xin Ma
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jian Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
7
|
Vasco AV, Ceballos LG, Wessjohann LA, Rivera DG. Multicomponent Functionalization of the Octreotide Peptide Macrocyclic Scaffold. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Aldrin V. Vasco
- Leibniz Institute of Plant Biochemistry: Leibniz-Institut fur Pflanzenbiochemie Bioorganic Chemistry GERMANY
| | | | - Ludger A. Wessjohann
- Leibniz Institute of Plant Biochemistry: Leibniz-Institut fur Pflanzenbiochemie Bioorganic Chemistry GERMANY
| | - Daniel García Rivera
- Universidad de la Habana Laboratory of Synthetic and Biomolecular Chemistry Zapata y G 10400 La Habana CUBA
| |
Collapse
|
8
|
Furman O, Zaporozhets A, Tobi D, Bazylevich A, Firer MA, Patsenker L, Gellerman G, Lubin BCR. Novel Cyclic Peptides for Targeting EGFR and EGRvIII Mutation for Drug Delivery. Pharmaceutics 2022; 14:1505. [PMID: 35890400 PMCID: PMC9318536 DOI: 10.3390/pharmaceutics14071505] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/05/2023] Open
Abstract
The epidermal growth factor-epidermal growth factor receptor (EGF-EGFR) pathway has become the main focus of selective chemotherapeutic intervention. As a result, two classes of EGFR inhibitors have been clinically approved, namely monoclonal antibodies and small molecule kinase inhibitors. Despite an initial good response rate to these drugs, most patients develop drug resistance. Therefore, new treatment approaches are needed. In this work, we aimed to find a new EGFR-specific, short cyclic peptide, which could be used for targeted drug delivery. Phage display peptide technology and biopanning were applied to three EGFR expressing cells, including cells expressing the EGFRvIII mutation. DNA from the internalized phage was extracted and the peptide inserts were sequenced using next-generation sequencing (NGS). Eleven peptides were selected for further investigation using binding, internalization, and competition assays, and the results were confirmed by confocal microscopy and peptide docking. Among these eleven peptides, seven showed specific and selective binding and internalization into EGFR positive (EGFR+ve) cells, with two of them-P6 and P9-also demonstrating high specificity for non-small cell lung cancer (NSCLC) and glioblastoma cells, respectively. These peptides were chemically conjugated to camptothecin (CPT). The conjugates were more cytotoxic to EGFR+ve cells than free CPT. Our results describe a novel cyclic peptide, which can be used for targeted drug delivery to cells overexpressing the EGFR and EGFRvIII mutation.
Collapse
Affiliation(s)
- Olga Furman
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel; (O.F.); (M.A.F.)
- Agriculture and Oenology Department, Eastern Regional R&D Center, Ariel 40700, Israel
| | - Alisa Zaporozhets
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
| | - Dror Tobi
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Department of Molecular Biology, Ariel University, Ariel 40700, Israel
| | - Andrii Bazylevich
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
| | - Michael A. Firer
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel; (O.F.); (M.A.F.)
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Ariel Center for Applied Cancer Research, Ariel 40700, Israel
| | - Leonid Patsenker
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.Z.); (A.B.); (L.P.); (G.G.)
- Ariel Center for Applied Cancer Research, Ariel 40700, Israel
| | - Bat Chen R. Lubin
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel; (O.F.); (M.A.F.)
- Agriculture and Oenology Department, Eastern Regional R&D Center, Ariel 40700, Israel
| |
Collapse
|
9
|
Chen X, Liu F, Yu X, Li L, Yan J, Chen X, Liu Q, Liu B. An auristatin-based peptide-drug conjugate targeting Kita-Kyushu lung cancer antigen 1 for precision chemoradiotherapy in gastric cancer. Eur J Med Chem 2022; 241:114617. [DOI: 10.1016/j.ejmech.2022.114617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
|
10
|
Pryyma A, Matinkhoo K, Bu YJ, Merkens H, Zhang Z, Bénard F, Perrin DM. Synthesis and preliminary evaluation of octreotate conjugates of bioactive synthetic amatoxins for targeting somatostatin receptor (sstr2) expressing cells. RSC Chem Biol 2022; 3:69-78. [PMID: 35128410 PMCID: PMC8729174 DOI: 10.1039/d1cb00036e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022] Open
Abstract
Targeted cancer therapy represents a paradigm-shifting approach that aims to deliver a toxic payload selectively to target-expressing cells thereby sparing normal tissues the off-target effects associated with traditional chemotherapeutics. Since most targeted constructs rely on standard microtubule inhibitors or DNA-reactive molecules as payloads, new toxins that inhibit other intracellular targets are needed to realize the full potential of targeted therapy. Among these new payloads, α-amanitin has gained attraction as a payload in targeted therapy. Here, we conjugate two synthetic amanitins at different sites to demonstrate their utility as payloads in peptide drug conjugates (PDCs). As an exemplary targeting agent, we chose octreotate, a well-studied somatostatin receptor (sstr2) peptide agonist for the conjugation to synthetic amatoxins via three tailor-built linkers. The linker chemistry permitted the evaluation of one non-cleavable and two cleavable self-immolative conjugates. The immolating linkers were chosen to take advantage of either the reducing potential of the intracellular environment or the high levels of lysosomal proteases in tumor cells to trigger toxin release. Cell-based assays on target-positive Ar42J cells revealed target-specific reduction in viability with up to 1000-fold enhancement in bioactivity compared to the untargeted amatoxins. Altogether, this preliminary study enabled the development of a highly modular synthetic platform for the construction of amanitin-based conjugates that can be readily extended to various targeting moieties.
Collapse
Affiliation(s)
- Alla Pryyma
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Kaveh Matinkhoo
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Yong Jia Bu
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - Zhengxing Zhang
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - Francois Bénard
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - David M Perrin
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| |
Collapse
|
11
|
Dual-dye systems comprising activatable fluorescein dye and hydrophobic or hydrophilic Cy5 reference fluorophore for ratiometric drug delivery monitoring. J Photochem Photobiol A Chem 2021. [DOI: 10.1016/j.jphotochem.2020.113113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
12
|
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
|
13
|
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: 26] [Impact Index Per Article: 5.2] [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
|
14
|
Ebaston TM, Rozovsky A, Zaporozhets A, Bazylevich A, Tuchinsky H, Marks V, Gellerman G, Patsenker LD. Peptide‐Driven Targeted Drug‐Delivery System Comprising Turn‐On Near‐Infrared Fluorescent Xanthene–Cyanine Reporter for Real‐Time Monitoring of Drug Release. ChemMedChem 2019; 14:1727-1734. [DOI: 10.1002/cmdc.201900464] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 12/25/2022]
Affiliation(s)
- T. M. Ebaston
- Department of Chemical SciencesAriel University Ariel 40700 Israel
| | - Alex Rozovsky
- Department of Chemical SciencesAriel University Ariel 40700 Israel
| | | | | | - Helena Tuchinsky
- Department of Molecular BiologyAriel University Ariel 40700 Israel
| | - Vered Marks
- Department of Chemical SciencesAriel University Ariel 40700 Israel
| | - Gary Gellerman
- Department of Chemical SciencesAriel University Ariel 40700 Israel
| | | |
Collapse
|
15
|
Solomonov AV, Marfin YS, Rumyantsev EV, Ragozin E, Zahavi TS, Gellerman G, Tesler AB, Muench F, Kumagai A, Miyawaki A. Self-assembled micellar clusters based on Triton-X-family surfactants for enhanced solubilization, encapsulation, proteins permeability control, and anticancer drug delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:794-804. [DOI: 10.1016/j.msec.2019.02.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/11/2022]
|
16
|
White BH, Whalen K, Kriksciukaite K, Alargova R, Au Yeung T, Bazinet P, Brockman A, DuPont M, Oller H, Lemelin CA, Lim Soo P, Moreau B, Perino S, Quinn JM, Sharma G, Shinde R, Sweryda-Krawiec B, Wooster R, Bilodeau MT. Discovery of an SSTR2-Targeting Maytansinoid Conjugate (PEN-221) with Potent Activity in Vitro and in Vivo. J Med Chem 2019; 62:2708-2719. [PMID: 30735385 DOI: 10.1021/acs.jmedchem.8b02036] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Somatostatin receptor 2 (SSTR2) is frequently overexpressed on several types of solid tumors, including neuroendocrine tumors and small-cell lung cancer. Peptide agonists of SSTR2 are rapidly internalized upon binding to the receptor and linking a toxic payload to an SSTR2 agonist is a potential method to kill SSTR2-expressing tumor cells. Herein, we describe our efforts towards an efficacious SSTR2-targeting cytotoxic conjugate; examination of different SSTR2-targeting ligands, conjugation sites, and payloads led to the discovery of 22 (PEN-221), a conjugate consisting of microtubule-targeting agent DM1 linked to the C-terminal side chain of Tyr3-octreotate. PEN-221 demonstrates in vitro activity which is both potent (IC50 = 10 nM) and receptor-dependent (IC50 shifts 90-fold upon receptor blockade). PEN-221 targets high levels of DM1 to SSTR2-expressing xenograft tumors, which has led to tumor regressions in several SSTR2-expressing xenograft mouse models. The safety and efficacy of PEN-221 is currently under evaluation in human clinical trials.
Collapse
Affiliation(s)
- Brian H White
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Kerry Whalen
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Kristina Kriksciukaite
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Rossitza Alargova
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Tsun Au Yeung
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Patrick Bazinet
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Adam Brockman
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Michelle DuPont
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Haley Oller
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Charles-Andre Lemelin
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Patrick Lim Soo
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Benoît Moreau
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Samantha Perino
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - James M Quinn
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Gitanjali Sharma
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Rajesh Shinde
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Beata Sweryda-Krawiec
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Richard Wooster
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| | - Mark T Bilodeau
- Tarveda Therapeutics , 134 Coolidge Avenue , Watertown , Massachusetts 02472 , United States
| |
Collapse
|
17
|
Rozovsky A, Ebaston TM, Zaporozhets A, Bazylevich A, Tuchinsky H, Patsenker L, Gellerman G. Theranostic system for ratiometric fluorescence monitoring of peptide-guided targeted drug delivery. RSC Adv 2019; 9:32656-32664. [PMID: 35529716 PMCID: PMC9073098 DOI: 10.1039/c9ra06334j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/04/2019] [Indexed: 11/21/2022] Open
Abstract
Novel theranostic system that first combines a cancer-targeting peptide with a long-wavelength dual fluorescent dye IRD in order to provide ratiometric monitoring of anticancer drug delivery is developed and evaluated in pancreatic cancer cell line.
Collapse
Affiliation(s)
- Alex Rozovsky
- Department of Chemical Sciences
- Ariel University
- Ariel 40700
- Israel
| | - T. M. Ebaston
- Department of Chemical Sciences
- Ariel University
- Ariel 40700
- Israel
| | | | | | - Helena Tuchinsky
- Department of Molecular Biology
- Ariel University
- Ariel 40700
- Israel
| | - Leonid Patsenker
- Department of Chemical Sciences
- Ariel University
- Ariel 40700
- Israel
| | - Gary Gellerman
- Department of Chemical Sciences
- Ariel University
- Ariel 40700
- Israel
| |
Collapse
|
18
|
Ragozin E, Hesin A, Bazylevich A, Tuchinsky H, Bovina A, Shekhter Zahavi T, Oron-Herman M, Kostenich G, Firer M, Rubinek T, Wolf I, Luboshits G, Sherman M, Gellerman G. New somatostatin-drug conjugates for effective targeting pancreatic cancer. Bioorg Med Chem 2018; 26:3825-3836. [DOI: 10.1016/j.bmc.2018.06.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/14/2018] [Accepted: 06/24/2018] [Indexed: 12/15/2022]
|
19
|
Therapeutic journery of nitrogen mustard as alkylating anticancer agents: Historic to future perspectives. Eur J Med Chem 2018; 151:401-433. [DOI: 10.1016/j.ejmech.2018.04.001] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/30/2018] [Accepted: 04/01/2018] [Indexed: 12/17/2022]
|
20
|
Yacobovich S, Tuchinsky L, Kirby M, Kardash T, Agranyoni O, Nesher E, Redko B, Gellerman G, Tobi D, Gurova K, Koman I, Ashur Fabian O, Pinhasov A. Novel synthetic cyclic integrin αvβ3 binding peptide ALOS4: Antitumor activity in mouse melanoma models. Oncotarget 2018; 7:63549-63560. [PMID: 27556860 PMCID: PMC5325384 DOI: 10.18632/oncotarget.11363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 07/27/2016] [Indexed: 12/15/2022] Open
Abstract
ALOS4, a unique synthetic cyclic peptide without resemblance to known integrin ligand sequences, was discovered through repeated biopanning with pIII phage expressing a disulfide-constrained nonapeptide library. Binding assays using a FITC-labeled analogue demonstrated selective binding to immobilized αvβ3 and a lack of significant binding to other common proteins, such as bovine serum albumin and collagen. In B16F10 cell cultures, ALOS4 treatment at 72 h inhibited cell migration (30%) and adhesion (up to 67%). Immunofluorescent imaging an ALOS4-FITC analogue with B16F10 cells demonstrated rapid cell surface binding, and uptake and localization in the cytoplasm. Daily injections of ALOS4 (0.1, 0.3 or 0.5 mg/kg i.p.) to mice inoculated with B16F10 mouse melanoma cells in two different cancer models, metastatic and subcutaneous tumor, resulted in reduction of lung tumor count (metastatic) and tumor mass (subcutaneous) and increased survival of animals monitored to 45 and 60 days, respectively. Examination of cellular activity indicated that ALOS4 produces inhibition of cell migration and adhesion in a concentration-dependent manner. Collectively, these results suggest that ALOS4 is a structurally-unique selective αvβ3 integrin ligand with potential anti-metastatic activity.
Collapse
Affiliation(s)
- Shiri Yacobovich
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Lena Tuchinsky
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Michael Kirby
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Tetiana Kardash
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Oryan Agranyoni
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Elimelech Nesher
- Department of Molecular Biology, Ariel University, Ariel, Israel.,Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Boris Redko
- Department of Chemical Sciences, Ariel University, Ariel, Israel
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel, Israel
| | - Dror Tobi
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Katerina Gurova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Igor Koman
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Osnat Ashur Fabian
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Ariel University, Ariel, Israel
| |
Collapse
|
21
|
Redko B, Tuchinsky H, Segal T, Tobi D, Luboshits G, Ashur-Fabian O, Pinhasov A, Gerlitz G, Gellerman G. Toward the development of a novel non-RGD cyclic peptide drug conjugate for treatment of human metastatic melanoma. Oncotarget 2018; 8:757-768. [PMID: 27768593 PMCID: PMC5352194 DOI: 10.18632/oncotarget.12748] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022] Open
Abstract
The newly discovered short (9 amino acid) non-RGD S-S bridged cyclic peptide ALOS-4 (H-cycl(Cys-Ser-Ser-Ala-Gly-Ser-Leu-Phe-Cys)-OH), which binds to integrin αvβ3 is investigated as peptide carrier for targeted drug delivery against human metastatic melanoma. ALOS4 binds specifically the αvβ3 overexpressing human metastatic melanoma WM-266-4 cell line both in vitro and in ex vivo assays. Coupling ALOS4 to the topoisomerase I inhibitor Camptothecin (ALOS4-CPT) increases the cytotoxicity of CPT against human metastatic melanoma cells while reduces dramatically the cytotoxicity against non-cancerous cells as measured by the levels of γH2A.X, active caspase 3 and cell viability. Moreover, conjugating ALOS4 to CPT even increases the chemo-stability of CPT under physiological pH. Bioinformatic analysis using Rosetta platform revealed potential docking sites of ALOS4 on the αvβ3 integrin which are distinct from the RGD binding sites. We propose to use this specific non-RGD cyclic peptide as the therapeutic carrier for conjugation of drugs in order to improve efficacy and reduce toxicity of currently available treatments of human malignant melanoma.
Collapse
Affiliation(s)
- Boris Redko
- Department of Chemical Sciences, Ariel University, Ariel, Israel
| | - Helena Tuchinsky
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Tamar Segal
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Dror Tobi
- Department of Molecular Biology, Ariel University, Ariel, Israel.,Department of Computer Science, Ariel University, Ariel, Israel
| | - Galia Luboshits
- Department of Chemical Engineering, Ariel University, Ariel, Israel
| | - Osnat Ashur-Fabian
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Gabi Gerlitz
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
22
|
Khan M, Huang T, Lin CY, Wu J, Fan BM, Bian ZX. Exploiting cancer's phenotypic guise against itself: targeting ectopically expressed peptide G-protein coupled receptors for lung cancer therapy. Oncotarget 2017; 8:104615-104637. [PMID: 29262666 PMCID: PMC5732832 DOI: 10.18632/oncotarget.18403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023] Open
Abstract
Lung cancer, claiming millions of lives annually, has the highest mortality rate worldwide. This advocates the development of novel cancer therapies that are highly toxic for cancer cells but negligibly toxic for healthy cells. One of the effective treatments is targeting overexpressed surface receptors of cancer cells with receptor-specific drugs. The receptors-in-focus in the current review are the G-protein coupled receptors (GPCRs), which are often overexpressed in various types of tumors. The peptide subfamily of GPCRs is the pivot of the current article owing to the high affinity and specificity to and of their cognate peptide ligands, and the proven efficacy of peptide-based therapeutics. The article summarizes various ectopically expressed peptide GPCRs in lung cancer, namely, Cholecystokinin-B/Gastrin receptor, the Bombesin receptor family, Bradykinin B1 and B2 receptors, Arginine vasopressin receptors 1a, 1b and 2, and the Somatostatin receptor type 2. The autocrine growth and pro-proliferative pathways they mediate, and the distinct tumor-inhibitory effects of somatostatin receptors are then discussed. The next section covers how these pathways may be influenced or 'corrected' through therapeutics (involving agonists and antagonists) targeting the overexpressed peptide GPCRs. The review proceeds on to Nano-scaled delivery platforms, which enclose chemotherapeutic agents and are decorated with peptide ligands on their external surface, as an effective means of targeting cancer cells. We conclude that targeting these overexpressed peptide GPCRs is potentially evolving as a highly promising form of lung cancer therapy.
Collapse
Affiliation(s)
- Mahjabin Khan
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
| | - Tao Huang
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
| | - Cheng-Yuan Lin
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
- YMU-HKBU Joint Laboratory of Traditional Natural Medicine, Yunnan Minzu University, Kunming, P.R. China
| | - Jiang Wu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, P. R. China
| | - Bao-Min Fan
- YMU-HKBU Joint Laboratory of Traditional Natural Medicine, Yunnan Minzu University, Kunming, P.R. China
| | - Zhao-Xiang Bian
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
| |
Collapse
|
23
|
Rayappan K, Murugan C, Sundarraj S, Lara RP, Kannan S. Peptide-Conjugated Nano-Drug Delivery System to Improve Synergistic Molecular Chemotherapy for Colon Carcinoma. ChemistrySelect 2017. [DOI: 10.1002/slct.201701153] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kathirvel Rayappan
- Division of Cancer Nanomedicine; Department of Zoology; Periyar University; Salem - 636 011, Tamil Nadu INDIA
| | - Chandran Murugan
- Division of Cancer Nanomedicine; Department of Zoology; Periyar University; Salem - 636 011, Tamil Nadu INDIA
| | - Shenbagamoorthy Sundarraj
- Division of Cancer Nanomedicine; Department of Zoology; Periyar University; Salem - 636 011, Tamil Nadu INDIA
- PG and Research Department of Zoology, Ayya Nadar Janaki Ammal College; Sivakasi - 626124, Tamil Nadu, INDIA
| | | | - Soundarapandian Kannan
- Division of Cancer Nanomedicine; Department of Zoology; Periyar University; Salem - 636 011, Tamil Nadu INDIA
| |
Collapse
|
24
|
Bashari O, Redko B, Cohen A, Luboshits G, Gellerman G, Firer MA. Discovery of peptide drug carrier candidates for targeted multi-drug delivery into prostate cancer cells. Cancer Lett 2017; 408:164-173. [PMID: 28888997 DOI: 10.1016/j.canlet.2017.08.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 11/24/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains essentially incurable. Targeted Drug Delivery (TDD) systems may overcome the limitations of current mCRPC therapies. We describe the use of strict criteria to isolate novel prostate cancer cell targeting peptides that specifically deliver drugs into target cells. Phage from a libraries displaying 7mer peptides were exposed to PC-3 cells and only internalized phage were recovered. The ability of these phage to internalize into other prostate cancer cells (LNCaP, DU-145) was validated. The displayed peptides of selected phage clones were synthesized and their specificity for target cells was validated in vitro and in vivo. One peptide (P12) which specifically targeted PC-3 tumors in vivo was incorporated into mono-drug (Chlorambucil, Combretastatin or Camptothecin) and dual-drug (Chlorambucil/Combretastatin or Chlorambucil/Camptothecin) PDCs and the cytotoxic efficacy of these conjugates for target cells was tested. Conjugation of P12 into dual-drug PDCs allowed discovery of new drug combinations with synergistic effects. The use of strict selection criteria can lead to discovery of novel peptides for use as drug carriers for TDD. PDCs represent an effective alternative to current modes of free drug chemotherapy for prostate cancer.
Collapse
Affiliation(s)
- O Bashari
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - B Redko
- Dept. Chemical Sciences, Ariel University, Ariel, 40700, Israel.
| | - A Cohen
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - G Luboshits
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| | - G Gellerman
- Dept. Chemical Sciences, Ariel University, Ariel, 40700, Israel.
| | - M A Firer
- Dept. Chemical Engineering, Ariel University, Ariel, 40700, Israel.
| |
Collapse
|
25
|
Vats K, Satpati D, Sharma R, Kumar C, Sarma HD, Dash A. 99m
Tc-labeled NGR-chlorambucil conjugate, 99m
Tc-HYNIC-CLB-c(NGR) for targeted chemotherapy and molecular imaging. J Labelled Comp Radiopharm 2017; 60:431-438. [DOI: 10.1002/jlcr.3522] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/12/2017] [Accepted: 05/19/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Kusum Vats
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai India
| | - Drishty Satpati
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai India
| | - Rohit Sharma
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai India
| | - Chandan Kumar
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai India
| | - Haladhar D. Sarma
- Radiation Biology and Health Science Division; Bhabha Atomic Research Centre; Mumbai India
| | - Ashutosh Dash
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai India
| |
Collapse
|
26
|
Gilad Y, Firer M, Gellerman G. Recent Innovations in Peptide Based Targeted Drug Delivery to Cancer Cells. Biomedicines 2016; 4:E11. [PMID: 28536378 PMCID: PMC5344250 DOI: 10.3390/biomedicines4020011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/16/2016] [Accepted: 05/23/2016] [Indexed: 12/21/2022] Open
Abstract
Targeted delivery of chemotherapeutics and diagnostic agents conjugated to carrier ligands has made significant progress in recent years, both in regards to the structural design of the conjugates and their biological effectiveness. The goal of targeting specific cell surface receptors through structural compatibility has encouraged the use of peptides as highly specific carriers as short peptides are usually non-antigenic, are structurally simple and synthetically diverse. Recent years have seen many developments in the field of peptide based drug conjugates (PDCs), particularly for cancer therapy, as their use aims to bypass off-target side-effects, reducing the morbidity common to conventional chemotherapy. However, no PDCs have as yet obtained regulatory approval. In this review, we describe the evolution of the peptide-based strategy for targeted delivery of chemotherapeutics and discuss recent innovations in the arena that should lead in the near future to their clinical application.
Collapse
Affiliation(s)
- Yosi Gilad
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel.
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel 40700, Israel.
| | - Michael Firer
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel 40700, Israel.
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel.
| |
Collapse
|
27
|
Ragozin E, Redko B, Tuchinsky E, Rozovsky A, Albeck A, Grynszpan F, Gellerman G. Biolabile peptidyl delivery systems toward sequential drug release. Biopolymers 2016; 106:119-32. [DOI: 10.1002/bip.22794] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/22/2015] [Accepted: 11/30/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Elena Ragozin
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
| | - Boris Redko
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
- Department of Chemistry; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Elena Tuchinsky
- Department of Molecular Biology; Ariel University; Ariel 40700 Israel
| | - Alex Rozovsky
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
- Department of Chemistry; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Amnon Albeck
- Department of Chemistry; Bar-Ilan University; Ramat Gan 52900 Israel
| | - Flavio Grynszpan
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
| | - Gary Gellerman
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
| |
Collapse
|