1
|
Jülke EM, Beck-Sickinger AG. Peptide therapeutics: current status and future opportunity with focus on nose-to-brain delivery☆. Peptides 2025; 188:171404. [PMID: 40222598 DOI: 10.1016/j.peptides.2025.171404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Peptide drugs are a highly diverse group of therapeutic agents. Over the last decade, more than 40 peptides have been approved for clinical use. They target different structures, ranging from G protein-coupled receptors (GPCRs) to pathogens and are used to treat a variety of indications, including metabolic disorders, genetic diseases, acute illnesses and more. Structurally, peptide therapeutics are a heterogeneous class. This diversity allows them to bridge the gap between small molecules and biologics. However, limited metabolic stability and bioavailability must be addressed. Strategies to improve the half-life include backbone and sequence modification, cyclization and the addition of stabilizing moieties. Great strides have been made in recent years towards achieving sufficient drug uptake for oral application have been achieved within recent years. However, these methods require specialized peptide design or involve permeabilization of the gastrointestinal tract. Consequently, other routes of administration are being explored. One promising approach is the nasal application of peptides. This method can be used for systemic uptake, but also allows for direct nose-to-brain delivery of compounds. While successful nose-to-brain delivery is already used in the clinic, the underlining mechanisms are poorly understood. Strategies for rational optimization are needed to make this method more applicable to a wider range of compounds. Overall, approved peptide therapeutics cover a wide range of applications and have demonstrated a growing and novel potential in recent drug discovery.
Collapse
Affiliation(s)
- Eva-Maria Jülke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, Leipzig 04103, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, Leipzig 04103, Germany.
| |
Collapse
|
2
|
Tomić K, Kostevšek N, Romeo S, Bassanini I. Neuropeptide Y receptors 1 and 2 as molecular targets in prostate and breast cancer therapy. Biomed Pharmacother 2025; 187:118117. [PMID: 40319656 DOI: 10.1016/j.biopha.2025.118117] [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: 02/24/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025] Open
Abstract
Recent advances have revealed the overexpression of Neuropeptide Y (NPY) receptors in multiple cancers, positioning them as attractive molecular targets for cancer diagnostics and therapeutics. Despite this, a comprehensive roadmap for the rational development of anticancer agents targeting NPY receptors remains lacking. Therefore, we present the characteristics of NPY receptor subtypes, their abundance, and the correlation of their expression in different cancer types. It was found that NPY receptor subtypes 1 and 2 were extensively studied, especially in connection with breast and prostate cancer. Many tumors express NPYR, but only breast cancer tissue shows a significant difference in NPYR subtype expression levels between tumor and normal tissues, and, therefore, can represent a promising target. In the context of anticancer therapy, this review provides key findings from the use of wild-type and synthetic NPY analogs. We highlight the critical residues in the NPY sequence that play a critical role in interactions with receptors and provide the recent literature findings on NPY analogues as efficient and specific cancer-targeting agents. Potential solutions to improve NPY analogs' stability are provided, such as sequence modifications of linear peptides, peptide stapling, and conjugation for drug delivery systems. In general, NPY treatment can not be used efficiently as a single therapy but as a combinatorial therapy with anticancer drugs to improve the specificity of the treatment via high-affinity binding to the cancer cells and sensitizing them to chemotherapy.
Collapse
Affiliation(s)
- Katarina Tomić
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova 39, Ljubljana 1000, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana 1000, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova 39, Ljubljana 1000, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana 1000, Slovenia.
| | - Sergio Romeo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, Milan, Italy; Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Via Mario Bianco 9, Milan, Italy
| | - Ivan Bassanini
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Via Mario Bianco 9, Milan, Italy
| |
Collapse
|
3
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Paolino M, Saletti M, Venditti J, Zacchei A, Donati A, Bonechi C, Giuliani G, Lamponi S, Cappelli A. Synthesis and Reactivity of Oligo(ethylene glycol)-Tethered Morita-Baylis-Hillman Dimers in the Formation of Macrocyclic Structures Showing Remarkable Cytotoxicity. Pharmaceuticals (Basel) 2025; 18:473. [PMID: 40283910 PMCID: PMC12030125 DOI: 10.3390/ph18040473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Crown ethers have received increasing interest owing to their ability to form stable complexes with cations. This molecular feature has been successfully exploited in the development of biologically relevant ionophores. Methods: In order to obtain innovative crown ethers derivatives, a Morita-Baylis-Hillman adduct (MBHA) acetate (4) bearing a phenylacetylene moiety was dimerized via the click-chemistry CuAAC reaction with oligo(ethylene glycol) diazide derivatives to build-up a small series of dimeric MBHA derivatives (5a-d). These dimeric MBHA derivatives were reacted with n-butylamine to afford tunable macrocyclic crown ether-paracyclophane hybrid architectures (6a-d). Results: Compounds (E,Z)-6a, (E,E)-6a, 6b-d showed, in human breast cancer MDA-MB-231 and human melanoma A375 cells, IC50 values comparable with those of reference anticancer agent Doxorubicin. Conclusions: This exploration approach provides original new macrocyclic architectures potentially useful as anticancer agents.
Collapse
Affiliation(s)
- Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy (A.C.)
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Zhang J, Kong X, Chen X. Development of Novel Peptides That Target the Ninjurin 1 and 2 Pathways to Inhibit Cell Growth and Survival via p53. Cells 2025; 14:401. [PMID: 40136650 PMCID: PMC11941050 DOI: 10.3390/cells14060401] [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: 02/05/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Ninjurin 1 and 2 (NINJ1, NINJ2) belong to the homophilic cell adhesion family and play significant roles in cellular communication and tissue development. While both NINJ1 and NINJ2 are found to be over-expressed in several types of cancers, it remains unclear whether they can be targeted for cancer treatment. In this study, we aimed to develop NINJ1/2 peptides derived from the N-terminal extracellular domain that can elicit growth suppression and thus possess therapeutic potentials. We found that peptide NINJ1-A, which is derived from the N-terminal adhesion motif of NINJ1, was able to inhibit cell growth in a NINJ1- or p53-dependent manner. Similarly, peptide NINJ2-A, which is derived from the N-terminal adhesion motif of NINJ2, was able to inhibit cell growth in a NINJ2- or p53-dependent manner. We also found that NINJ1 and NINJ2 physically interact via their respective N-terminal domains. Interestingly, NINJ1-B and NINJ2-B peptides, which were derived from the N-terminal amphipathic helix domains of NINJ1 and NINJ2, respectively, were able to disrupt NINJ1-NINJ2 interaction and inhibit cell growth in a NINJ1/NINJ2-dependent manner. Notably, NINJ1-B and NINJ2-B peptides demonstrated greater potency in growth suppression than NINJ1-A and NINJ2-A peptides, respectively. Mechanistically, we found that NINJ1-B and NINJ2-B peptides were able to induce p53 expression and suppress cell growth in a p53-dependent manner. Together, our findings provide valuable insights into the development of NINJ1/NINJ2 peptides as potential cancer therapeutics, particularly for cancers harboring wild-type p53.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616, USA;
| | | | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616, USA;
| |
Collapse
|
6
|
Xiao W, Jiang W, Chen Z, Huang Y, Mao J, Zheng W, Hu Y, Shi J. Advance in peptide-based drug development: delivery platforms, therapeutics and vaccines. Signal Transduct Target Ther 2025; 10:74. [PMID: 40038239 PMCID: PMC11880366 DOI: 10.1038/s41392-024-02107-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 11/01/2024] [Accepted: 12/13/2024] [Indexed: 03/06/2025] Open
Abstract
The successful approval of peptide-based drugs can be attributed to a collaborative effort across multiple disciplines. The integration of novel drug design and synthesis techniques, display library technology, delivery systems, bioengineering advancements, and artificial intelligence have significantly expedited the development of groundbreaking peptide-based drugs, effectively addressing the obstacles associated with their character, such as the rapid clearance and degradation, necessitating subcutaneous injection leading to increasing patient discomfort, and ultimately advancing translational research efforts. Peptides are presently employed in the management and diagnosis of a diverse array of medical conditions, such as diabetes mellitus, weight loss, oncology, and rare diseases, and are additionally garnering interest in facilitating targeted drug delivery platforms and the advancement of peptide-based vaccines. This paper provides an overview of the present market and clinical trial progress of peptide-based therapeutics, delivery platforms, and vaccines. It examines the key areas of research in peptide-based drug development through a literature analysis and emphasizes the structural modification principles of peptide-based drugs, as well as the recent advancements in screening, design, and delivery technologies. The accelerated advancement in the development of novel peptide-based therapeutics, including peptide-drug complexes, new peptide-based vaccines, and innovative peptide-based diagnostic reagents, has the potential to promote the era of precise customization of disease therapeutic schedule.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wenjie Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yu Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Junyi Mao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wei Zheng
- Department of Integrative Medicine, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yonghe Hu
- School of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
7
|
Sagar B, Gupta S, Verma SK, Reddy YVM, Shukla S. Navigating cancer therapy: Harnessing the power of peptide-drug conjugates as precision delivery vehicles. Eur J Med Chem 2025; 283:117131. [PMID: 39647418 DOI: 10.1016/j.ejmech.2024.117131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Cancer treatment is a formidable challenge due to the adverse effects associated with non-selective therapies like chemotherapy and radiotherapy. This review article primarily centers on the application of Peptide-Drug Conjugates (PDCs) for delivering cancer treatment. PDCs represent a promising class of precision medicines, harnessing the unique attributes of peptides in conjunction with non-peptide components. The covalent linking of peptides and drugs through specialized connectors characterizes PDCs. These constructs play a pivotal role in delivering drugs directly to tumor sites with high precision. PDCs encompass three pivotal components: a targeting ligand, a cytotoxic ligand, and a carefully chosen linker. The selection of these elements is crucial to maximize the efficiency of PDCs. PDCs offer a multitude of advantages over conventional drug molecules, including enhanced specificity, reduced off-target effects, and an improved therapeutic profile. The peptide component within PDCs can be customized to specifically adhere to disease-specific receptors or biomarkers, facilitating targeted drug delivery and accumulation in afflicted cells or tissues. This targeted approach enables the controlled release of therapeutic payloads at the localized site, resulting in heightened effectiveness and minimized systemic toxicity. Diverse linker strategies are employed to ensure the stable connection between the peptide and non-peptide components, ensuring controlled drug release at the desired location of action. The peptides utilized in these treatments encompass cell-penetrating peptides, peptides designed to target tumor cells, and those aimed at the nucleus of cancer cells. While certain clinical trials have been conducted, and some PDCs are currently in use for cancer treatment, it's essential to acknowledge that PDCs have their limitations, such as low stability in plasma, fast elimination and limited oral bioavailability. Ongoing research endeavors seek to surmount these challenges and further establish PDCs as potent agents for cancer treatment. This review sheds light on recent advancements in the design, delivery, and applications of PDCs, while also highlighting the prevailing challenges and charting a path for future research directions.
Collapse
Affiliation(s)
- Bulbul Sagar
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarthak Gupta
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarvesh Kumar Verma
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, 302017, Rajasthan, India
| | | | - Shefali Shukla
- Sri Venkateswara College, University of Delhi, New Delhi, India.
| |
Collapse
|
8
|
Chavda VP, Bojarska J. Peptides on patrol: Carrier systems for targeted delivery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 212:129-161. [PMID: 40122644 DOI: 10.1016/bs.pmbts.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The peptide is a small unit of protein that exhibits a diverse range of therapeutic applications, including but not limited to respiratory, inflammatory, oncologic, metabolic and neurological disorders. Peptides also play a significant role in signal transduction in cells. This chapter focuses on the delivery of peptides through the utilization of various carrier molecules, including liposomes, micelles, polymeric nanoparticles, and inorganic materials. These carriers facilitate targeted delivery and site-specific delivery of peptides. Different nanocarriers and therapeutic drug molecules also help with the delivery of peptides. Application to various diseases and different routes of delivery are described in this manuscript, along with current limitations and future prospects.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India.
| | - Joanna Bojarska
- Chemistry Department, Institute of Ecological and Inorganic Chemistry, Technical, University of Lodz, Zeromskiego St., Lodz, Poland
| |
Collapse
|
9
|
Gare CL, Palombi IR, White AM, Chavchich M, Edstein MD, Lock A, Avery VM, Craik DJ, McMorran BJ, Lawrence N, Malins LR. Exploring the Utility of Cell-Penetrating Peptides as Vehicles for the Delivery of Distinct Antimalarial Drug Cargoes. ChemMedChem 2025; 20:e202400637. [PMID: 39379289 DOI: 10.1002/cmdc.202400637] [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: 08/16/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
The devastating impact of malaria includes significant mortality and illness worldwide. Increasing resistance of the causative parasite, Plasmodium, to existing antimalarial drugs underscores a need for additional compounds with distinct modes of action in the therapeutic development pipeline. Here we showcase peptide-drug conjugates (PDCs) as an attractive compound class, in which therapeutic or lead antimalarials are chemically conjugated to cell-penetrating peptides. This approach aims to enhance selective uptake into Plasmodium-infected red blood cells and impart additional cytotoxic actions on the intraerythrocytic parasite, thereby enabling targeted drug delivery and dual modes of action. We describe the development of PDCs featuring four compounds with antimalarial activity-primaquine, artesunate, tafenoquine and methotrexate-conjugated to three cell-penetrating peptide scaffolds with varied antiplasmodial activity, including active and inactive analogues of platelet factor 4 derived internalization peptide (PDIP), and a cyclic polyarginine peptide. Development of this diverse set of PDCs featured distinct and adaptable conjugation strategies, to produce conjugates with in vitro antiplasmodial activities ranging from low nanomolar to low micromolar potencies according to the drug cargo and bioactivity of the partner peptide. Overall, this study establishes a strategic and methodological framework for the further development of dual mode of action peptide-drug antimalarial therapeutics.
Collapse
Affiliation(s)
- Caitlin L Gare
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| | - Isabella R Palombi
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| | - Andrew M White
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| | - Marina Chavchich
- Department of Drug Evaluation, Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, QLD, 4051, Australia
| | - Michael D Edstein
- Department of Drug Evaluation, Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, QLD, 4051, Australia
| | - Aaron Lock
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
| | - Vicky M Avery
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Brendan J McMorran
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - Nicole Lawrence
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Lara R Malins
- Research School of Chemistry, The Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
10
|
Skalickova M, Hadrava Vanova K, Uher O, Leischner Fialova J, Petrlakova K, Masarik M, Kejík Z, Martasek P, Pacak K, Jakubek M. Injecting hope: the potential of intratumoral immunotherapy for locally advanced and metastatic cancer. Front Immunol 2025; 15:1479483. [PMID: 39850897 PMCID: PMC11754201 DOI: 10.3389/fimmu.2024.1479483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Despite enormous progress, advanced cancers are still one of the most serious medical problems in current society. Although various agents and therapeutic strategies with anticancer activity are known and used, they often fail to achieve satisfactory long-term patient outcomes and survival. Recently, immunotherapy has shown success in patients by harnessing important interactions between the immune system and cancer. However, many of these therapies lead to frequent side effects when administered systemically, prompting treatment modifications or discontinuation or, in severe cases, fatalities. New therapeutic approaches like intratumoral immunotherapy, characterized by reduced side effects, cost, and systemic toxicity, offer promising prospects for future applications in clinical oncology. In the context of locally advanced or metastatic cancer, combining diverse immunotherapeutic and other treatment strategies targeting multiple cancer hallmarks appears crucial. Such combination therapies hold promise for improving patient outcomes and survival and for promoting a sustained systemic response. This review aims to provide a current overview of immunotherapeutic approaches, specifically focusing on the intratumoral administration of drugs in patients with locally advanced and metastatic cancers. It also explores the integration of intratumoral administration with other modalities to maximize therapeutic response. Additionally, the review summarizes recent advances in intratumoral immunotherapy and discusses novel therapeutic approaches, outlining future directions in the field.
Collapse
Affiliation(s)
- Marketa Skalickova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Katerina Hadrava Vanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jindriska Leischner Fialova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katerina Petrlakova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Michal Masarik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Pavel Martasek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
11
|
Gutierrez S, Boada MD. NK1 receptor blockade disrupts microtumor growth and aggregation in a three-dimensional murine breast cancer model. Neuropeptides 2025; 109:102479. [PMID: 39591909 DOI: 10.1016/j.npep.2024.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 11/28/2024]
Abstract
Several data indicate that Substance P (SP) neurokinin type 1 receptor (NK1R) is at the center of the interaction between cancer cells and peripheral sensory neurons. Selecting the appropriate cancer cell line and its susceptibility to being modulated by NK1 antagonists are critical to studying this complex interaction. In the current study, we have focused on this selection by comparing several aspects of the triple-negative breast cancer (TNBC) cell line (MDA-MB-231LUC+) with a modified murine cell line (E0771LUC+), both expressing luciferase. This comparison was made using several methods, SP stimulation and 3D cell culture models, to better reproduce the heterogenous microenvironment of solid tumors observed in vivo. Furthermore, the susceptibility of the murine cell line (E0771LUC+) to NK1R antagonist (Aprepitant) was tested. Our results indicate that E0771LUC+ recapitulates several essential aspects of the human cell line, rendering this murine line ideal to be used on immune-competent animals during in vivo studies. We have also found that both cell lines are susceptible to SP stimulation, and their proliferation is disrupted by NK1R antagonists (Aprepitant). In vivo studies are required to verify and refine these findings.
Collapse
Affiliation(s)
- Silvia Gutierrez
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - M Danilo Boada
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
12
|
Yang Q, Hu Z, Jiang H, Wang J, Han H, Shi W, Qian H. Recent advances, strategies, and future perspectives of peptide-based drugs in clinical applications. Chin J Nat Med 2025; 23:31-42. [PMID: 39855829 DOI: 10.1016/s1875-5364(25)60800-4] [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: 07/07/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 01/27/2025]
Abstract
Peptide-based therapies have attracted considerable interest in the treatment of cancer, diabetes, bacterial infections, and neurodegenerative diseases due to their promising therapeutic properties and enhanced safety profiles. This review provides a comprehensive overview of the major trends in peptide drug discovery and development, emphasizing preclinical strategies aimed at improving peptide stability, specificity, and pharmacokinetic properties. It assesses the current applications and challenges of peptide-based drugs in these diseases, illustrating the pharmaceutical areas where peptide-based drugs demonstrate significant potential. Furthermore, this review analyzes the obstacles that must be overcome in the future, aiming to provide valuable insights and references for the continued advancement of peptide-based drugs.
Collapse
Affiliation(s)
- Qimeng Yang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhipeng Hu
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hongyu Jiang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jialing Wang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Han Han
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Wei Shi
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Hai Qian
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
13
|
Palombi IR, White AM, Koda Y, Craik DJ, Lawrence N, Malins LR. Synthesis and Investigation of Peptide-Drug Conjugates Comprising Camptothecin and a Human Protein-Derived Cell-Penetrating Peptide. Chem Biol Drug Des 2025; 105:e70051. [PMID: 39834140 PMCID: PMC11747586 DOI: 10.1111/cbdd.70051] [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: 10/04/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Drug targeting strategies, such as peptide-drug conjugates (PDCs), have arisen to combat the issue of off-target toxicity that is commonly associated with chemotherapeutic small molecule drugs. Here we investigated the ability of PDCs comprising a human protein-derived cell-penetrating peptide-platelet factor 4-derived internalization peptide (PDIP)-as a targeting strategy to improve the selectivity of camptothecin (CPT), a topoisomerase I inhibitor that suffers from off-target toxicity. The intranuclear target of CPT allowed exploration of PDC design features required for optimal potency. A suite of PDCs with various structural characteristics, including alternative conjugation strategies (such as azide-alkyne cycloaddition and disulfide conjugation) and linker types (non-cleavable or cleavable), were synthesized and investigated for their anticancer activity. Membrane permeability and cytotoxicity studies revealed that intact PDIP-CPT PDCs can cross membranes, and that PDCs with disulfide- and protease-cleavable linkers liberated free CPT and killed melanoma cells with nanomolar potency. However, selectivity of the PDIP carrier peptide for melanoma compared to noncancerous epidermal cells was not maintained for the PDCs. This study emphasizes the distinct role of the peptide, linker, and drug for optimal PDC activity and highlights the need to carefully match components when assembling PDCs as targeted therapies.
Collapse
Affiliation(s)
- Isabella R. Palombi
- Research School of ChemistryAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Andrew M. White
- Research School of ChemistryAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Yasuko Koda
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQueenslandAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQueenslandAustralia
| | - David J. Craik
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQueenslandAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQueenslandAustralia
| | - Nicole Lawrence
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQueenslandAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQueenslandAustralia
| | - Lara R. Malins
- Research School of ChemistryAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
| |
Collapse
|
14
|
Bazylevich A, Miller A, Tkachenko I, Merlani M, Patsenker L, Gellerman G, Lubin BCR. Novel Cyclic Peptide-Drug Conjugate P6-SN38 Toward Targeted Treatment of EGFR Overexpressed Non-Small Cell Lung Cancer. Pharmaceutics 2024; 16:1613. [PMID: 39771591 PMCID: PMC11676734 DOI: 10.3390/pharmaceutics16121613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Here, we report on the synthesis and biological evaluation of a novel peptide-drug conjugate, P6-SN38, which consists of the EGFR-specific short cyclic peptide, P6, and the Topo I inhibitor SN38, which is a bioactive metabolite of the anticancer drug irinotecan. Methods: SN38 is attached to the peptide at position 20 of the E ring's tertiary hydroxyl group via a mono-succinate linker. Results: The developed peptide-drug conjugate (PDC) exhibited sub-micromolar anticancer activity on EGFR-positive (EGFR+) cell lines but no effect on EGFR-negative (EGFR-) cells. In vivo studies have shown that this PDC specifically accumulates in EGFR+ non-small cell lung cancer (NSCLC) xenografts and presents superior anticancer activity compared to the EGFR-specific antibody cetuximab (ErbituxTM) and free SN38. The 10 mg/kg dose of P6-SN38 in a side-by-side EGFR+/EGFR- xenograft shows eradication of the EGFR+ tumor with good tolerance, but no inhibition of tumor growth of the EGFR- counterpart. Conclusions: The PDC examined in this study was proven to be highly efficient for NSCLC, broadening its utilization for targeted cancer therapy in EGFR overexpressed cancers.
Collapse
Affiliation(s)
- Andrii Bazylevich
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.B.); (I.T.); (L.P.); (G.G.)
| | - Ayala Miller
- Agriculture and Oenology Department, Eastern Regional R&D Center, Ariel 40700, Israel
| | - Iryna Tkachenko
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.B.); (I.T.); (L.P.); (G.G.)
| | - Maia Merlani
- I. Kutateladze Institute of Pharmacochemistry, Tbilisi State Medical University (TSMU), Vashlijvari 0159, Georgia
| | - Leonid Patsenker
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.B.); (I.T.); (L.P.); (G.G.)
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel; (A.B.); (I.T.); (L.P.); (G.G.)
| | - Bat Chen R. Lubin
- Department of Chemical Engineering, Biotechnology and Materials, Ariel University, Ariel 40700, Israel
- Agriculture and Oenology Department, Eastern Regional R&D Center, Ariel 40700, Israel
| |
Collapse
|
15
|
Lin Z, Assaraf YG, Kwok HF. Peptides for microbe-induced cancers: latest therapeutic strategies and their advanced technologies. Cancer Metastasis Rev 2024; 43:1315-1336. [PMID: 39008152 DOI: 10.1007/s10555-024-10197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
Cancer is a significant global health concern associated with multiple distinct factors, including microbial and viral infections. Numerous studies have elucidated the role of microorganisms, such as Helicobacter pylori (H. pylori), as well as viruses for example human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV), in the development of human malignancies. Substantial attention has been focused on the treatment of these microorganism- and virus-associated cancers, with promising outcomes observed in studies employing peptide-based therapies. The current paper provides an overview of microbe- and virus-induced cancers and their underlying molecular mechanisms. We discuss an assortment of peptide-based therapies which are currently being developed, including tumor-targeting peptides and microbial/viral peptide-based vaccines. We describe the major technological advancements that have been made in the design, screening, and delivery of peptides as anticancer agents. The primary focus of the current review is to provide insight into the latest research and development in this field and to provide a realistic glimpse into the future of peptide-based therapies for microbe- and virus-induced neoplasms.
Collapse
Affiliation(s)
- Ziqi Lin
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Instituteof Technology, Haifa, 3200003, Israel
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
16
|
Lamb HO, Benfield AH, Henriques ST. Peptides as innovative strategies to combat drug resistance in cancer therapy. Drug Discov Today 2024; 29:104206. [PMID: 39395530 DOI: 10.1016/j.drudis.2024.104206] [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: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
Drug resistance is the leading cause of treatment failure in patients with cancer. Thus, innovative therapeutic strategies are required to overcome this critical challenge and improve patient outcomes. In this review, we examine the potential of peptide-based therapies to combat drug resistance in cancer. We highlight the unique strategies and mechanisms that can be explored by using peptides, including their ability to selectively target tumours, facilitate drug delivery into cancer cells, and inhibit key intracellular proteins that drive cancer progression and resistance. Peptides offer a promising approach to overcoming both intrinsic and adaptative cancer resistance against chemotherapy, targeted therapies, and biologics.
Collapse
Affiliation(s)
- Henry O Lamb
- School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Aurélie H Benfield
- School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Sónia Troeira Henriques
- School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Brisbane, QLD 4102, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
17
|
O’Flaherty S, Luzina OA, Dyrkheeva NS, Krier Y, Leprince J, Zakharenko AL, Pokrovsky MA, Pokrovsky AG, Lavrik OI, Salakhutdinov NF, Varbanov M, Devocelle M, Volcho KP. Novel Peptide-Drug Conjugates with Dual Anticancer Activity. Int J Mol Sci 2024; 25:12411. [PMID: 39596476 PMCID: PMC11594562 DOI: 10.3390/ijms252212411] [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: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Cationic antimicrobial peptides (AMPs), also called host defence peptides, have established antimicrobial and anticancer activities. Conjugation of an AMP to a bioactive molecule with complementary activity can address some of the clinical limitations of the peptide candidate. This approach has been particularly applied in antimicrobial applications of AMPs, but it remains relatively less explored in the generation of anticancer candidates. In this study, two usnic acid derivatives, based on hydrazinothiazole and benzylidenefuranone pharmacophore moieties, respectively, were conjugated to L-K6, a lysine/leucine-rich AMP, through a new pyrazole ligation intrinsically driven by the cargo molecule. Both components, the usnic acid derivative and the peptide, are selectively active against cancer cells, by targeting the human DNA repair enzyme tyrosyl-DNA phosphodiesterase 1 (TDP1) and through DNA damage, respectively. The two conjugates, based on a hydrazone linkage, exhibited pleiotropic effects, ranging from reduction in the activity of the parent drugs to their conservation or even enhancement. Notably, the conjugates retained some anti-TDP1 activity and displayed intermediate, or even higher, cytotoxicities against glioblastoma cells, compared to their individual components.
Collapse
Affiliation(s)
- Siobhán O’Flaherty
- Department of Chemistry, RCSI University of Medicine and Health Sciences, 123, St. Stephen’s Green, D02 YN77 Dublin, Ireland;
- SSPC (Synthesis and Solid State Pharmaceutical Centre) Research Centre, V94 T9PX Limerick, Ireland
| | - Olga A. Luzina
- Department of Medicinal Chemistry, N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 9 Acad. Lavrentjev Ave., 630090 Novosibirsk, Russia; (O.A.L.); (N.F.S.); (K.P.V.)
| | - Nadezhda S. Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 8, Lavrentjev Ave., 630090 Novosibirsk, Russia; (N.S.D.); (A.L.Z.); (O.I.L.)
| | - Ysaline Krier
- Laboratoire Lorraine de Chimie Moléculaire, Université de Lorraine, CNRS, L2CM, 54000 Nancy, France; (Y.K.); (M.V.)
| | - Jérôme Leprince
- Inserm, Rouen Normandie Université, NorDiC UMR 1239, 76000 Rouen, France;
- Rouen Normandie Université, HeRacLes UMS 51, PRIMACEN, 76000 Rouen, France
| | - Alexandra L. Zakharenko
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 8, Lavrentjev Ave., 630090 Novosibirsk, Russia; (N.S.D.); (A.L.Z.); (O.I.L.)
| | - Mikhail A. Pokrovsky
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 2 Pirogova str., 630090 Novosibirsk, Russia; (M.A.P.); (A.G.P.)
| | - Andrey G. Pokrovsky
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 2 Pirogova str., 630090 Novosibirsk, Russia; (M.A.P.); (A.G.P.)
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, SB RAS, 8, Lavrentjev Ave., 630090 Novosibirsk, Russia; (N.S.D.); (A.L.Z.); (O.I.L.)
| | - Nariman F. Salakhutdinov
- Department of Medicinal Chemistry, N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 9 Acad. Lavrentjev Ave., 630090 Novosibirsk, Russia; (O.A.L.); (N.F.S.); (K.P.V.)
| | - Mihayl Varbanov
- Laboratoire Lorraine de Chimie Moléculaire, Université de Lorraine, CNRS, L2CM, 54000 Nancy, France; (Y.K.); (M.V.)
| | - Marc Devocelle
- Department of Chemistry, RCSI University of Medicine and Health Sciences, 123, St. Stephen’s Green, D02 YN77 Dublin, Ireland;
- SSPC (Synthesis and Solid State Pharmaceutical Centre) Research Centre, V94 T9PX Limerick, Ireland
| | - Konstantin P. Volcho
- Department of Medicinal Chemistry, N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 9 Acad. Lavrentjev Ave., 630090 Novosibirsk, Russia; (O.A.L.); (N.F.S.); (K.P.V.)
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 2 Pirogova str., 630090 Novosibirsk, Russia; (M.A.P.); (A.G.P.)
| |
Collapse
|
18
|
Llanes D, Rennert R, Jänicke P, Morgan I, Reguera L, Rivera DG, Ricardo MG, Wessjohann LA. Development of bombesin-tubulysin conjugates using multicomponent chemistry to functionalize both the payload and the homing peptide. Front Pharmacol 2024; 15:1408091. [PMID: 39600359 PMCID: PMC11589458 DOI: 10.3389/fphar.2024.1408091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Peptide-drug conjugates (PDCs) have recently gained significant attention for the targeted delivery of anticancer therapeutics, mainly due to their cost-effective and chemically defined production and lower antigenicity compared to ADCs, among other benefits. In this study, we designed and synthesized novel PDCs by conjugating new thiol-functionalized tubulysin analogs (tubugis) to bombesin, a peptide ligand with a relevant role in cancer research. Two tubulysin analogs bearing ready-for-conjugation thiol groups were prepared by an on-resin multicomponent peptide synthesis strategy and subsequently tested for their stand-alone in vitro anti-proliferative activity against human cancer cells, which resulted in IC50 values in the nanomolar range. In addition, various fluorescently labeled [K5]-bombesin(6-14) peptides, non-lipidated and lipidated with fatty acid chains of variable length, were also synthesized using the versatile multicomponent chemistry. These bombesin derivatives were tested for their gastrin-related peptide receptor (GRPR)-mediated internalization into cancer cells using flow cytometry, proving that the lipid tail (especially C14) enhances the cell internalization. Using the tubugi toxins and bombesin peptides, three different bombesin-tubugi conjugates were synthesized with different cleavage propensity and lipophilicity. Preliminary in vitro experiments revealed that, depending on the linker and the presence of a lipid tail, these novel PDCs possess good to potent anticancer activity and moderate selectivity for GRPR-overexpressing cancer cells.
Collapse
Affiliation(s)
- Dayma Llanes
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Robert Rennert
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Paul Jänicke
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Ibrahim Morgan
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Leslie Reguera
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Havana, Cuba
| | - Daniel G. Rivera
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Havana, Cuba
| | - Manuel G. Ricardo
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Havana, Cuba
| | - Ludger A. Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| |
Collapse
|
19
|
Raveendran A, Ser J, Park SH, Jang P, Choi HS, Cho H. Lysosome-Targeted Bifunctional Therapeutics Induce Autodynamic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401424. [PMID: 39231370 PMCID: PMC11538690 DOI: 10.1002/advs.202401424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/01/2024] [Indexed: 09/06/2024]
Abstract
Autodynamic cancer therapy possesses tremendous potential for enhancing therapeutic efficacy by initiating the treatment process autonomously within targeted cells. However, challenges related to biocompatibility and targeted delivery have hindered its clinical translation owing to the induction of adverse effects and cytotoxicity in healthy cells. In this study, a novel approach for auto-initiated dynamic therapy by conjugating zwitterionic near-infrared fluorophores to a cell-penetrating peptide is proposed. This enables efficient cellular uptake and specific targeting of therapy to desired cells while avoiding off-target uptake. The zwitterionic bioconjugate causes cancer-specific toxicity following its internalization into the targeted cells, triggered by specific intracellular conditions in lysosomes. This innovative approach enables selective targeting of lysosomes in malignant cells while minimizing cytotoxic effects on normal cells. By targeting lysosomes, the method overcomes inherent risks and side effects associated with conventional cancer treatments, offering a selective and effective approach to cancer therapy.
Collapse
Affiliation(s)
- Athira Raveendran
- Department of Materials Science and EngineeringChonnam National UniversityGwangju61186Republic of Korea
| | - Jinhui Ser
- Department of Materials Science and EngineeringChonnam National UniversityGwangju61186Republic of Korea
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Seung Hun Park
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Paul Jang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hak Soo Choi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hoonsung Cho
- Department of Materials Science and EngineeringChonnam National UniversityGwangju61186Republic of Korea
| |
Collapse
|
20
|
Rizvi SFA, Zhang H, Fang Q. Engineering peptide drug therapeutics through chemical conjugation and implication in clinics. Med Res Rev 2024; 44:2420-2471. [PMID: 38704826 DOI: 10.1002/med.22046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2024] [Accepted: 04/21/2024] [Indexed: 05/07/2024]
Abstract
The development of peptide drugs has made tremendous progress in the past few decades because of the advancements in modification chemistry and analytical technologies. The novel-designed peptide drugs have been modified through various biochemical methods with improved diagnostic, therapeutic, and drug-delivery strategies. Researchers found it a helping hand to overcome the inherent limitations of peptides and bring continued advancements in their applications. Furthermore, the emergence of peptide-drug conjugates (PDCs)-utilizes target-oriented peptide moieties as a vehicle for cytotoxic payloads via conjugation with cleavable chemical agents, resulting in the key foundation of the new era of targeted peptide drugs. This review summarizes the various classifications of peptide drugs, suitable chemical modification strategies to improve the ADME (adsorption, distribution, metabolism, and excretion) features of peptide drugs, and recent (2015-early 2024) progress/achievements in peptide-based drug delivery systems as well as their fruitful implication in preclinical and clinical studies. Furthermore, we also summarized the brief description of other types of PDCs, including peptide-MOF conjugates and peptide-UCNP conjugates. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development and progress toward a bright future of novel peptide drugs.
Collapse
Affiliation(s)
- Syed Faheem Askari Rizvi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
21
|
Wiggins R, Woo J, Mito S. Optimizing Niclosamide for Cancer Therapy: Improving Bioavailability via Structural Modification and Nanotechnology. Cancers (Basel) 2024; 16:3548. [PMID: 39456642 PMCID: PMC11506536 DOI: 10.3390/cancers16203548] [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: 09/15/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Inhibition of multiple cancer-related pathways has made niclosamide a promising candidate for the treatment of various cancers. However, its clinical application has been significantly limited by poor bioavailability. This review will discuss current findings on improving niclosamide bioavailability through modification of its chemical structure and utilization of novel nanotechnologies, like electrospraying and supercritical fluids, to improve drug delivery. For example, niclosamide derivatives, such as o-alkylamino-tethered niclosamide derivates, niclosamide ethanolamine salt, and niclosamide piperazine salt, have demonstrated increased water solubility without compromising anticancer activity in vitro. Additionally, this review briefly discusses recent findings on the first pass metabolism of niclosamide in vivo, the role of cytochrome P450-mediated hydroxylation, UDP-glucuronosyltransferase mediated glucuronidation, and how enzymatic inhibition could enhance niclosamide bioavailability. Ultimately, there is a need for researchers to synthesize, evaluate, and improve upon niclosamide derivatives while experimenting with the employment of nanotechnologies, such as targeted delivery and nanoparticle modification, as a way to improve drug administration. Researchers should strive to improve drug-target accuracy, its therapeutic index, and increase the drug's efficacy as an anti-neoplastic agent.
Collapse
Affiliation(s)
| | | | - Shizue Mito
- Department of Medical Education, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA; (R.W.); (J.W.)
| |
Collapse
|
22
|
Jalil AT, Abdulhadi MA, Al-Ameer LR, Taher WM, Abdulameer SJ, Abosaooda M, Fadhil AA. Peptide-Based Therapeutics in Cancer Therapy. Mol Biotechnol 2024; 66:2679-2696. [PMID: 37768503 DOI: 10.1007/s12033-023-00873-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023]
Abstract
A monster called cancer is still one of the most challenging human problems and one of the leading causes of death in the world. Different types of treatment methods are used for cancer therapy; however, there are challenges such as high cost and harmful side effects in using these methods. Recent years have witnessed a surge in the development of therapeutic peptides for a wide range of diseases, notably cancer. Peptides are preferred over antibiotics, radiation therapy, and chemotherapy in the treatment of cancer due to a number of aspects, including flexibility, easy modification, low immunogenicity, and inexpensive cost of production. The use of therapeutic peptides in cancer treatment is a novel and intriguing strategy. These peptides provide excellent prospects for targeted drug delivery because of their high selectivity, specificity, small dimensions, good biocompatibility, and simplicity of modification. Target specificity and minimal toxicity are benefits of therapeutic peptides. Additionally, peptides can be used to design antigens or adjuvants for vaccine development. Here, types of therapeutic peptides for cancer therapy will be discussed, such as peptide-based cancer vaccines and tumor-targeting peptides (TTP) and cell-penetrating peptides (CPP).
Collapse
Affiliation(s)
- Abduladheem Turki Jalil
- Department of Medical Laboratories Techniques, Al-Mustaqbal University College, Hilla, Babylon, 51001, Iraq.
| | - Mohanad Ali Abdulhadi
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | - Lubna R Al-Ameer
- College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
| | | | - Sada Jasim Abdulameer
- Biology Department, College of Education for Pure Science, Wasit University, Kut, Wasit, Iraq
| | | | - Ali A Fadhil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
23
|
Verma VS, Pandey A, Jha AK, Badwaik HKR, Alexander A, Ajazuddin. Polyethylene Glycol-Based Polymer-Drug Conjugates: Novel Design and Synthesis Strategies for Enhanced Therapeutic Efficacy and Targeted Drug Delivery. Appl Biochem Biotechnol 2024; 196:7325-7361. [PMID: 38519751 DOI: 10.1007/s12010-024-04895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
Due to their potential to enhance therapeutic results and enable targeted drug administration, polymer-drug conjugates that use polyethylene glycol (PEG) as both the polymer and the linker for drug conjugation have attracted much research. This study seeks to investigate recent developments in the design and synthesis of PEG-based polymer-drug conjugates, emphasizing fresh ideas that fill in existing knowledge gaps and satisfy the increasing need for more potent drug delivery methods. Through an extensive review of the existing literature, this study identifies key challenges and proposes innovative strategies for future investigations. The paper presents a comprehensive framework for designing and synthesizing PEG-based polymer-drug conjugates, including rational molecular design, linker selection, conjugation methods, and characterization techniques. To further emphasize the importance and adaptability of PEG-based polymer-drug conjugates, prospective applications are highlighted, including cancer treatment, infectious disorders, and chronic ailments.
Collapse
Affiliation(s)
- Vinay Sagar Verma
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India
| | - Aakansha Pandey
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Arvind Kumar Jha
- Shri Shankaracharya Professional University, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Hemant Kumar Ramchandra Badwaik
- Shri Shankaracharya College of Pharmaceutical Sciences, Junwani, Bhilai, 490020, Chhattisgarh, India.
- Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India.
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Ministry of Chemical and Fertilizers, Guwahati, 781101, Assam, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India.
| |
Collapse
|
24
|
Hou M, Liu S. Recent Progress of pH-Responsive Peptides, Polypeptides, and Their Supramolecular Assemblies for Biomedical Applications. Biomacromolecules 2024; 25:5402-5416. [PMID: 39105715 DOI: 10.1021/acs.biomac.4c00688] [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: 08/07/2024]
Abstract
Peptides and polypeptides feature a variety of active functional groups on their side chains (including carboxylic acid, hydroxyl, amino, and thiol groups), enabling diverse chemical modifications. This versatility makes them highly valuable in stimuli-responsive systems. Notably, pH-responsive peptides and polypeptides, due to their ability to respond to pH changes, hold significant promise for applications in cellular pathology and tumor targeting. Extensive researches have highlighted the potentials of low pH insertion peptides (pHLIPs), peptide-drug conjugates (PDCs), and antibody-drug conjugates (ADCs) in biomedicine. Peptide self-assemblies, with their structural stability, ease of regulation, excellent biocompatibility, and biodegradability, offer immense potentials in the development of novel materials and biomedical applications. We also explore specific examples of their applications in drug delivery, tumor targeting, and tissue engineering, while discussing future challenges and potential advancements in the field of pH-responsive self-assembling peptide-based biomaterials.
Collapse
Affiliation(s)
- Mingxuan Hou
- Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jin-zhai Road, Hefei, Anhui Province 230026, China
| | - Shiyong Liu
- Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jin-zhai Road, Hefei, Anhui Province 230026, China
| |
Collapse
|
25
|
Sharma AK, Sharma R, Chauhan N, Das A, Satpati D. Peptide-drug conjugate designated for targeted delivery to HER2-expressing cancer cells. J Pept Sci 2024; 30:e3602. [PMID: 38600778 DOI: 10.1002/psc.3602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/15/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Targeted therapy of the highest globally incident breast cancer shall resolve the issue of off-target toxicity concurring with augmented killing of specific diseased cells. Thus, the goal of this study was to prepare a peptide-drug conjugate targeting elevated expression of HER2 receptors in breast cancer. Towards this, the rL-A9 peptide was conjugated with the chemotherapeutic drug doxorubicin (DOX) through a N-succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC) linker. The synthesized peptide-drug conjugate, rL-A9-DOX, was characterized by mass spectrometry. Molecular docking studies, based on binding energy data, suggested a stronger interaction of rL-A9-DOX with the HER2 receptor in comparison to the unconjugated peptide, rL-A9. The cytotoxic effect of the rL-A9-DOX conjugate was observed to be higher in HER2-positive SKOV3 cells compared to HER2-negative MDA-MB-231 cells, indicating selective cell killing. Cellular internalization of the rL-A9-DOX conjugate was evident from the flow cytometry analysis, where a noticeable shift in mean fluorescent intensity (MFI) was observed for the conjugate compared to the control group. This data was further validated by confocal microscopy, where the fluorescent signal ascertained nuclear accumulation of rL-A9-DOX. The present studies highlight the promising potential of rL-A9-DOX for targeted delivery of the drug into a defined group of cancer cells.
Collapse
Affiliation(s)
- Amit Kumar Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rohit Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Nitish Chauhan
- Homi Bhabha National Institute, Mumbai, India
- Bio-Organic Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Amit Das
- Homi Bhabha National Institute, Mumbai, India
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
26
|
Lee JW, Yoon HY, Ko YJ, Kim EH, Song S, Hue S, Gupta N, Malin D, Kim J, Kong B, Kim S, Kim IS, Kwon IC, Yang Y, Kim SH. Dual-Action Protein-siRNA Conjugates for Targeted Disruption of CD47-Signal Regulatory Protein α Axis in Cancer Therapy. ACS NANO 2024; 18:22298-22315. [PMID: 39117621 DOI: 10.1021/acsnano.4c06471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
A series of successes in RNA interference (RNAi) therapies for liver diseases using lipid nanoparticles and N-acetylgalactosamine have heralded a current era of RNA therapeutics. However, alternative delivery strategies are required to take RNAi out of the comfort zone of hepatocytes. Here we report SIRPα IgV/anti-CD47 siRNA (vS-siCD47) conjugates that selectively and persistently disrupt the antiphagocytic CD47/SIRPα axis in solid tumors. Conjugation of the SIRPα IgV domain protein to siRNAs enables tumor dash through CD47-mediated erythrocyte piggyback, primarily blocking the physical interaction between CD47 on cancer cells and SIRPα on phagocytes. After internalization of the vS-siCD47 conjugates within cancer cells, the detached free-standing anti-CD47 siRNAs subsequently attack CD47 through the RNAi mechanism. The dual-action approach of the vS-siCD47 conjugate effectively overcomes the "don't eat me" barrier and stimulates phagocyte-mediated tumor destruction, demonstrating a highly selective and potent CD47-blocking immunotherapy. This delivery strategy, employing IgV domain protein-siRNA conjugates with a dual mode of target suppression, holds promise for expanding RNAi applications beyond hepatocytes and advancing RNAi-based cancer immunotherapies for solid tumors.
Collapse
Affiliation(s)
- Jong Won Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Hwarang-ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Young Ji Ko
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eun Hye Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Sukyung Song
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seungmi Hue
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Nilaksh Gupta
- K2B Therapeutics, Cambridge, Massachusetts 02139, United States
| | - Dmitry Malin
- K2B Therapeutics, Cambridge, Massachusetts 02139, United States
| | - Jay Kim
- K2B Therapeutics, Cambridge, Massachusetts 02139, United States
| | - Byoungjae Kong
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Sehoon Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Hwarang-ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sun Hwa Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
27
|
Sadique FL, Subramaiam H, Krishnappa P, Chellappan DK, Ma JH. Recent advances in breast cancer metastasis with special emphasis on metastasis to the brain. Pathol Res Pract 2024; 260:155378. [PMID: 38850880 DOI: 10.1016/j.prp.2024.155378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Understanding the underlying mechanisms of breast cancer metastasis is of vital importance for developing treatment approaches. This review emphasizes contemporary breakthrough studies with special focus on breast cancer brain metastasis. Acquired mutational changes in metastatic lesions are often distinct from the primary tumor, suggesting altered mutagenesis pathways. The concept of micrometastases and heterogeneity within the tumors unravels novel therapeutic targets at genomic and molecular levels through epigenetic and proteomic profiling. Several pre-clinical studies have identified mechanisms involving the immune system, where tumor associated macrophages are key players. Expression of cell proteins like Syndecan1, fatty acid-binding protein 7 and tropomyosin kinase receptor B have been implicated in aiding the transmigration of breast cancer cells to the brain. Changes in the proteomic landscape of the blood-brain-barrier show altered permeability characteristics, supporting entry of cancer cells. Findings from laboratory studies pave the path for the emergence of new biomarkers, especially blood-based miRNA and circulating tumor cell markers for prognostic staging. The constantly evolving therapeutics call for clinical trials backing supportive evidence of efficacies of both novel and existing approaches. The challenge lying ahead is discovering innovative techniques to replace use of human samples and optimize small-scale patient recruitment in trials.
Collapse
Affiliation(s)
- Fairooz Labiba Sadique
- Department of Biomedical Science, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Hemavathy Subramaiam
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia.
| | - Purushotham Krishnappa
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Jin Hao Ma
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
28
|
Ricardo MG, Llanes D, Rennert R, Jänicke P, Rivera DG, Wessjohann LA. Improved Access to Potent Anticancer Tubulysins and Linker-Functionalized Payloads Via an All-On-Resin Strategy. Chemistry 2024; 30:e202401943. [PMID: 38771268 DOI: 10.1002/chem.202401943] [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: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 05/22/2024]
Abstract
Tubulysins are among the most recent antimitotic compounds to enter into antibody/peptide-drug conjugate (ADC/PDC) development. Thus far, the design of the most promising tubulysin payloads relied on simplifying their structures, e. g., by using small tertiary amide N-substituents (Me, Et, Pr) on the tubuvaline residue. Cumbersome solution-phase approaches are typically used for both syntheses and functionalization with cleavable linkers. p-Aminobenzyl quaternary ammonium (PABQ) linkers were a remarkable advancement for targeted delivery, but the procedures to incorporate them into tubulysins are only of moderate efficiency. Here we describe a novel all-on-resin strategy permitting a loss-free resin linkage and an improved access to super potent tubulysin analogs showing close resemblance to the natural compounds. For the first time, a protocol enables the integration of on-resin tubulysin derivatization with, e. g., a maleimido-Val-Cit-PABQ linker, which is a notable progress for the payload-PABQ-linker technology. The strategy also allows tubulysin diversification of the internal amide N-substituent, thus enabling to screen a tubulysin library for the discovery of new potent analogs. This work provides ADC/PDC developers with new tools for both rapid access to new derivatives and easier linker-attachment and functionalization.
Collapse
Affiliation(s)
- Manuel G Ricardo
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120, Halle (Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata & G, Havana, 10400, Cuba
- Present address: Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, D-14476, Potsdam, Germany
| | - Dayma Llanes
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120, Halle (Saale), Germany
| | - Robert Rennert
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120, Halle (Saale), Germany
| | - Paul Jänicke
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120, Halle (Saale), Germany
| | - Daniel G Rivera
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120, Halle (Saale), Germany
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata & G, Havana, 10400, Cuba
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120, Halle (Saale), Germany
| |
Collapse
|
29
|
Song H, Sgouros G. Alpha and Beta Radiation for Theragnostics. PET Clin 2024; 19:307-323. [PMID: 38688775 DOI: 10.1016/j.cpet.2024.03.006] [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] [Indexed: 05/02/2024]
Abstract
Targeted radionuclide therapy (TRT) has significantly evolved from its beginnings with iodine-131 to employing carrier molecules with beta emitting isotopes like lutetium-177. With the success of Lu-177-DOTATATE for neuroendocrine tumors and Lu-177-PSMA-617 for prostate cancer, several other beta emitting radioisotopes, such as Cu-67 and Tb-161, are being explored for TRT. The field has also expanded into targeted alpha therapy (TAT) with agents like radium-223 for bone metastases in prostate cancer, and several other alpha emitter radioisotopes with carrier molecules, such as Ac-225, and Pb-212 under clinical trials. Despite these advancements, the scope of TRT in treating diverse solid tumors and integration with other therapies like immunotherapy remains under investigation. The success of antibody-drug conjugates further complements treatments with TRT, though challenges in treatment optimization continue.
Collapse
Affiliation(s)
- Hong Song
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA.
| | - George Sgouros
- Division of Radiological Physics, Department of Radiology and Radiological Sciences, The Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
30
|
Milewska S, Sadowska A, Stefaniuk N, Misztalewska-Turkowicz I, Wilczewska AZ, Car H, Niemirowicz-Laskowska K. Tumor-Homing Peptides as Crucial Component of Magnetic-Based Delivery Systems: Recent Developments and Pharmacoeconomical Perspective. Int J Mol Sci 2024; 25:6219. [PMID: 38892406 PMCID: PMC11172452 DOI: 10.3390/ijms25116219] [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: 04/10/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
According to data from the World Health Organization (WHO), cancer is considered to be one of the leading causes of death worldwide, and new therapeutic approaches, especially improved novel cancer treatment regimens, are in high demand. Considering that many chemotherapeutic drugs tend to have poor pharmacokinetic profiles, including rapid clearance and limited on-site accumulation, a combined approach with tumor-homing peptide (THP)-functionalized magnetic nanoparticles could lead to remarkable improvements. This is confirmed by an increasing number of papers in this field, showing that the on-target peptide functionalization of magnetic nanoparticles improves their penetration properties and ensures tumor-specific binding, which results in an increased clinical response. This review aims to highlight the potential applications of THPs in combination with magnetic carriers across various fields, including a pharmacoeconomic perspective.
Collapse
Affiliation(s)
- Sylwia Milewska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | - Natalia Stefaniuk
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | | | - Agnieszka Z. Wilczewska
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland; (I.M.-T.); (A.Z.W.)
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| | - Katarzyna Niemirowicz-Laskowska
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland; (S.M.); (A.S.); (N.S.); (H.C.)
| |
Collapse
|
31
|
Bai H, Huang W, Li J, Ji Y, He S, Hu H. Enhancing anticancer treatment: Development of cRGD-Conjugated F-OH-Evo prodrugs for targeted delivery. Bioorg Med Chem 2024; 107:117759. [PMID: 38795572 DOI: 10.1016/j.bmc.2024.117759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/28/2024]
Abstract
Small molecule drugs sourced from natural products are pivotal for novel therapeutic discoveries. However, their clinical deployment is often impeded by non-specific activity and severe adverse effects. This study focused on 3-fluoro-10-hydroxy-Evodiamine (F-OH-Evo), a potent derivative of Evodiamine, whose development is curtailed due to suboptimal tumor selectivity and heightened cytotoxicity. By harnessing the remarkable stability, specificity, and αvβ3 integrin affinity of c(RGDFK), a novel prodrug by conjugating F-OH-Evo with cRGD was synthesized. This innovative prodrug substantially enhanced the tumor-specific targeting of F-OH-Evo and improved the anti-tumor activities. Among them, compound 3c demonstrated the best selective inhibitory activity toward U87 cancer cells in vitro. It selectively enterd U87 cells by binding to αvβ3 integrin, releasing the parent molecule under the dual response of ROS and GSH to exert inhibitory activity on topo I. The results highlight the potential of cRGD-conjugated prodrugs in targeted cancer therapy. This approach signifies a significant advancement in developing safer and more effective chemotherapy drugs, emphasizing the role of prodrug strategies in overcoming the limitations of traditional cancer treatments.
Collapse
Affiliation(s)
- Haohao Bai
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Wenjing Huang
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Jinqiu Li
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Yajing Ji
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Shipeng He
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China.
| | - Honggang Hu
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China.
| |
Collapse
|
32
|
Sun X, Liu Y, Ma T, Zhu N, Lao X, Zheng H. DCTPep, the data of cancer therapy peptides. Sci Data 2024; 11:541. [PMID: 38796630 PMCID: PMC11128002 DOI: 10.1038/s41597-024-03388-9] [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: 07/19/2023] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
With the discovery of the therapeutic activity of peptides, they have emerged as a promising class of anti-cancer agents due to their specific targeting, low toxicity, and potential for high selectivity. In particular, as peptide-drug conjugates enter clinical, the coupling of targeted peptides with traditional chemotherapy drugs or cytotoxic agents will become a new direction in cancer treatment. To facilitate the drug development of cancer therapy peptides, we have constructed DCTPep, a novel, open, and comprehensive database for cancer therapy peptides. In addition to traditional anticancer peptides (ACPs), the peptide library also includes peptides related to cancer therapy. These data were collected manually from published research articles, patents, and other protein or peptide databases. Data on drug library include clinically investigated and/or approved peptide drugs related to cancer therapy, which mainly come from the portal websites of drug regulatory authorities and organisations in different countries and regions. DCTPep has a total of 6214 entries, we believe that DCTPep will contribute to the design and screening of future cancer therapy peptides.
Collapse
Affiliation(s)
- Xin Sun
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P. R. China
| | - Yanchao Liu
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P. R. China
| | - Tianyue Ma
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P. R. China
| | - Ning Zhu
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P. R. China
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P. R. China.
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, P. R. China.
| |
Collapse
|
33
|
Franco Machado J, Sá M, Pires I, da Silva MT, Marques F, Coelho JAS, Mendes F, Piedade MFM, Machuqueiro M, Jiménez MA, Garcia MH, Correia JDG, Morais TS. Dual FGFR-targeting and pH-activatable ruthenium-peptide conjugates for targeted therapy of breast cancer. Dalton Trans 2024; 53:7682-7693. [PMID: 38573236 DOI: 10.1039/d4dt00497c] [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: 04/05/2024]
Abstract
Dysregulation of Fibroblast Growth Factor Receptors (FGFRs) signaling has been associated with breast cancer, yet employing FGFR-targeted delivery systems to improve the efficacy of cytotoxic agents is still sparsely exploited. Herein, we report four new bi-functional ruthenium-peptide conjugates (RuPCs) with FGFR-targeting and pH-dependent releasing abilities, envisioning the selective delivery of cytotoxic Ru complexes to FGFR(+)-breast cancer cells, and controlled activation at the acidic tumoral microenvironment. The antiproliferative potential of the RuPCs and free Ru complexes was evaluated in four breast cancer cell lines with different FGFR expression levels (SKBR-3, MDA-MB-134-VI, MCF-7, and MDA-MB-231) and in human dermal fibroblasts (HDF), at pH 6.8 and pH 7.4 aimed at mimicking the tumor microenvironment and normal tissues/bloodstream pHs, respectively. The RuPCs showed higher cytotoxicity in cells with higher level of FGFR expression at acidic pH. Additionally, RuPCs showed up to 6-fold higher activity in the FGFR(+) breast cancer lines compared to the normal cell line. The release profile of Ru complexes from RuPCs corroborates the antiproliferative effects observed. Remarkably, the cytotoxicity and releasing ability of RuPCs were shown to be strongly dependent on the conjugation of the peptide position in the Ru complex. Complementary molecular dynamic simulations and computational calculations were performed to help interpret these findings at the molecular level. In summary, we identified a lead bi-functional RuPC that holds strong potential as a FGFR-targeted chemotherapeutic agent.
Collapse
Affiliation(s)
- João Franco Machado
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
| | - Marco Sá
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Inês Pires
- BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Miguel Tarita da Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Jaime A S Coelho
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Filipa Mendes
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - M Fátima M Piedade
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Miguel Machuqueiro
- BioISI - Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - María Angeles Jiménez
- Institute of Physical Chemistry Blas Cabreras (IQF-CSIC), Serrano 119, E-28006 Madrid, Spain
| | - Maria Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal.
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139, 7), 2695-066 Bobadela LRS, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal.
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
34
|
Kumar N, Sharma AK, Guleria M, Shelar SB, Chakraborty A, Rakshit S, Kolay S, Satpati D, Das T. Nuclear Localization Signal Enhances the Targeting and Therapeutic Efficacy of a Porphyrin-Based Molecular Cargo: A Systemic In Vitro and Ex Vivo Evaluation. Mol Pharm 2024; 21:2351-2364. [PMID: 38477252 DOI: 10.1021/acs.molpharmaceut.3c01152] [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] [Indexed: 03/14/2024]
Abstract
The objective of the present work was to evaluate the potential of a nuclear localization signal (NLS) toward facilitating intracellular delivery and enhancement in the therapeutic efficacy of the molecular cargo. Toward this, an in-house synthesized porphyrin derivative, namely, 5-carboxymethyelene-oxyphenyl-10,15,20-tris(4-methoxyphenyl) porphyrin (UTriMA), was utilized for conjugation with the NLS sequence [PKKKRKV]. The three compounds synthesized during the course of the present work, namely DOTA-Lys-NLS, DOTA-UTriMA-Lys-NLS, and DOTA-Lys-UTriMA, were evaluated for cellular toxicity in cancer cell lines (HT1080), wherein all exhibited minimal dark toxicity. However, during photocytotoxicity studies with DOTA-Lys-UTriMA and DOTA-UTriMA-Lys-NLS conjugates in the same cell line, the latter exhibited significantly higher light-dependent toxicity compared to the former. Furthermore, the photocytotoxicity for DOTA-UTriMA-Lys-NLS in a healthy cell line (WI26VA4) was found to be significantly lower than that observed in the cancer cells. Fluorescence cell imaging studies carried out in HT1080 cancer cells revealed intracellular accumulation for the NLS-conjugated porphyrin (DOTA-UTriMA-Lys-NLS), whereas unconjugated porphyrin (DOTA-Lys-UTriMA) failed to do so. To evaluate the radiotherapeutic effects of the synthesized conjugates, all three compounds were radiolabeled with 177Lu, a well-known therapeutic radionuclide with high radiochemical purity (>95%). During in vitro studies, the [177Lu]Lu-DOTA-UTriMA-Lys-NLS complex exhibited the highest cell binding as well as internalization among the three radiolabeled complexes. Biological distribution studies for the radiolabeled compounds were performed in a fibrosarcoma-bearing small animal model, wherein significantly higher accumulation and prolonged retention of [177Lu]Lu-DOTA-UTriMA-Lys-NLS (9.32 ± 1.27% IA/g at 24 h p.i.) in the tumorous lesion compared to [177Lu]Lu-UTriMA-Lys-DOTA (2.3 ± 0.13% IA/g at 24 h p.i.) and [177Lu]Lu-DOTA-Lys-NLS complexes (0.26 ± 0.17% IA/g at 24 h p.i.) were observed. The results of the biodistribution studies were further corroborated by recording serial SPECT-CT images of fibrosarcoma-bearing Swiss mice administered with [177Lu]Lu-DOTA-UTriMA-Lys-NLS at different time points. Tumor regression studies performed with [177Lu]Lu-DOTA-UTriMA-Lys-NLS in the same animal model with two different doses [250 μCi (9.25 MBq) and 500 μCi (18.5 MBq)] resulted in a significant reduction in tumor mass in the treated group of animals. The above results revealed a definite enhancement in the targeting ability of molecular cargo upon conjugation with NLS and hence indicated that this strategy may be helpful for the preparation of drug-NLS conjugates as multimodal agents.
Collapse
Affiliation(s)
- Naveen Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Amit K Sharma
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Mohini Guleria
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Sandeep B Shelar
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Avik Chakraborty
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai 400012, India
| | - Sutapa Rakshit
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Parel, Mumbai 400012, India
| | - Soumi Kolay
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Tapas Das
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
35
|
Wei H, Dong C, Li X. Treatment Options for Hepatocellular Carcinoma Using Immunotherapy: Present and Future. J Clin Transl Hepatol 2024; 12:389-405. [PMID: 38638377 PMCID: PMC11022065 DOI: 10.14218/jcth.2023.00462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 04/20/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a common cancer, and the body's immune responses greatly affect its progression and the prognosis of patients. Immunological suppression and the maintenance of self-tolerance in the tumor microenvironment are essential responses, and these form part of the theoretical foundations of immunotherapy. In this review, we first discuss the tumor microenvironment of HCC, describe immunosuppression in HCC, and review the major biomarkers used to track HCC progression and response to treatment. We then examine antibody-based therapies, with a focus on immune checkpoint inhibitors (ICIs), monoclonal antibodies that target key proteins in the immune response (programmed cell death protein 1, anti-cytotoxic T-lymphocyte associated protein 4, and programmed death-ligand 1) which have transformed the treatment of HCC and other cancers. ICIs may be used alone or in conjunction with various targeted therapies for patients with advanced HCC who are receiving first-line treatments or subsequent treatments. We also discuss the use of different cellular immunotherapies, including T cell receptor (TCR) T cell therapy and chimeric antigen receptor (CAR) T cell therapy. We then review the use of HCC vaccines, adjuvant immunotherapy, and oncolytic virotherapy, and describe the goals of future research in the development of treatments for HCC.
Collapse
Affiliation(s)
- Hongbin Wei
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chunlu Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, Gansu, China
- Cancer Prevention and Treatment Center of Lanzhou University School of Medicine, Lanzhou, Gansu, China
- Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou, Gansu, China
- Clinical Research Center for General Surgery of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
36
|
Beloborodov E, Iurova E, Fomin A, Saenko Y. Development and Synthesis of Bombesin-Based Radiopharmaceutical Precursors Modified with Knottin. Sovrem Tekhnologii Med 2024; 16:5-14. [PMID: 39539754 PMCID: PMC11556054 DOI: 10.17691/stm2024.16.2.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 11/16/2024] Open
Abstract
Bombesin receptors on the cell surface are of great interest as a target for targeted cancer therapy. One of the strategies of targeting bombesin receptors involves the use of tropic short peptides. However, the main limitation for the wide application of peptides as drugs is their low stability in vivo due to their sensitivity to extreme conditions of the internal body environment such as temperature and action of enzymes. In our work, a short bombesin peptide, taken as a basis, was modified with a knottin, a toxin with an inhibitor cystine knot, increasing thereby the stability of the short peptide under various conditions. The aim of the investigation is to study the chemical and radiochemical stability of the structure based on the short bombesin peptide and knottin, as well as the ability of the obtained structure to bind to tumor cells. Materials and Methods The work analyzed the chemical and radiochemical stability of the synthesized peptide labeled with a lutetium radioisotope using high-performance liquid chromatography. A fluorescent-labeled peptide, obtained by a solid-phase peptide synthesis, was used to analyze binding to cultures expressing bombesin receptors. Results The analysis has shown increased chemical and radiochemical stability of the knottin-modified peptide, as compared to the commercial analog, and maintenance of a high ability to bind to receptors on the surface of cancer cells. Conclusion The structure created on the basis of a short bombesin peptide and knottin possesses increased stability and retains the ability to bind to cancer cells. All this allows us to consider the creation of these structures as a strategy for fabricating stabilizing scaffolds for short peptides for a peptide-receptor therapy.
Collapse
Affiliation(s)
- E.A. Beloborodov
- Researcher, Laboratory for Peptide Drugs and Vaccines Development, S.P. Kapitsa Research Institute of Technology; Ulyanovsk State University, 42 Leo Tolstoy St., Ulyanovsk, 432017, Russia
| | - E.V. Iurova
- Junior Researcher, Laboratory for Peptide Drugs and Vaccine Development, S.P. Kapitsa Research Institute of Technology; Ulyanovsk State University, 42 Leo Tolstoy St., Ulyanovsk, 432017, Russia
| | - A.N. Fomin
- PhD, Senior Researcher, Laboratory for Peptide Drugs and Vaccines Development, S.P. Kapitsa Research Institute of Technology; Ulyanovsk State University, 42 Leo Tolstoy St., Ulyanovsk, 432017, Russia
| | - Yu.V. Saenko
- DSc, Leading Researcher, Laboratory for Peptide Drugs and Vaccines Development, S.P. Kapitsa Research Institute of Technology; Ulyanovsk State University, 42 Leo Tolstoy St., Ulyanovsk, 432017, Russia
| |
Collapse
|
37
|
Sugiharti RJ, Maharani R, Kurniawan F, Kartasasmita RE, Tjahjono DH. Computational studies and synthesis of 131iodine-labeled nocardiotide A analogs as a peptide-based theragnostic radiopharmaceutical ligand for cancer targeting SSTR2. RSC Adv 2024; 14:10962-10968. [PMID: 38577429 PMCID: PMC10993231 DOI: 10.1039/d4ra00684d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/22/2024] [Indexed: 04/06/2024] Open
Abstract
Radiolabeled peptides belong to a highly specific group of radiotracers used in oncology, particularly for diagnostics and cancer therapy. With the notable advantages of high binding affinity and selectivity to cancer cells, they have proven to be very useful in nuclear medicine. As a result, efforts have been focused on discovering new peptide sequences for radiopeptide preparation. Nocardiotide A, a cyclic hexapeptide comprising the amino acids cyclo-Trp-Ile-Trp-Leu-Val-Ala (cWIWLVA) isolated from Nocardiopsis sp., has shown significant cytotoxicity against cancer cells, rendering it a suitable candidate for the process. Therefore, the present study aimed to design a stable and effective radiopeptide by labeling nocardiotide A with iodine-131 (131I), ensuring that its affinity to SSTR2 is not compromised. In silico study showed that structural modification of nocardiotide A labeled with 131iodine exhibited good affinity value, forming hydrogen bonds with key residues, such as Q.102 and T.194, which are essential in SSTR2. Based on the results, cyclic hexapeptides of cWIWLYA were selected for further synthesis, and its peptide product was confirmed by the presence of an ionic molecule peak m/z [M + Na]+ 855.4332 (yield, 25.60%). In vitro tests conducted on cWIWLYA showed that cWIWLYA can bind to HeLa cancer cells. Radiopeptide synthesis was initiated with radiolabeling of cWIWLYA by 131I using the chloramine-T method that showed a radiochemical yield of 93.37%. Non-radioactive iodine labeling reaction showed that iodination was successful, which detected the presence of di-iodinated peptide (I2-cWIWLYA) with m/z [M + Na]+ 1107.1138. In summary, a radiopeptide derived from nocardiotide A showed great potential for further development as a diagnostic and therapeutic agent in cancer treatment.
Collapse
Affiliation(s)
- Rizky Juwita Sugiharti
- School of Pharmacy, Bandung Institute of Technology Bandung Indonesia
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency Indonesia
| | - Rani Maharani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor Indonesia
| | | | | | | |
Collapse
|
38
|
Vankadara S, Ke Z, Wang S, Foo SY, Gunaratne J, Lee MA, Koh X, Chia CSB. Cytotoxic activity and cell specificity of a novel LHRH peptide drug conjugate, D-Cys6-LHRH vedotin, against ovarian cancer cell lines. Chem Biol Drug Des 2024; 103:e14516. [PMID: 38618710 DOI: 10.1111/cbdd.14516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/27/2024] [Accepted: 03/16/2024] [Indexed: 04/16/2024]
Abstract
Ovarian cancer is the most deadly female gynaecological malignancy in developed countries and new treatments are urgently needed. The luteinising hormone releasing hormone (LHRH) peptide drug conjugate Zoptarelin doxorubicin is one such potential new drug modality that entered clinical trials for treating LHRH receptor-positive gynaecological cancers. However, development stopped after disappointing Phase 3 results in 2017. We believe the lack of efficacy was due to linker instability and payload potency. In this work, we replaced its linker-toxin with vedotin (MC-VC-PABC-MMAE), yielding the novel peptide drug conjugate D-Cys6-LHRH vedotin. A GI50 and cell specificity comparison against cancerous and non-cancerous ovarian cell lines showed significantly superior bioactivity and selectivity over Zoptarelin doxorubicin (GI50 4 vs. 453 nM) and other chemotherapeutic drugs used for treating ovarian cancers. Our results suggest D-Cys6-LHRH vedotin can potentially be used as a treatment for ovarian cancer.
Collapse
Affiliation(s)
- Subramanyam Vankadara
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Zhiyuan Ke
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Sifang Wang
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Sin Yun Foo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Jayantha Gunaratne
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, 4 Medical Drive, MD10, National University of Singapore, Singapore, Republic of Singapore
| | - May Ann Lee
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Xiaoying Koh
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - C S Brian Chia
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| |
Collapse
|
39
|
Sun B, Zhang L, Li M, Wang X, Wang W. Applications of peptide-based nanomaterials in targeting cancer therapy. Biomater Sci 2024; 12:1630-1642. [PMID: 38404259 DOI: 10.1039/d3bm02026f] [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: 02/27/2024]
Abstract
To meet the demand for precision medicine, researchers are committed to developing novel strategies to reduce systemic toxicity and side effects in cancer treatment. Targeting peptides are widely applied due to their affinity and specificity, and their ability to be high-throughput screened, chemically synthesized and modified. More importantly, peptides can form ordered self-assembled structures through non-covalent supramolecular interactions, which can form nanostructures with different morphologies and functions, playing crucial roles in targeted diagnosis and treatment. Among them, in targeted immunotherapy, utilizing targeting peptides to block the binding between immune checkpoints and ligands, thereby activating the immune system to eliminate cancer cells, is an advanced therapeutic strategy. In this mini-review, we summarize the screening, self-assembly, and biomedical applications of targeting peptide-based nanomaterials. Furthermore, this mini-review summarizes the potential and optimization strategies of targeting peptides.
Collapse
Affiliation(s)
- Beilei Sun
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China.
| | - Limin Zhang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China.
| | - Mengzhen Li
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China.
| | - Xin Wang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China.
| | - Weizhi Wang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China.
| |
Collapse
|
40
|
Al Musaimi O. Peptide Therapeutics: Unveiling the Potential against Cancer-A Journey through 1989. Cancers (Basel) 2024; 16:1032. [PMID: 38473389 PMCID: PMC11326481 DOI: 10.3390/cancers16051032] [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: 02/06/2024] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
The United States Food and Drug Administration (FDA) has approved a plethora of peptide-based drugs as effective drugs in cancer therapy. Peptides possess high specificity, permeability, target engagement, and a tolerable safety profile. They exhibit selective binding with cell surface receptors and proteins, functioning as agonists or antagonists. They also serve as imaging agents for diagnostic applications or can serve a dual-purpose as both diagnostic and therapeutic (theragnostic) agents. Therefore, they have been exploited in various forms, including linkers, peptide conjugates, and payloads. In this review, the FDA-approved prostate-specific membrane antigen (PSMA) peptide antagonists, peptide receptor radionuclide therapy (PRRT), somatostatin analogs, antibody-drug conjugates (ADCs), gonadotropin-releasing hormone (GnRH) analogs, and other peptide-based anticancer drugs are analyzed in terms of their chemical structures and properties, therapeutic targets and mechanisms of action, development journey, administration routes, and side effects.
Collapse
Affiliation(s)
- Othman Al Musaimi
- School of Pharmacy, Faculty of Medical Sciences, Newcastle upon Tyne NE1 7RU, UK
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
41
|
Zhou L, Lu Y, Liu W, Wang S, Wang L, Zheng P, Zi G, Liu H, Liu W, Wei S. Drug conjugates for the treatment of lung cancer: from drug discovery to clinical practice. Exp Hematol Oncol 2024; 13:26. [PMID: 38429828 PMCID: PMC10908151 DOI: 10.1186/s40164-024-00493-8] [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: 01/06/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
A drug conjugate consists of a cytotoxic drug bound via a linker to a targeted ligand, allowing the targeted delivery of the drug to one or more tumor sites. This approach simultaneously reduces drug toxicity and increases efficacy, with a powerful combination of efficient killing and precise targeting. Antibody‒drug conjugates (ADCs) are the best-known type of drug conjugate, combining the specificity of antibodies with the cytotoxicity of chemotherapeutic drugs to reduce adverse reactions by preferentially targeting the payload to the tumor. The structure of ADCs has also provided inspiration for the development of additional drug conjugates. In recent years, drug conjugates such as ADCs, peptide‒drug conjugates (PDCs) and radionuclide drug conjugates (RDCs) have been approved by the Food and Drug Administration (FDA). The scope and application of drug conjugates have been expanding, including combination therapy and precise drug delivery, and a variety of new conjugation technology concepts have emerged. Additionally, new conjugation technology-based drugs have been developed in industry. In addition to chemotherapy, targeted therapy and immunotherapy, drug conjugate therapy has undergone continuous development and made significant progress in treating lung cancer in recent years, offering a promising strategy for the treatment of this disease. In this review, we discuss recent advances in the use of drug conjugates for lung cancer treatment, including structure-based drug design, mechanisms of action, clinical trials, and side effects. Furthermore, challenges, potential approaches and future prospects are presented.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shanglong Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
42
|
Srivastava V, Godara P, Jena SP, Naik B, Singh S, Prajapati VK, Prusty D. Peptide-ligand conjugate based immunotherapeutic approach for targeted dismissal of non-structural protein 1 of dengue virus: A novel therapeutic solution for mild and severe dengue infections. Int J Biol Macromol 2024; 260:129562. [PMID: 38246445 DOI: 10.1016/j.ijbiomac.2024.129562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Dengue virus infection has significantly increased, with reported cases soaring from 505,430 in 2000 to 2,809,818 in 2022, emphasizing the need for effective treatments. Among the eleven structural and non-structural proteins of DENV, Non-structural protein 1 (NS1) has emerged as a promising target due to its diverse role in modulating the immune response, inducing vascular leakage, and facilitating viral replication and assembly. Monoclonal antibodies are the sole therapeutics to target NS1, but concerns about their cross-reactivity persist. Given these concerns, our study focuses on designing a novel Peptide Ligand Conjugate (PLC) as a potential alternative immunotherapeutic agent against NS1. This PLC aims to mediate the immune elimination of soluble NS1 and NS1-presenting DENV-infected host cells by pre-existing vaccine-induced immunity. By employing the High Throughput Virtual Screening (HTVS) method, QikProp analysis, and Molecular Dynamics studies, we identified three hits from Asinex Biodesigned Ligands out of 220,177 compounds that show strong binding affinity towards the monoclonal binding site of NS1 protein. After a rigorous analysis of physicochemical characteristics, antigenicity, allergenicity, and toxicity using various servers, we selected two peptides: the minimum epitopic region of the Diphtheria and Tetanus toxins as the peptide components of the PLCs. A non-cleavable, non-reactive oxime linker connected the ligand with the peptide through oxime and amide bonds. DPT vaccine is widely used in dengue-endemic countries, and it has been reported that antibodies titer against MER of Diphtheria toxin and Tetanus toxins persist lifelong in DPT-vaccinated people. Therefore, once the rationally designed PLCs bind to NS1 through the ligands, the peptide will induce an immune response against NS1 by triggering pre-existing DPT antibodies and activating memory cells. This orchestrated immune response will destroy soluble NS1 and NS1-expressing DENV-infected cells, thereby reducing the illness of severe dengue hemorrhagic fever and the DENV infection, respectively. Given the increasing demand for new therapeutics for DENV treatment, further investigation into this novel immune-therapeutic strategy may offer a new avenue for treating mild and severe dengue infections.
Collapse
Affiliation(s)
- Varshita Srivastava
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Priya Godara
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Sudip Prasad Jena
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Biswajit Naik
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Satyendra Singh
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Dhaneswar Prusty
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, 305817, India.
| |
Collapse
|
43
|
Guo S, Wang J, Wang Q, Wang J, Qin S, Li W. Advances in peptide-based drug delivery systems. Heliyon 2024; 10:e26009. [PMID: 38404797 PMCID: PMC10884816 DOI: 10.1016/j.heliyon.2024.e26009] [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: 07/03/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
Drug delivery systems (DDSs) are designed to deliver drugs to their specific targets to minimize their toxic effects and improve their susceptibility to clearance during targeted transport. Peptides have high affinity, low immunogenicity, simple amino acid composition, and adjustable molecular size; therefore, most peptides can be coupled to drugs via linkers to form peptide-drug conjugates (PDCs) and act as active pro-drugs. PDCs are widely thought to be promising DDSs, given their ability to improve drug bio-compatibility and physiological stability. Peptide-based DDSs are often used to deliver therapeutic substances such as anti-cancer drugs and nucleic acid-based drugs, which not only slow the degradation rate of drugs in vivo but also ensure the drug concentration at the targeted site and prolong the half-life of drugs in vivo. This article provides an profile of the advancements and future development in functional peptide-based DDSs both domestically and internationally in recent years, in the expectation of achieving targeted drug delivery incorporating functional peptides and taking full advantage of synergistic effects.
Collapse
Affiliation(s)
- Sijie Guo
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Jing Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Qi Wang
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Jinxin Wang
- College of Life Sciences, Yantai University, Yantai, 264005, China
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Wenjun Li
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| |
Collapse
|
44
|
Rizvi SF, Zhang L, Zhang H, Fang Q. Peptide-Drug Conjugates: Design, Chemistry, and Drug Delivery System as a Novel Cancer Theranostic. ACS Pharmacol Transl Sci 2024; 7:309-334. [PMID: 38357281 PMCID: PMC10863443 DOI: 10.1021/acsptsci.3c00269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 02/16/2024]
Abstract
The emergence of peptide-drug conjugates (PDCs) that utilize target-oriented peptide moieties as carriers of cytotoxic payloads, interconnected with various cleavable/noncleavable linkers, resulted in the key-foundation of the new era of targeted therapeutics. They are capable of retaining the integrity of conjugates in the blood circulatory system as well as releasing the drugs at the tumor microenvironment. Other valuable advantages are specificity and selectivity toward targeted-receptors, higher penetration ability, and drug-loading capacity, making them a suitable candidate to play their vital role as promising carrier agents. In this review, we summarized the types of cell-targeting (CTPs) and cell-penetrating peptides (CPPs) that have broad applications in the advancement of targeted drug-delivery systems (DDS). Moreover, the techniques to overcome the limitations of peptide-chemistry for their extensive implementation to construct the PDCs. Besides this, the diversified breakthrough of linker chemistry, and ample knowledge of various cytotoxic payloads used in PDCs in recent years, as well as the mechanism of action of PDCs was critically discussed. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development, also their progression toward a bright future for PDCs as novel theranostics in clinical practice.
Collapse
Affiliation(s)
- Syed Faheem
Askari Rizvi
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, and
Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
- Institute
of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, 54000, Punjab Pakistan
| | - Linjie Zhang
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| | - Haixia Zhang
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| | - Quan Fang
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, and
Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| |
Collapse
|
45
|
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
|
46
|
Mező G, Gomena J, Ranđelović I, Dókus EL, Kiss K, Pethő L, Schuster S, Vári B, Vári-Mező D, Lajkó E, Polgár L, Kőhidai L, Tóvári J, Szabó I. Oxime-Linked Peptide-Daunomycin Conjugates as Good Tools for Selection of Suitable Homing Devices in Targeted Tumor Therapy: An Overview. Int J Mol Sci 2024; 25:1864. [PMID: 38339141 PMCID: PMC10855781 DOI: 10.3390/ijms25031864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Chemotherapy is still one of the main therapeutic approaches in cancer therapy. Nevertheless, its poor selectivity causes severe toxic side effects that, together with the development of drug resistance in tumor cells, results in a limitation for its application. Tumor-targeted drug delivery is a possible choice to overcome these drawbacks. As well as monoclonal antibodies, peptides are promising targeting moieties for drug delivery. However, the development of peptide-drug conjugates (PDCs) is still a big challenge. The main reason is that the conjugates have to be stable in circulation, but the drug or its active metabolite should be released efficiently in the tumor cells. For this purpose, suitable linker systems are needed that connect the drug molecule with the homing peptide. The applied linker systems are commonly categorized as cleavable and non-cleavable linkers. Both the groups possess advantages and disadvantages that are summarized briefly in this manuscript. Moreover, in this review paper, we highlight the benefit of oxime-linked anthracycline-peptide conjugates in the development of PDCs. For instance, straightforward synthesis as well as a conjugation reaction proceed in excellent yields, and the autofluorescence of anthracyclines provides a good tool to select the appropriate homing peptides. Furthermore, we demonstrate that these conjugates can be used properly in in vivo studies. The results indicate that the oxime-linked PDCs are potential candidates for targeted tumor therapy.
Collapse
Affiliation(s)
- Gábor Mező
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Institute of Chemistry, ELTE, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Jacopo Gomena
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Institute of Chemistry, ELTE, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Ivan Ranđelović
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
| | - Endre Levente Dókus
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
| | - Krisztina Kiss
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, 1111 Budapest, Hungary
| | - Lilla Pethő
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
| | - Sabine Schuster
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Institute of Chemistry, ELTE, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Balázs Vári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
- School of Ph.D. Studies, Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Diána Vári-Mező
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
- School of Ph.D. Studies, Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Eszter Lajkó
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary; (E.L.); (L.P.); (L.K.)
| | - Lívia Polgár
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary; (E.L.); (L.P.); (L.K.)
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary; (E.L.); (L.P.); (L.K.)
| | - József Tóvári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122 Budapest, Hungary; (I.R.); (B.V.); (J.T.)
- School of Ph.D. Studies, Doctoral School of Pathological Sciences, Semmelweis University, 1085 Budapest, Hungary
| | - Ildikó Szabó
- HUN-REN-ELTE Research Group of Peptide Chemistry, 1117 Budapest, Hungary; (J.G.); (E.L.D.); (L.P.); (S.S.); (D.V.-M.); (I.S.)
| |
Collapse
|
47
|
Liu X, Cheng Y, Mu Y, Zhang Z, Tian D, Liu Y, Hu X, Wen T. Diverse drug delivery systems for the enhancement of cancer immunotherapy: an overview. Front Immunol 2024; 15:1328145. [PMID: 38298192 PMCID: PMC10828056 DOI: 10.3389/fimmu.2024.1328145] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Despite the clear benefits demonstrated by immunotherapy, there is still an inevitable off-target effect resulting in serious adverse immune reactions. In recent years, the research and development of Drug Delivery System (DDS) has received increased prominence. In decades of development, DDS has demonstrated the ability to deliver drugs in a precisely targeted manner to mitigate side effects and has the advantages of flexible control of drug release, improved pharmacokinetics, and drug distribution. Therefore, we consider that combining cancer immunotherapy with DDS can enhance the anti-tumor ability. In this paper, we provide an overview of the latest drug delivery strategies in cancer immunotherapy and briefly introduce the characteristics of DDS based on nano-carriers (liposomes, polymer nano-micelles, mesoporous silica, extracellular vesicles, etc.) and coupling technology (ADCs, PDCs and targeted protein degradation). Our aim is to show readers a variety of drug delivery platforms under different immune mechanisms, and analyze their advantages and limitations, to provide more superior and accurate targeting strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Xu Liu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Cheng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yao Mu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | | | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yunpeng Liu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ti Wen
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
48
|
Bataille Backer P, Adekiya TA, Kim Y, Reid TER, Thomas M, Adesina SK. Development of a Targeted SN-38-Conjugate for the Treatment of Glioblastoma. ACS OMEGA 2024; 9:2615-2628. [PMID: 38250376 PMCID: PMC10795035 DOI: 10.1021/acsomega.3c07486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024]
Abstract
Glioblastoma (GBM) is the most aggressive and fatal brain tumor, with approximately 10,000 people diagnosed every year in the United States alone. The typical survival period for individuals with glioblastoma ranges from 12 to 18 months, with significant recurrence rates. Common therapeutic modalities for brain tumors are chemotherapy and radiotherapy. The main challenges with chemotherapy for the treatment of glioblastoma are high toxicity, poor selectivity, and limited accumulation of therapeutic anticancer agents in brain tumors as a result of the presence of the blood-brain barrier. To overcome these challenges, researchers have explored strategies involving the combination of targeting peptides possessing a specific affinity for overexpressed cell-surface receptors with conventional chemotherapy agents via the prodrug approach. This approach results in the creation of peptide drug conjugates (PDCs), which facilitate traversal across the blood-brain barrier (BBB), enable preferential accumulation of chemotherapy within the neoplastic microenvironment, and selectively target cancerous cells. This approach increases accumulation in tumors, thereby improving therapeutic efficiency and minimizing toxicity. Leveraging the affinity of the HAIYPRH (T7) peptide for the transferrin receptor (TfR) overexpressed on the blood-brain barrier and glioma cells, a novel T7-SN-38 peptide drug conjugate was developed. The T7-SN-38 peptide drug conjugate demonstrates about a 2-fold reduction in glide score (binding affinity) compared to T7 while maintaining a comparable orientation within the TfR target site using Schrödinger-2022-3 Maestro 13.3 for ligand preparation and Glide SP-Peptide docking. Additionally, SN-38 extends into a solvent-accessible region, enhancing its susceptibility to protease hydrolysis at the cathepsin B (Cat B) cleavable site. The SN-38-ether-peptide drug conjugate displayed high stability in buffer at physiological pH, and cleavage of the conjugate to release free cytotoxic SN-38 was observed in the presence of exogenous cathepsin B. The synthesized peptide drug conjugate exhibited potent cytotoxic activities in cellular models of glioblastoma in vitro. In addition, blocking transferrin receptors using the free T7 peptide resulted in a notable inhibition of cytotoxicity of the conjugate, which was reversed when exogenous cathepsin B was added to cells. This work demonstrates the potential for targeted drug delivery to the brain in the treatment of glioblastoma using the transferrin receptor-targeted T7-SN-38 conjugate.
Collapse
Affiliation(s)
| | - Tayo Alex Adekiya
- Department
of Pharmaceutical Sciences, Howard University, Washington D.C. 20059, United States
| | - Yushin Kim
- Department
of Pharmaceutical Sciences, Concordia University
of Wisconsin, Mequon, Wisconsin 53097-2402, United States
| | - Terry-Elinor R. Reid
- Department
of Pharmaceutical Sciences, Concordia University
of Wisconsin, Mequon, Wisconsin 53097-2402, United States
| | - Michael Thomas
- Department
of Biology, Howard University, Washington D.C. 20059, United States
| | - Simeon K. Adesina
- Department
of Pharmaceutical Sciences, Howard University, Washington D.C. 20059, United States
| |
Collapse
|
49
|
Trencsényi G, Csikos C, Képes Z. Targeted Radium Alpha Therapy in the Era of Nanomedicine: In Vivo Results. Int J Mol Sci 2024; 25:664. [PMID: 38203834 PMCID: PMC10779852 DOI: 10.3390/ijms25010664] [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: 11/01/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Targeted alpha-particle therapy using radionuclides with alpha emission is a rapidly developing area in modern cancer treatment. To selectively deliver alpha-emitting isotopes to tumors, targeting vectors, including monoclonal antibodies, peptides, small molecule inhibitors, or other biomolecules, are attached to them, which ensures specific binding to tumor-related antigens and cell surface receptors. Although earlier studies have already demonstrated the anti-tumor potential of alpha-emitting radium (Ra) isotopes-Radium-223 and Radium-224 (223/224Ra)-in the treatment of skeletal metastases, their inability to complex with target-specific moieties hindered application beyond bone targeting. To exploit the therapeutic gains of Ra across a wider spectrum of cancers, nanoparticles have recently been embraced as carriers to ensure the linkage of 223/224Ra to target-affine vectors. Exemplified by prior findings, Ra was successfully bound to several nano/microparticles, including lanthanum phosphate, nanozeolites, barium sulfate, hydroxyapatite, calcium carbonate, gypsum, celestine, or liposomes. Despite the lengthened tumor retention and the related improvement in the radiotherapeutic effect of 223/224Ra coupled to nanoparticles, the in vivo assessment of the radiolabeled nanoprobes is a prerequisite prior to clinical usage. For this purpose, experimental xenotransplant models of different cancers provide a well-suited scenario. Herein, we summarize the latest achievements with 223/224Ra-doped nanoparticles and related advances in targeted alpha radiotherapy.
Collapse
Affiliation(s)
- György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; (G.T.); (C.C.)
| | - Csaba Csikos
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; (G.T.); (C.C.)
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; (G.T.); (C.C.)
| |
Collapse
|
50
|
Paul T, Palaniyandi K, Gnanasampanthapandian D. Therapeutic Approaches to Increase the Survival Rate of Cancer Patients in the Younger and Older Population. Curr Aging Sci 2024; 17:16-30. [PMID: 38062658 DOI: 10.2174/0118746098241507231127114248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/25/2023] [Accepted: 09/22/2023] [Indexed: 05/18/2024]
Abstract
Various developments have been observed in the treatment of cancer patients, such as higher survival rates and better treatment outcomes. However, expecting similar outcomes in older patients remains a challenge. The main reason for this conclusion is the exclusion of older people from clinical trials for cancer drugs, as well as other factors, such as comorbidity, side effects, age-related frailties and their willingness to undergo multiple treatments. However, the discovery of new techniques and drug combinations has led to a significant improvement in the survival of the elderly population after the onset of the disease. On the other hand, cancer treatments have not become more complex for the younger population when compared to the older population, as the younger population tends to respond well to treatment trials and their physiological conditions are stable in response to treatments. In summary, this review correlates recent cancer treatment strategies and the corresponding responses and survival outcomes of older and younger patients.
Collapse
Affiliation(s)
- Tharrun Paul
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, India
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, India
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, India
| |
Collapse
|