1
|
Alkhamach D, Khan SA, Greish K, Hassan HAFM, Haider M. Nanostructured lipid carriers in cancer Therapy: Advances in passive and active targeting strategies. Int J Pharm 2025:125736. [PMID: 40389069 DOI: 10.1016/j.ijpharm.2025.125736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025]
Abstract
Nanostructured lipid carriers (NLCs) have emerged as a promising drug delivery platform in cancer therapy, offering advantages such as enhanced drug solubility, stability, and controlled release. Recent efforts have focused on utilizing NLCs for passive and active tumor targeting to improve therapeutic outcomes. This review provides a comprehensive analysis of the role of NLCs in cancer therapy, with particular emphasis on their application in passive and active targeting strategies for precision oncology. Relevant studies were selected from recent literature, focusing on NLC formulation, targeting approaches, and therapeutic applications. NLCs enhance tumor-specific drug delivery through passive targeting via the enhanced permeability and retention (EPR) effect and active targeting via ligand-mediated mechanisms. Lymphatic-targeting NLCs enable improved drug delivery to metastatic niches, while stimuli-responsive NLCs facilitate site-specific release under tumor-associated conditions (e.g., pH, enzymatic activity, redox gradients). Advances in lipid composition, surfactant systems, and conjugation strategies significantly influence drug loading (DL), biodistribution, therapeutic efficacy, and clinical translation across various malignancies. NLCs represent a versatile and adaptable platform for precision cancer therapy. Continued optimization of formulation parameters, functionalization strategies, and clinical translation pathways is essential to fully realize their potential in targeted oncology applications.
Collapse
Affiliation(s)
- Dana Alkhamach
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates
| | - Saeed Ahmad Khan
- Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates
| | - Khaled Greish
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences Arabian Gulf University, Manama 328329, Bahrain
| | - Hatem A F M Hassan
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK
| | - Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
2
|
Yadav AK, Kushwaha R, Mandal AA, Mandal A, Banerjee S. Intracellular Photocatalytic NADH/NAD(P)H Oxidation for Cancer Drug Development. J Am Chem Soc 2025; 147:7161-7181. [PMID: 39980079 DOI: 10.1021/jacs.4c18328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Photocatalytic cancer therapy (PCT) has emerged as a cutting-edge anticancer mechanism of action, harnessing light energy to mediate the catalytic oxidation of intracellular substrates. PCT is of significant current importance due to its potential to address the limitations of conventional chemotherapy, particularly drug resistance and side effects. This approach offers a noninvasive, targeted cancer treatment option by utilizing metal-based photocatalysts to induce redox and metabolic disorders within cancer cells. The photocatalysts disrupt the cancer cell metabolism by converting NADH/NAD(P)H to NAD+/NAD(P)+ via catalytic photoredox processes, altering intracellular NAD+/NADH or NAD(P)+/NAD(P)H ratios, which are crucial for cellular metabolism. Ir(III), Ru(II), Re(I), and Os(II) photocatalysts demonstrated promising PCT efficacy. Despite these developments, gaps remain in the literature for translating this new anticancer mechanism into clinical trials. This Perspective critically examines the developments in this research area and provides future directions for designing efficient photocatalysts for PCT.
Collapse
Affiliation(s)
- Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Apurba Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
3
|
Zhang DE, He T, Shi T, Huang K, Peng A. Trends in the research and development of peptide drug conjugates: artificial intelligence aided design. Front Pharmacol 2025; 16:1553853. [PMID: 40083376 PMCID: PMC11903715 DOI: 10.3389/fphar.2025.1553853] [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: 12/31/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Peptide-drug conjugates (PDCs) represent an emerging class of targeted therapeutic agents that consist of small molecular drugs coupled to multifunctional peptides through cleavable or non-cleavable linkers. The principal advantage of PDCs lies in their capacity to deliver drugs to diseased tissues at increased local concentrations, thereby reducing toxicity and mitigating adverse effects by limiting damage to non-diseased tissues. Despite the increasing number of PDCs being developed for various diseases, their advancements remain relatively slow due to several development constraints, which include limited available peptides and linkers, narrow therapeutic applications, and incomplete evaluation and information platforms for PDCs. Marked by the recent Nobel Prize awarded to artificial intelligence (AI) and de novo protein design for "protein design and structure prediction," AI is playing an increasingly important role in drug discovery and development. In this review, we summarize the recent developments and limitations of PDCs, highlights the potential of AI in revolutionizing the design and evaluation of PDC.
Collapse
Affiliation(s)
- Dong-E Zhang
- The Third Hospital of Wuhan, Hubei University of Chinese Medicine, Wuhan, China
| | - Tong He
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyi Shi
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Tongji-RongCheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anlin Peng
- The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
He H, Deng X, Wang Z, Chen J. Recent progress in the development of peptide-drug conjugates (PDCs) for cancer therapy. Eur J Med Chem 2025; 284:117204. [PMID: 39731788 DOI: 10.1016/j.ejmech.2024.117204] [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: 11/04/2024] [Revised: 12/10/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
Peptide-drug conjugates (PDCs) are emerging therapeutic agents composed of peptides, linkers, and payloads, which possess favorable targeting capability and can deliver enough payloads to the tumor sites with minimized impact on healthy tissues. However, only a few PDCs have been approved for clinical use so far. To advance the research on PDCs, this review summarizes the approved PDCs, and PDCs in clinical and preclinical stages based on the payload types. Additionally, the biological activity and pharmacokinetic properties of preclinical PDCs are detailedly described. Lastly, the challenges and future development directions of PDCs are discussed. This review aims to inspire insights into the development of PDCs for cancer treatment.
Collapse
Affiliation(s)
- Haiqi He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic Al Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhijie Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Bogdanović B, Fagret D, Ghezzi C, Montemagno C. Integrin Targeting and Beyond: Enhancing Cancer Treatment with Dual-Targeting RGD (Arginine-Glycine-Aspartate) Strategies. Pharmaceuticals (Basel) 2024; 17:1556. [PMID: 39598465 PMCID: PMC11597078 DOI: 10.3390/ph17111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Integrins, an important superfamily of cell adhesion receptors, play an essential role in cancer progression, metastasis, and angiogenesis, establishing them as prime targets for both diagnostic and therapeutic applications. Despite their significant potential, integrin-targeted therapies have faced substantial challenges in clinical trials, including variable efficacy and unmet high expectations. Nevertheless, the consistent expression of integrins on tumor and stromal cells underscores their ongoing relevance and potential. Traditional RGD-based imaging and therapeutic agents have faced limitations, such as inconsistent target expression and rapid systemic clearance, which have reduced their effectiveness. To overcome these challenges, recent research has focused on advancing RGD-based strategies and exploring innovative solutions. This review offers a thorough analysis of the latest developments in the RGD-integrin field, with a particular focus on addressing previous limitations. It delves into new dual-targeting approaches and cutting-edge RGD-based agents designed to improve both tumor diagnosis and therapeutic outcomes. By examining these advancements, this review illuminates new pathways for enhancing the specificity and efficacy of integrin-targeted therapies, paving the way for more effective cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bojana Bogdanović
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Daniel Fagret
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Catherine Ghezzi
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | | |
Collapse
|
6
|
Sanz-Villafruela J, Bermejo-Casadesús C, Riesco-Llach G, Iglesias M, Martínez-Alonso M, Planas M, Feliu L, Espino G, Massaguer A. Bombesin-Targeted Delivery of β-Carboline-Based Ir(III) and Ru(II) Photosensitizers for a Selective Photodynamic Therapy of Prostate Cancer. Inorg Chem 2024; 63:19140-19155. [PMID: 39361042 PMCID: PMC11483813 DOI: 10.1021/acs.inorgchem.4c02583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024]
Abstract
Despite advances in Ir(III) and Ru(II) photosensitizers (PSs), their lack of selectivity for cancer cells has hindered their use in photodynamic therapy (PDT). We disclose the synthesis and characterization of two pairs of Ir(III) and Ru(II) polypyridyl complexes bearing two β-carboline ligands (N^N') functionalized with -COOMe (L1) or -COOH (L2), resulting in PSs of formulas [Ir(C^N)2(N^N')]Cl (Ir-Me: C^N = ppy, N^N' = L1; Ir-H: C^N = ppy, N^N' = L2) and [Ru(N^N)2(N^N')](Cl)2 (Ru-Me: N^N = bpy, N^N' = L1; Ru-H: N^N = bpy, N^N' = L2). To enhance their selectivity toward cancer cells, Ir-H and Ru-H were coupled to a bombesin derivative (BN3), resulting in the metallopeptides Ir-BN and Ru-BN. Ir(III) complexes showed higher anticancer activity than their Ru(II) counterparts, particularly upon blue light irradiation, but lacked cancer cell selectivity. In contrast, Ir-BN and Ru-BN exhibited selective photocytoxicity against prostate cancer cells, with a lower effect against nonmalignant fibroblasts. All compounds generated ROS and induced severe mitochondrial toxicity upon photoactivation, leading to apoptosis. Additionally, the ability of Ir-Me to oxidize NADH was demonstrated, suggesting a mechanism for mitochondrial damage. Our findings indicated that the conjugation of metal PSs with BN3 creates efficient PDT agents, achieving selectivity through targeting bombesin receptors and local photoactivation.
Collapse
Affiliation(s)
- Juan Sanz-Villafruela
- Universidad
de Burgos, Departamento de
Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, Burgos 09001, Spain
| | - Cristina Bermejo-Casadesús
- Universitat
de Girona, Departament de
Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, Girona 17003, Spain
| | - Gerard Riesco-Llach
- LIPPSO,
Departament de Química, Facultat de Ciències, Universitat de Girona, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Mònica Iglesias
- Universitat
de Girona, Departament de Química,
Facultat de Ciències, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Marta Martínez-Alonso
- Universidad
de Burgos, Departamento de
Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, Burgos 09001, Spain
| | - Marta Planas
- LIPPSO,
Departament de Química, Facultat de Ciències, Universitat de Girona, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Lidia Feliu
- LIPPSO,
Departament de Química, Facultat de Ciències, Universitat de Girona, Maria Aurelia Capmany 69, Girona 17003, Spain
| | - Gustavo Espino
- Universidad
de Burgos, Departamento de
Química, Facultad de Ciencias, Plaza Misael Bañuelos s/n, Burgos 09001, Spain
| | - Anna Massaguer
- Universitat
de Girona, Departament de
Biologia, Facultat de Ciències, Maria Aurelia Capmany 40, Girona 17003, Spain
| |
Collapse
|
7
|
Wang Y, Zhang L, Liu C, Luo Y, Chen D. Peptide-Mediated Nanocarriers for Targeted Drug Delivery: Developments and Strategies. Pharmaceutics 2024; 16:240. [PMID: 38399294 PMCID: PMC10893007 DOI: 10.3390/pharmaceutics16020240] [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: 01/15/2024] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Effective drug delivery is essential for cancer treatment. Drug delivery systems, which can be tailored to targeted transport and integrated tumor therapy, are vital in improving the efficiency of cancer treatment. Peptides play a significant role in various biological and physiological functions and offer high design flexibility, excellent biocompatibility, adjustable morphology, and biodegradability, making them promising candidates for drug delivery. This paper reviews peptide-mediated drug delivery systems, focusing on self-assembled peptides and peptide-drug conjugates. It discusses the mechanisms and structural control of self-assembled peptides, the varieties and roles of peptide-drug conjugates, and strategies to augment peptide stability. The review concludes by addressing challenges and future directions.
Collapse
Affiliation(s)
- Yubo Wang
- Medical College, Guangxi University, Da-Xue-Dong Road No. 100, Nanning 530004, China;
| | - Lu Zhang
- School of Life Sciences, Xiamen University, Xiamen 361005, China;
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| | - Yiming Luo
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen 361003, China
- The School of Clinical Medicine, Fujian Medical University, Fuzhou 351002, China
| | - Dengyue Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| |
Collapse
|
8
|
Beloborodov E, Iurova E, Sugak D, Rastorgueva E, Pogodina E, Fomin A, Viktorov D, Slesarev S, Saenko Y. Stabilizing Scaffold for Short Peptides Based on Knottins. Curr Cancer Drug Targets 2024; 24:1275-1285. [PMID: 38357956 DOI: 10.2174/0115680096285288240118090050] [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: 10/03/2023] [Revised: 12/23/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Bombesin (BBN) is a short peptide with a high affinity for receptors that are expressed on the surface of various types of cancer cells. However, a full length BBN molecule has low in vivo stability. OBJECTIVE In our study, we propose the use of peptide toxins, derived from animal and plant toxins, as scaffold molecules to enhance the bioavailability and stability of bombesin. These peptides possess a unique structure known as an inhibitory cystine knot. METHODS We synthesized structures in which short bombesin was incorporated into various domains of arthropod and plant toxins using solid-phase peptide synthesis. The stability under different conditions was assessed through high-performance liquid chromatography, and binding to cell cultures expressing the bombesin receptor was analyzed. Additionally, toxicity to cell cultures was evaluated using fluorescence microscopy. RESULTS The data obtained demonstrated that placing the short peptide between the first and second cysteine residues in arachnid toxins results in increased in vitro stability and bioavailability, as well as low cytotoxicity. CONCLUSION Arachnid toxins with an inhibitory cystine knot can be considered as a scaffold for increasing the stability of therapeutic peptides.
Collapse
Affiliation(s)
- Evgenii Beloborodov
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| | - Elena Iurova
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| | - Dmitrii Sugak
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| | - Eugenia Rastorgueva
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
- Department of General and Clinical Pharmacology and Microbiology, Faculty of Medicine, Ulyanovsk State University, Ulyanovsk, Russia
| | - Evgeniya Pogodina
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| | - Aleksandr Fomin
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| | - Denis Viktorov
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| | - Sergei Slesarev
- Department of Biology, Ecology and Natural Resources Management, Faculty of Ecology, Ulyanovsk State University, Ulyanovsk, Russia
| | - Yury Saenko
- Laboratory of Research and Development of Peptide Drugs and Vaccines, S.P. Kapitsa Technological Research Institute, Ulyanovsk State University, Ulyanovsk, Russia
| |
Collapse
|
9
|
Vila-Julià G, Rubio-Martinez J, Perez JJ. Assessment of the bound conformation of bombesin to the BB1 and BB2 receptors. Int J Biol Macromol 2024; 255:127843. [PMID: 37956803 DOI: 10.1016/j.ijbiomac.2023.127843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
Bombesin is an endogenous peptide involved in a wide spectrum of physiological activities ranging from satiety, control of circadian rhythm and thermoregulation in the central nervous system, to stimulation of gastrointestinal hormone release, activation of macrophages and effects on development in peripheral tissues. Actions of the peptide are mediated through the two high affinity G-protein coupled receptors BB1R and BB2R. Under pathophysiological conditions, these receptors are overexpressed in many different types of tumors, such as prostate cancer, breast cancer, small and non-small cell lung cancer and pancreatic cancer. This observation has been used for designing cell markers, but it has not been yet exploited for therapeutical purposes. Despite the enormous biological interest of the peptide, little is known about the stereochemical features that contribute to their activity. On the one hand, mutagenesis studies identified a few receptor residues important for high bombesin affinity and on the other, a few studies focused on the relevance of diverse residues of the peptide for receptor activation. Models of the peptide bound to BB1R and BB2R can be helpful to improve our understanding of the stereochemical features granting bombesin activity. Accordingly, the present study describes the computational process followed to construct such models by means of Steered Molecular Dynamics, using models of the peptide and its receptors. Present results provide new insights into the structure-activity relationships of bombesin and its receptors, as well as render an explanation for the differential binding affinity observed towards BB1R and BB2R. Finally, these models can be further exploited to help for designing novel small molecule peptidomimetics with improved pharmacokinetics profile.
Collapse
Affiliation(s)
- Guillem Vila-Julià
- Department of Materials Science and Physical Chemistry, University of Barcelona and the Institut de Recerca en Quimica Teorica i Computacional (IQTCUB), Barcelona, Spain; Department of Chemical Engineering, Universitat Politecnica de Catalunya- Barcelona Tech., Av. Diagonal, 647, 08028 Barcelona, Spain
| | - Jaime Rubio-Martinez
- Department of Materials Science and Physical Chemistry, University of Barcelona and the Institut de Recerca en Quimica Teorica i Computacional (IQTCUB), Barcelona, Spain
| | - Juan J Perez
- Department of Chemical Engineering, Universitat Politecnica de Catalunya- Barcelona Tech., Av. Diagonal, 647, 08028 Barcelona, Spain.
| |
Collapse
|
10
|
Brosch-Lenz JF, Delker A, Schmidt F, Tran-Gia J. On the Use of Artificial Intelligence for Dosimetry of Radiopharmaceutical Therapies. Nuklearmedizin 2023; 62:379-388. [PMID: 37827503 DOI: 10.1055/a-2179-6872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Routine clinical dosimetry along with radiopharmaceutical therapies is key for future treatment personalization. However, dosimetry is considered complex and time-consuming with various challenges amongst the required steps within the dosimetry workflow. The general workflow for image-based dosimetry consists of quantitative imaging, the segmentation of organs and tumors, fitting of the time-activity-curves, and the conversion to absorbed dose. This work reviews the potential and advantages of the use of artificial intelligence to improve speed and accuracy of every single step of the dosimetry workflow.
Collapse
Affiliation(s)
| | - Astrid Delker
- Department of Nuclear Medicine, LMU University Hospital, Munich, Germany
| | - Fabian Schmidt
- Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital Tuebingen, Tuebingen, Germany
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Tuebingen, Germany
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
11
|
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: 42] [Impact Index Per Article: 21.0] [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
|
12
|
Brosch-Lenz J, Ke S, Wang H, Frey E, Dewaraja YK, Sunderland J, Uribe C. An International Study of Factors Affecting Variability of Dosimetry Calculations, Part 2: Overall Variabilities in Absorbed Dose. J Nucl Med 2023; 64:1109-1116. [PMID: 37024302 PMCID: PMC10315703 DOI: 10.2967/jnumed.122.265094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 04/08/2023] Open
Abstract
Dosimetry for personalized radiopharmaceutical therapy has gained considerable attention. Many methods, tools, and workflows have been developed to estimate absorbed dose (AD). However, standardization is still required to reduce variability of AD estimates across centers. One effort for standardization is the Society of Nuclear Medicine and Molecular Imaging 177Lu Dosimetry Challenge, which comprised 5 tasks (T1-T5) designed to assess dose estimate variability associated with the imaging protocol (T1 vs. T2 vs. T3), segmentation (T1 vs. T4), time integration (T4 vs. T5), and dose calculation (T5) steps of the dosimetry workflow. The aim of this work was to assess the overall variability in AD calculations for the different tasks. Methods: Anonymized datasets consisting of serial planar and quantitative SPECT/CT scans, organ and lesion contours, and time-integrated activity maps of 2 patients treated with 177Lu-DOTATATE were made available globally for participants to perform dosimetry calculations and submit their results in standardized submission spreadsheets. The data were carefully curated for formal mistakes and methodologic errors. General descriptive statistics for ADs were calculated, and statistical analysis was performed to compare the results of different tasks. Variability in ADs was measured using the quartile coefficient of dispersion. Results: ADs to organs estimated from planar imaging protocols (T2) were lower by about 60% than those from pure SPECT/CT (T1), and the differences were statistically significant. Importantly, the average differences in dose estimates when at least 1 SPECT/CT acquisition was available (T1, T3, T4, T5) were within ±10%, and the differences with respect to T1 were not statistically significant for most organs and lesions. When serial SPECT/CT images were used, the quartile coefficients of dispersion of ADs for organs and lesions were on average less than 20% and 26%, respectively, for T1; 20% and 18%, respectively, for T4 (segmentations provided); and 10% and 5%, respectively, for T5 (segmentation and time-integrated activity images provided). Conclusion: Variability in ADs was reduced as segmentation and time-integration data were provided to participants. Our results suggest that SPECT/CT-based imaging protocols generate more consistent and less variable results than planar imaging methods. Effort at standardizing segmentation and fitting should be made, as this may substantially reduce variability in ADs.
Collapse
Affiliation(s)
- Julia Brosch-Lenz
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Suqi Ke
- Division of Quantitative Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Division of Quantitative Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eric Frey
- Rapid, LLC, Baltimore, Maryland
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Yuni K Dewaraja
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - John Sunderland
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | - Carlos Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada;
- Department of Functional Imaging, BC Cancer, Vancouver, British Columbia, Canada; and
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
13
|
Nanostrategies for Therapeutic and Diagnostic Targeting of Gastrin-Releasing Peptide Receptor. Int J Mol Sci 2023; 24:ijms24043455. [PMID: 36834867 PMCID: PMC9958678 DOI: 10.3390/ijms24043455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Advances in nanomedicine bring the attention of researchers to the molecular targets that can play a major role in the development of novel therapeutic and diagnostic modalities for cancer management. The choice of a proper molecular target can decide the efficacy of the treatment and endorse the personalized medicine approach. Gastrin-releasing peptide receptor (GRPR) is a G-protein-coupled membrane receptor, well known to be overexpressed in numerous malignancies including pancreatic, prostate, breast, lung, colon, cervical, and gastrointestinal cancers. Therefore, many research groups express a deep interest in targeting GRPR with their nanoformulations. A broad spectrum of the GRPR ligands has been described in the literature, which allows tuning of the properties of the final formulation, particularly in the field of the ligand affinity to the receptor and internalization possibilities. Hereby, the recent advances in the field of applications of various nanoplatforms that are able to reach the GRPR-expressing cells are reviewed.
Collapse
|
14
|
Alhaj-Suliman SO, Wafa EI, Salem AK. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv Drug Deliv Rev 2022; 189:114482. [PMID: 35944587 DOI: 10.1016/j.addr.2022.114482] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Over the past two decades, multidisciplinary investigations into the development of nanoparticles for medical applications have continually increased. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has motivated the implementation of innovational modifications to a range of nanoparticle formulations designed for cancer imaging and/or cancer treatment to overcome specific barriers and shift the accumulation of payloads toward the diseased tissues. In recent years, novel technological and chemical approaches have been employed to modify or functionalize the surface of nanoparticles or manipulate the characteristics of nanoparticles. Combining these approaches with the identification of critical biomarkers provides new strategies for enhancing nanoparticle specificity for both cancer diagnostic and therapeutic applications. This review discusses the most recent advances in the design and engineering of nanoparticles as well as future directions for developing the next generation of nanomedicines.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, United States.
| |
Collapse
|
15
|
Desai P, Rimal R, Sahnoun SEM, Mottaghy FM, Möller M, Morgenroth A, Singh S. Radiolabeled Nanocarriers as Theranostics-Advancement from Peptides to Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200673. [PMID: 35527333 DOI: 10.1002/smll.202200673] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/15/2022] [Indexed: 06/14/2023]
Abstract
Endogenous targeted radiotherapy is emerging as an integral modality to treat a variety of cancer entities. Nevertheless, despite the positive clinical outcome of the treatment using radiolabeled peptides, small molecules, antibodies, and nanobodies, a high degree of hepatotoxicity and nephrotoxicity still persist. This limits the amount of dose that can be injected. In an attempt to mitigate these side effects, the use of nanocarriers such as nanoparticles (NPs), dendrimers, micelles, liposomes, and nanogels (NGs) is currently being explored. Nanocarriers can prolong circulation time and tumor retention, maximize radiation dosage, and offer multifunctionality for different targeting strategies. In this review, the authors first provide a summary of radiation therapy and imaging and discuss the new radiotracers that are used preclinically and clinically. They then highlight and identify the advantages of radio-nanomedicine and its potential in overcoming the limitations of endogenous radiotherapy. Finally, the review points to the ongoing efforts to maximize the use of radio-nanomedicine for efficient clinical translation.
Collapse
Affiliation(s)
- Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Rahul Rimal
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Sabri E M Sahnoun
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
- Department of Radiology and Nuclear Medicine, School for Cardiovascular Diseases (CARIM) and School of oncology (GROW), Maastricht University, Maastricht, 6229 HX, The Netherlands
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
| | - Smriti Singh
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
- Max-Planck-Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| |
Collapse
|
16
|
Calatayud DG, Neophytou S, Nicodemou E, Giuffrida SG, Ge H, Pascu SI. Nano-Theranostics for the Sensing, Imaging and Therapy of Prostate Cancers. Front Chem 2022; 10:830133. [PMID: 35494646 PMCID: PMC9039169 DOI: 10.3389/fchem.2022.830133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/16/2022] [Indexed: 01/28/2023] Open
Abstract
We highlight hereby recent developments in the emerging field of theranostics, which encompasses the combination of therapeutics and diagnostics in a single entity aimed for an early-stage diagnosis, image-guided therapy as well as evaluation of therapeutic outcomes of relevance to prostate cancer (PCa). Prostate cancer is one of the most common malignancies in men and a frequent cause of male cancer death. As such, this overview is concerned with recent developments in imaging and sensing of relevance to prostate cancer diagnosis and therapeutic monitoring. A major advantage for the effective treatment of PCa is an early diagnosis that would provide information for an appropriate treatment. Several imaging techniques are being developed to diagnose and monitor different stages of cancer in general, and patient stratification is particularly relevant for PCa. Hybrid imaging techniques applicable for diagnosis combine complementary structural and morphological information to enhance resolution and sensitivity of imaging. The focus of this review is to sum up some of the most recent advances in the nanotechnological approaches to the sensing and treatment of prostate cancer (PCa). Targeted imaging using nanoparticles, radiotracers and biomarkers could result to a more specialised and personalised diagnosis and treatment of PCa. A myriad of reports has been published literature proposing methods to detect and treat PCa using nanoparticles but the number of techniques approved for clinical use is relatively small. Another facet of this report is on reviewing aspects of the role of functional nanoparticles in multimodality imaging therapy considering recent developments in simultaneous PET-MRI (Positron Emission Tomography-Magnetic Resonance Imaging) coupled with optical imaging in vitro and in vivo, whilst highlighting feasible case studies that hold promise for the next generation of dual modality medical imaging of PCa. It is envisaged that progress in the field of imaging and sensing domains, taken together, could benefit from the biomedical implementation of new synthetic platforms such as metal complexes and functional materials supported on organic molecular species, which can be conjugated to targeting biomolecules and encompass adaptable and versatile molecular architectures. Furthermore, we include hereby an overview of aspects of biosensing methods aimed to tackle PCa: prostate biomarkers such as Prostate Specific Antigen (PSA) have been incorporated into synthetic platforms and explored in the context of sensing and imaging applications in preclinical investigations for the early detection of PCa. Finally, some of the societal concerns around nanotechnology being used for the detection of PCa are considered and addressed together with the concerns about the toxicity of nanoparticles–these were aspects of recent lively debates that currently hamper the clinical advancements of nano-theranostics. The publications survey conducted for this review includes, to the best of our knowledge, some of the most recent relevant literature examples from the state-of-the-art. Highlighting these advances would be of interest to the biomedical research community aiming to advance the application of theranostics particularly in PCa diagnosis and treatment, but also to those interested in the development of new probes and methodologies for the simultaneous imaging and therapy monitoring employed for PCa targeting.
Collapse
Affiliation(s)
- David G. Calatayud
- Department of Chemistry, University of Bath, Bath, United Kingdom
- Department of Electroceramics, Instituto de Ceramica y Vidrio - CSIC, Madrid, Spain
- *Correspondence: Sofia I. Pascu, ; David G. Calatayud,
| | - Sotia Neophytou
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Eleni Nicodemou
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | | | - Haobo Ge
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Sofia I. Pascu
- Department of Chemistry, University of Bath, Bath, United Kingdom
- Centre of Therapeutic Innovations, University of Bath, Bath, United Kingdom
- *Correspondence: Sofia I. Pascu, ; David G. Calatayud,
| |
Collapse
|
17
|
Moody TW, Lee L, Ramos-Alvarez I, Iordanskaia T, Mantey SA, Jensen RT. Bombesin Receptor Family Activation and CNS/Neural Tumors: Review of Evidence Supporting Possible Role for Novel Targeted Therapy. Front Endocrinol (Lausanne) 2021; 12:728088. [PMID: 34539578 PMCID: PMC8441013 DOI: 10.3389/fendo.2021.728088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are increasingly being considered as possible therapeutic targets in cancers. Activation of GPCR on tumors can have prominent growth effects, and GPCRs are frequently over-/ectopically expressed on tumors and thus can be used for targeted therapy. CNS/neural tumors are receiving increasing attention using this approach. Gliomas are the most frequent primary malignant brain/CNS tumor with glioblastoma having a 10-year survival <1%; neuroblastomas are the most common extracranial solid tumor in children with long-term survival<40%, and medulloblastomas are less common, but one subgroup has a 5-year survival <60%. Thus, there is an increased need for more effective treatments of these tumors. The Bombesin-receptor family (BnRs) is one of the GPCRs that are most frequently over/ectopically expressed by common tumors and is receiving particular attention as a possible therapeutic target in several tumors, particularly in prostate, breast, and lung cancer. We review in this paper evidence suggesting why a similar approach in some CNS/neural tumors (gliomas, neuroblastomas, medulloblastomas) should also be considered.
Collapse
Affiliation(s)
- Terry W. Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Training, Office of the Director, Bethesda, MD, United States
| | - Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Gastroenterology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tatiana Iordanskaia
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Samuel A. Mantey
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Robert T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
18
|
Dash BS, Lu YJ, Chen HA, Chuang CC, Chen JP. Magnetic and GRPR-targeted reduced graphene oxide/doxorubicin nanocomposite for dual-targeted chemo-photothermal cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112311. [PMID: 34474862 DOI: 10.1016/j.msec.2021.112311] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/17/2021] [Accepted: 07/04/2021] [Indexed: 02/06/2023]
Abstract
Herein, we design a rGO-based magnetic nanocomposite by decorating rGO with citrate-coated magnetic nanoparticles (CMNP). The magnetic rGO (mrGO) was modified by phospholipid-polyethylene glycol to prepare PEGylated mrGO, for conjugating with gastrin-releasing peptide receptor (GRPR)-binding peptide (mrGOG). The anticancer drug doxorubicin (DOX) was bound to mrGO (mrGOG) by π-π stacking for drug delivery triggered by the low pH value in the endosome. The mrGOG showed enhanced photothermal effect under NIR irradiation, endorsing its role for dual targeted DOX delivery. With efficient DOX release in the endosomal environment and heat generation from light absorption in the NIR range, mrGOG/DOX could be used for combination chemo-photothermal therapy after intracellular uptake by cancer cells. We characterized the physico-chemical as well as biological properties of the synthesized nanocomposites. The mrGOG is stable in biological buffer solution, showing high biocompatibility and minimum hemolytic properties. Using U87 glioblastoma cells, we confirmed the magnetic drug targeting effect in vitro for selective cancer cell killing. The peptide ligand-mediated targeted delivery increases the efficiency of intracellular uptake of both nanocomposite and DOX up to ~3 times due to the over-expressed GRPR on U87 surface, leading to higher cytotoxicity. The increased cytotoxicity using mrGOG over mrGO was shown from a decreased IC50 value (0.70 to 0.48 μg/mL) and an increased cell apoptosis rate (19.8% to 47.1%). The IC50 and apoptosis rate changed further to 0.19 μg/mL and 76.8% in combination with NIR laser irradiation, with the photothermal effect supported from upregulation of heat shock protein HSP70 expression. Using U87 tumor xenograft model created in nude mice, we demonstrated that magnetic guidance after intravenous delivery of mrGOG/DOX could significantly reduce tumor size and prolong animal survival over free DOX and non-magnetic guided groups. Augmented with NIR laser treatment for 5 min, the anti-cancer efficacy significantly improves with elevated cell apoptosis and reduced cell proliferation. Together with safety profiles from hematological as well as major organ histological analysis of treated animals, the mrGOG nanocomposite is an effective nanomaterial for combination chemo-photothermal cancer therapy.
Collapse
Affiliation(s)
- Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Huai-An Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Chi-Cheng Chuang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
19
|
Gupta N, Yadav V, Patel R. A brief review of the essential role of nanovehicles for improving the therapeutic efficacy of pharmacological agents against tumours. Curr Drug Deliv 2021; 19:301-316. [PMID: 34391379 DOI: 10.2174/1567201818666210813144105] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 11/22/2022]
Abstract
Cancer is the leading cause of death globally. There are several differences between cancer cells and normal cells. From all the therapies, chemotherapy is the most prominent therapy to treat cancer. However, the conventional drug delivery that is used to deliver poorly aqueous soluble chemotherapeutic agents has several obstacles such as whole-body distribution, rapid excretion, degradation before reaching the infected site, side effects, etc. Nanoformulation of these aqueous insoluble agents is the emerging delivery system for targeted and increasing solubility. Among all the three methods (physical, chemical and biological) chemical and biological methods are mostly used for the synthesis of nanovehicles (NVs) of different sizes, shapes and dimensions. A passive targeting delivery system in which NVs supports the pharmacological agents (drugs/genes) is a good way for resolving the obstacles with a conventional delivery system. It enhances the therapeutic efficacy of pharmacological agents (drugs/genes). These NVs have several specific characters like small size, large surface area to volume ratio, surface functionalization, etc. However, this delivery is not able to deliver site-specific delivery of drugs. An active targeting delivery system in which pharmacological agents are loaded on NVs to attack directly on cancer cells and tissues is a superior way for delivering the pharmacological agents compared to a passive targeting delivery system. Various targeting ligands have been investigated and applied for targeting the delivery of drugs such as sugar, vitamin, antibodies, protein, peptides, etc. These targeted ligand supports to guide the NVs accumulated directly on the cancer cells with a higher level of cellular internalization compared to passive targeting and conventional delivery system.
Collapse
Affiliation(s)
- Nitin Gupta
- School of Nano Sciences, Central University of Gujarat, Gandhinagar- 382030, Gujarat, India
| | - Virendra Yadav
- Department of Microbiology, School of Life Sciences, Jaipur National University, Jaipur- 341503, Rajasthan, India
| | - Rakesh Patel
- Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana- 384012, Gujarat, India
| |
Collapse
|
20
|
Pretze M, Reffert L, Diehl S, Schönberg SO, Wängler C, Hohenberger P, Wängler B. GMP-compliant production of [ 68Ga]Ga-NeoB for positron emission tomography imaging of patients with gastrointestinal stromal tumor. EJNMMI Radiopharm Chem 2021; 6:22. [PMID: 34228236 PMCID: PMC8260665 DOI: 10.1186/s41181-021-00137-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
Background [68Ga]Ga-NeoB is a novel DOTA-coupled Gastrin Releasing Peptide Receptor (GRPR) antagonist with high affinity for GRPR and good in vivo stability. This study aimed at (1) the translation of preclinical results to the clinics and establish the preparation of [68Ga]Ga-NeoB using a GMP conform kit approach and a licensed 68Ge/68Ga generator and (2) to explore the application of [68Ga]Ga-NeoB in patients with gastrointestinal stromal tumors (GIST) before and/or after interventional treatment (selective internal radiotherapy, irreversible electroporation, microwave ablation). Results Validation of the production and quality control of [68Ga]Ga-NeoB for patient use had to be performed before starting the GMP production. Six independent batches of [68Ga]Ga-NeoB were produced, all met the quality and sterility criteria and yielded 712 ± 73 MBq of the radiotracer in a radiochemical purity of > 95% and a molar activity of 14.2 ± 1.5 GBq/μmol within 20 min synthesis time and additional 20 min quality control. Three patients (2 females, 1 male, 51–77 yrs. of age) with progressive gastrointestinal stromal tumor metastases in the liver or peritoneum not responsive to standard tyrosine kinase inhibitor therapy underwent both [68Ga]Ga-NeoB scans prior and after interventional therapy. Radiosynthesis of 68Ga-NeoB was performed using a kit approach under GMP conditions. No specific patient preparation such as fasting or hydration was required for [68Ga]Ga-NeoB PET/CT imaging. Contrast-enhanced PET/CT studies were performed. A delayed, second abdominal image after the administration of the of [68Ga]Ga-NeoB was acquired at 120 min post injection. Conclusions A fully GMP compliant kit preparation of [68Ga]Ga-NeoB enabling the routine production of the tracer under GMP conditions was established for clinical routine PET/CT imaging of patients with metastatic GIST and proved to adequately visualize tumor deposits in the abdomen expressing GRPR. Patients could benefit from additional information derived from [68Ga]Ga-NeoB diagnosis to assess the presence of GRPR in the tumor tissue and monitor antitumor treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s41181-021-00137-w.
Collapse
Affiliation(s)
- Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany. .,Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Laura Reffert
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Steffen Diehl
- Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Mannheim, Germany
| | - Stefan O Schönberg
- Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Mannheim, Germany
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, Mannheim, Germany
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
21
|
Faviana P, Boldrini L, Erba PA, Di Stefano I, Manassero F, Bartoletti R, Galli L, Gentile C, Bardi M. Gastrin-Releasing Peptide Receptor in Low Grade Prostate Cancer: Can It Be a Better Predictor Than Prostate-Specific Membrane Antigen? Front Oncol 2021; 11:650249. [PMID: 33854977 PMCID: PMC8039448 DOI: 10.3389/fonc.2021.650249] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/05/2021] [Indexed: 01/22/2023] Open
Abstract
The aim of the present study was to evaluate whether prostate cancer (PC) patients can be accurately classified on the bases of tissue expression of gastrin-releasing peptide receptor (GRPR) and prostate-specific membrane antigen (PSMA). This retrospective study included 28 patients with PC. Formalin-fixed paraffin-embedded samples were used for diagnosis. Immunohistochemistry staining techniques were used to evaluate PSMA and GRPR expression (both number of cells expressed and % of area stained). To assess the independent associations among selected variables, a multi-dimensional scaling (MDS) analysis was used. It was found that the PSMA expression was inversely correlated with GRPR expression. Only the number of cells expressing GRPR was significantly related to the Gleason score. Both the percentage of area expressing GRPR and the number of cells expressing PSMA were close to reaching significance at the 0.05 level. MDS provided a map of the overall, independent association confirming that GRPR and PSMA represent inversely correlated measures of the same dimension. In conclusion, our data showed that GRPR expression should be evaluated in prostate biopsy specimens to improve our ability to detect PC with low grades at the earliest phases of development. Considering that GRPRs appear to be directly involved in the mechanisms of tumor proliferation, advancements in nuclear medicine radiotherapy can focus on this receptor to improve the therapeutic approach to PC. Further studies in our laboratory will investigate the molecular mechanisms of activation based on GRPR.
Collapse
Affiliation(s)
- Pinuccia Faviana
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Laura Boldrini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paola Anna Erba
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Iosè Di Stefano
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Francesca Manassero
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Riccardo Bartoletti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luca Galli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Carlo Gentile
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Massimo Bardi
- Department of Psychology and Behavioral Neuroscience, Randolph-Macon College, Ashland, VA, United States
| |
Collapse
|
22
|
Palmioli A, Nicolini G, Tripodi F, Orsato A, Ceresa C, Donzelli E, Arici M, Coccetti P, Rocchetti M, La Ferla B, Airoldi C. Targeting GRP receptor: Design, synthesis and preliminary biological characterization of new non-peptide antagonists of bombesin. Bioorg Chem 2021; 109:104739. [PMID: 33626451 DOI: 10.1016/j.bioorg.2021.104739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 01/29/2023]
Abstract
We report the rational design, synthesis, and in vitro preliminary evaluation of a new small library of non-peptide ligands of Gastrin Releasing Peptide Receptor (GRP-R), able to antagonize its natural ligand bombesin (BN) in the nanomolar range of concentration. GRP-R is a transmembrane G-protein coupled receptor promoting the stimulation of cancer cell proliferation. Being overexpressed on the surface of different human cancer cell lines, GRP-R is ideal for the selective delivery to tumor cells of both anticancer drug and diagnostic devices. What makes very challenging the design of non-peptide BN analogues is that the 3D structure of the GRP-R is not available, which is the case for many membrane-bound receptors. Thus, the design of GRP-R ligands has to be based on the structure of its natural ligands, BN and GRP. We recently mapped the BN binding epitope by NMR and here we exploited the same spectroscopy, combined with MD, to define BN conformation in proximity of biological membranes, where the interaction with GRP-R takes place. The gained structural information was used to identify a rigid C-galactosidic scaffold able to support pharmacophore groups mimicking the BN key residues' side chains in a suitable manner for binding to GRP-R. Our BN antagonists represent hit compounds for the rational design and synthesis of new ligands and modulators of GRP-R. The further optimization of the pharmacophore groups will allow to increase the biological activity. Due to their favorable chemical properties and stability, they could be employed for the active receptor-mediated targeting of GRP-R positive tumors.
Collapse
Affiliation(s)
- Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy; Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy
| | - Gabriella Nicolini
- Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy; School of Medicine and Surgery, Experimental Neurology Unit, University of Milano - Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | - Farida Tripodi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Alexandre Orsato
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy; Departamento de Química, CCE, Universidade Estadual de Londrina, CEP 86057-970 Londrina, Paraná, Brazil
| | - Cecilia Ceresa
- Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy; School of Medicine and Surgery, Experimental Neurology Unit, University of Milano - Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | - Elisabetta Donzelli
- Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy; School of Medicine and Surgery, Experimental Neurology Unit, University of Milano - Bicocca, Via Cadore 48, 20900 Monza, MB, Italy
| | - Martina Arici
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Paola Coccetti
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Marcella Rocchetti
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy.
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126 Milan, Italy; Milan Center for Neuroscience, University of Milano-Bicocca, P.zza dell'Ateneo Nuovo 1, 20126 Milano, Italy.
| |
Collapse
|
23
|
Ciaffaglione V, Waghorn PA, Exner RM, Cortezon-Tamarit F, Godfrey SP, Sarpaki S, Quilter H, Dondi R, Ge H, Kociok-Kohn G, Botchway SW, Eggleston IM, Dilworth JR, Pascu SI. Structural Investigations, Cellular Imaging, and Radiolabeling of Neutral, Polycationic, and Polyanionic Functional Metalloporphyrin Conjugates. Bioconjug Chem 2021; 32:1374-1392. [PMID: 33525868 PMCID: PMC8299459 DOI: 10.1021/acs.bioconjchem.0c00691] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Over the past decade,
porphyrin derivatives have emerged as invaluable
synthetic building blocks and theranostic kits for the delivery of
cellular fluorescence imaging and photodynamic therapy. Tetraphenylporphyrin
(TPP), its metal complexes, and related derivatives have been investigated
for their use as dyes in histology and as components of multimodal
imaging probes. The photophysical properties of porphyrin–metal
complexes featuring radiometals have been a focus of our attention
for the realization of fluorescence imaging probes coupled with radioimaging
capabilities and therapeutic potential having “true”
theranostic promise. We report hereby on the synthesis, radiochemistry,
structural investigations, and preliminary in vitro and in vivo uptake studies on a range of functionalized
porphyrin-based derivatives. In pursuit of developing new porphyrin-based
probes for multimodality imaging applications, we report new functionalized
neutral, polycationic, and polyanionic porphyrins incorporating nitroimidazole
and sulfonamide moieties, which were used as targeting groups to improve
the notoriously poor pharmacokinetics of porphyrin tags. The resulting
functional metalloporphyrin species were stable under serum challenges
and the nitroimidazole and sulfonamide derivatives remained fluorescent,
allowing in vitro confocal studies and visualization
of the lysosomal uptake in a gallium(III) sulfonamide derivative.
The molecular structures of selected porphyrin derivatives were determined
by single crystal X-ray diffraction using synchrotron radiation. We
also investigated the nature of the emission/excitation behavior of
model functional porphyrins using in silico approaches
such as TD DFT in simple solvation models. The conjugation of porphyrins
with the [7-13] and [7-14] fragments of bombesin was also achieved,
to provide targeting of the gastrin releasing peptide receptor (GRPR).
Depending on the metal, probe conjugates of relevance for single photon
emission computed tomography (SPECT) or positron emission tomography
(PET) probes have been designed and tested hereby, using TPP and related
functional free base porphyrins as the bifunctional chelator synthetic
scaffold and 111In[In] or 68Ga[Ga], respectively,
as the central metal ions. Interestingly, for simple porphyrin conjugates
good radiochemical incorporation was obtained for both radiometals,
but the presence of peptides significantly diminished the radio-incorporation
yields. Although the gallium-68 radiochemistry of the bombesin conjugates
did not show radiochemical incorporation suitable for in vivo studies, likely because the presence of the peptide changed the
behavior of the TPP-NH2 synthon taken alone, the optical
imaging assays indicated that the conjugated peptide tags do mediate
uptake of the porphyrin units into cells.
Collapse
Affiliation(s)
- Valeria Ciaffaglione
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Philip A Waghorn
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Rüdiger M Exner
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | | | - Samuel P Godfrey
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Sophia Sarpaki
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Helena Quilter
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom.,Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom.,Centre for Sustainable and Circular Technologies, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Ruggero Dondi
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Haobo Ge
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Gabriele Kociok-Kohn
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom.,Material and Chemical Characterisation (MC2), University of Bath, Bath, BA2 7AY, United Kingdom
| | - Stanley W Botchway
- Research Complex at Harwell, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0QX, United Kingdom
| | - Ian M Eggleston
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Jonathan R Dilworth
- Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Sofia I Pascu
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom.,Centre for Sustainable and Circular Technologies, University of Bath, Bath, BA2 7AY, United Kingdom
| |
Collapse
|
24
|
Preclinical Evaluation of the Copper-64 Labeled GRPR-Antagonist RM26 in Comparison with the Cobalt-55 Labeled Counterpart for PET-Imaging of Prostate Cancer. Molecules 2020; 25:molecules25245993. [PMID: 33352838 PMCID: PMC7766840 DOI: 10.3390/molecules25245993] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 01/21/2023] Open
Abstract
Gastrin-releasing peptide receptor (GRPR) is overexpressed in the majority of prostate cancers. This study aimed to investigate the potential of 64Cu (radionuclide for late time-point PET-imaging) for imaging of GRPR expression using NOTA-PEG2-RM26 and NODAGA-PEG2-RM26. Methods: NOTA/NODAGA-PEG2-RM26 were labeled with 64Cu and evaluated in GRPR-expressing PC-3 cells. Biodistribution of [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 was studied in PC-3 xenografted mice and compared to the biodistribution of [57Co]Co-NOTA/NODAGA-PEG2-RM26 at 3 and 24 h p.i. Preclinical PET/CT imaging was performed in tumor-bearing mice. NOTA/NODAGA-PEG2-RM26 were stably labeled with 64Cu with quantitative yields. In vitro, binding of [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 was rapid and GRPR-specific with slow internalization. In vivo, [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 bound specifically to GRPR-expressing tumors with fast clearance from blood and normal organs and displayed generally comparable biodistribution profiles to [57Co]Co-NOTA/NODAGA-PEG2-RM26; tumor uptake exceeded normal tissue uptake 3 h p.i.. Tumor-to-organ ratios did not increase significantly with time. [64Cu]Cu-NOTA-PEG2-RM26 had a significantly higher liver and pancreas uptake compared to other agents. 57Co-labeled radioconjugates showed overall higher tumor-to-non-tumor ratios, compared to the 64Cu-labeled counterparts. [64Cu]Cu-NOTA/NODAGA-PEG2-RM26 was able to visualize GRPR-expression in a murine PC model using PET. However, [55/57Co]Co-NOTA/NODAGA-PEG2-RM26 provided better in vivo stability and overall higher tumor-to-non-tumor ratios compared with the 64Cu-labeled conjugates.
Collapse
|
25
|
Sgouros G, Bodei L, McDevitt MR, Nedrow JR. Radiopharmaceutical therapy in cancer: clinical advances and challenges. Nat Rev Drug Discov 2020; 19:589-608. [PMID: 32728208 PMCID: PMC7390460 DOI: 10.1038/s41573-020-0073-9] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2020] [Indexed: 12/25/2022]
Abstract
Radiopharmaceutical therapy (RPT) is emerging as a safe and effective targeted approach to treating many types of cancer. In RPT, radiation is systemically or locally delivered using pharmaceuticals that either bind preferentially to cancer cells or accumulate by physiological mechanisms. Almost all radionuclides used in RPT emit photons that can be imaged, enabling non-invasive visualization of the biodistribution of the therapeutic agent. Compared with almost all other systemic cancer treatment options, RPT has shown efficacy with minimal toxicity. With the recent FDA approval of several RPT agents, the remarkable potential of this treatment is now being recognized. This Review covers the fundamental properties, clinical development and associated challenges of RPT.
Collapse
Affiliation(s)
- George Sgouros
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jessie R Nedrow
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Current State of Radiolabeled Heterobivalent Peptidic Ligands in Tumor Imaging and Therapy. Pharmaceuticals (Basel) 2020; 13:ph13080173. [PMID: 32751666 PMCID: PMC7465997 DOI: 10.3390/ph13080173] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Over the past few years, an approach emerged that combines different receptor-specific peptide radioligands able to bind different target structures on tumor cells concomitantly or separately. The reason for the growing interest in this special field of radiopharmaceutical development is rooted in the fact that bispecific peptide heterodimers can exhibit a strongly increased target cell avidity and specificity compared to their corresponding monospecific counterparts by being able to bind to two different target structures that are overexpressed on the cell surface of several malignancies. This increase of avidity is most pronounced in the case of concomitant binding of both peptides to their respective targets but is also observed in cases of heterogeneously expressed receptors within a tumor entity. Furthermore, the application of a radiolabeled heterobivalent agent can solve the ubiquitous problem of limited tumor visualization sensitivity caused by differential receptor expression on different tumor lesions. In this article, the concept of heterobivalent targeting and the general advantages of using radiolabeled bispecific peptidic ligands for tumor imaging or therapy as well as the influence of molecular design and the receptors on the tumor cell surface are explained, and an overview is given of the radiolabeled heterobivalent peptides described thus far.
Collapse
|
27
|
Kurmi BD, Patel P, Paliwal R, Paliwal SR. Molecular approaches for targeted drug delivery towards cancer: A concise review with respect to nanotechnology. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
Hoppenz P, Els-Heindl S, Kellert M, Kuhnert R, Saretz S, Lerchen HG, Köbberling J, Riedl B, Hey-Hawkins E, Beck-Sickinger AG. A Selective Carborane-Functionalized Gastrin-Releasing Peptide Receptor Agonist as Boron Delivery Agent for Boron Neutron Capture Therapy. J Org Chem 2019; 85:1446-1457. [PMID: 31813224 DOI: 10.1021/acs.joc.9b02406] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Boron neutron capture therapy (BNCT) allows the selective elimination of malignant tumor cells without affecting healthy tissue. Although this binary radiotherapy approach has been known for decades, BNCT failed to reach the daily clinics to date. One of the reasons is the lack of selective boron delivery agents. Using boron loaded peptide conjugates, which address G protein-coupled receptors overexpressed on tumor cells allow the intracellular accumulation of boron. The gastrin-releasing peptide receptor (GRPR) is a well-known target in cancer diagnosis and can potentially be used for BNCT. Here, we present the successful introduction of multiple bis-deoxygalactosyl-carborane building blocks to the GRPR-selective ligand [d-Phe6, β-Ala11, Ala13, Nle14]Bn(6-14) (sBB2L) generating peptide conjugates with up to 80 boron atoms per molecule. Receptor activation was retained, metabolic stability was increased, and uptake into PC3 cells was proven without showing any intrinsic cytotoxicity. Furthermore, undesired uptake into liver cells was suppressed by using l-deoxygalactosyl modified carborane building blocks. Due to its high boron loading and excellent GRPR selectivity, this conjugate can be considered as a promising boron delivery agent for BNCT.
Collapse
Affiliation(s)
- Paul Hoppenz
- Institute of Biochemistry, Faculty of Life Sciences , Leipzig University , Brüderstrasse 34 , 04103 Leipzig , Germany
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Faculty of Life Sciences , 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 , Wuppertal , Germany
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry , Leipzig University , Johannisallee 29 , 04103 Leipzig , Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences , Leipzig University , Brüderstrasse 34 , 04103 Leipzig , Germany
| |
Collapse
|
29
|
Hoppenz P, Els‐Heindl S, Beck‐Sickinger AG. Identification and stabilization of a highly selective gastrin‐releasing peptide receptor agonist. J Pept Sci 2019; 25:e3224. [DOI: 10.1002/psc.3224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Paul Hoppenz
- Institute of BiochemistryLeipzig University Leipzig Germany
| | | | | |
Collapse
|