1
|
Morel VJ, Rössler J, Bernasconi M. Targeted immunotherapy and nanomedicine for rhabdomyosarcoma: The way of the future. Med Res Rev 2024; 44:2730-2773. [PMID: 38885148 DOI: 10.1002/med.22059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. Histology separates two main subtypes: embryonal RMS (eRMS; 60%-70%) and alveolar RMS (aRMS; 20%-30%). The aggressive aRMS carry one of two characteristic chromosomal translocations that result in the expression of a PAX3::FOXO1 or PAX7::FOXO1 fusion transcription factor; therefore, aRMS are now classified as fusion-positive (FP) RMS. Embryonal RMS have a better prognosis and are clinically indistinguishable from fusion-negative (FN) RMS. Next to histology and molecular characteristics, RMS risk groupings are now available defining low risk tumors with excellent outcomes and advanced stage disease with poor prognosis, with an overall survival of about only 20% despite intensified multimodal treatment. Therefore, development of novel effective targeted strategies to increase survival and to decrease long-term side effects is urgently needed. Recently, immunotherapies and nanomedicine have been emerging for potent and effective tumor treatments with minimal side effects, raising hopes for effective and safe cures for RMS patients. This review aims to describe the most relevant preclinical and clinical studies in immunotherapy and targeted nanomedicine performed so far in RMS and to provide an insight in future developments.
Collapse
Affiliation(s)
- Victoria Judith Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Dzhumashev D, Anton-Joseph S, Morel VJ, Timpanaro A, Bordon G, Piccand C, Aleandri S, Luciani P, Rössler J, Bernasconi M. Rapid liposomal formulation for nucleolin targeting to rhabdomyosarcoma cells. Eur J Pharm Biopharm 2024; 194:49-61. [PMID: 38029941 DOI: 10.1016/j.ejpb.2023.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. More effective and less toxic therapies are urgently needed for high-risk patients. Peptide-guided targeted drug delivery can increase the therapeutic index of encapsulated drugs and improve patients' well-being. To apply this strategy to RMS, we identified the peptide F3 in a screening for peptides binding to RMS cells surface. F3 binds to nucleolin, which is present on the surface of RMS cells and is abundantly expressed at the mRNA level in RMS patients' biopsies compared to healthy tissues. We developed a rapid microfluidic formulation of F3-decorated PEGylated liposomes and remote loading of the chemotherapeutic drug vincristine. Size, surface charge, drug loading and retention of targeted and control liposomes were studied. Enhanced cellular binding and uptake were observed in three different nucleolin-positive RMS cell lines. Importantly, F3-functionalized liposomes loaded with vincristine were up to 11 times more cytotoxic than non-targeted liposomes for RMS cell lines. These results demonstrate that F3-functionalized liposomes are promising for targeted drug delivery to RMS and warrant further in vivo investigations.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Stenija Anton-Joseph
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Victoria J Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Caroline Piccand
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland.
| |
Collapse
|
3
|
Zarrabi A, Perrin D, Kavoosi M, Sommer M, Sezen S, Mehrbod P, Bhushan B, Machaj F, Rosik J, Kawalec P, Afifi S, Bolandi SM, Koleini P, Taheri M, Madrakian T, Łos MJ, Lindsey B, Cakir N, Zarepour A, Hushmandi K, Fallah A, Koc B, Khosravi A, Ahmadi M, Logue S, Orive G, Pecic S, Gordon JW, Ghavami S. Rhabdomyosarcoma: Current Therapy, Challenges, and Future Approaches to Treatment Strategies. Cancers (Basel) 2023; 15:5269. [PMID: 37958442 PMCID: PMC10650215 DOI: 10.3390/cancers15215269] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Rhabdomyosarcoma is a rare cancer arising in skeletal muscle that typically impacts children and young adults. It is a worldwide challenge in child health as treatment outcomes for metastatic and recurrent disease still pose a major concern for both basic and clinical scientists. The treatment strategies for rhabdomyosarcoma include multi-agent chemotherapies after surgical resection with or without ionization radiotherapy. In this comprehensive review, we first provide a detailed clinical understanding of rhabdomyosarcoma including its classification and subtypes, diagnosis, and treatment strategies. Later, we focus on chemotherapy strategies for this childhood sarcoma and discuss the impact of three mechanisms that are involved in the chemotherapy response including apoptosis, macro-autophagy, and the unfolded protein response. Finally, we discuss in vivo mouse and zebrafish models and in vitro three-dimensional bioengineering models of rhabdomyosarcoma to screen future therapeutic approaches and promote muscle regeneration.
Collapse
Affiliation(s)
- Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - David Perrin
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Micah Sommer
- Section of Orthopaedic Surgery, Department of Surgery, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; (D.P.); (M.S.)
- Section of Physical Medicine and Rehabilitation, Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Parvaneh Mehrbod
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Bhavya Bhushan
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Science, McGill University, Montreal, QC H3A 0C7, Canada
| | - Filip Machaj
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jakub Rosik
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Philip Kawalec
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Health Sciences Centre, Winnipeg, MB R3A 1R9, Canada
| | - Saba Afifi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Seyed Mohammadreza Bolandi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Peiman Koleini
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Mohsen Taheri
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland;
| | - Benjamin Lindsey
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Nilufer Cakir
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Türkiye; (A.Z.); (A.Z.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Ali Fallah
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
| | - Bahattin Koc
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Türkiye; (S.S.); (N.C.); (B.K.)
- Integrated Manufacturing Technologies Research and Application Center, Sabanci University, Tuzla, Istanbul 34956, Türkiye;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Türkiye
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (T.M.); (M.A.)
| | - Susan Logue
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), 01007 Vitoria-Gasteiz, Spain;
- University Institute for Regenerative Medicine and Oral Implantology–UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, USA;
| | - Joseph W. Gordon
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- College of Nursing, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; (M.K.); (B.B.); (F.M.); (J.R.); (P.K.); (S.A.); (S.M.B.); (P.K.); (B.L.); (S.L.); (J.W.G.)
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555 Katowice, Poland
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
4
|
Dzhumashev D, Timpanaro A, Ali S, De Micheli AJ, Mamchaoui K, Cascone I, Rössler J, Bernasconi M. Quantum Dot-Based Screening Identifies F3 Peptide and Reveals Cell Surface Nucleolin as a Therapeutic Target for Rhabdomyosarcoma. Cancers (Basel) 2022; 14:5048. [PMID: 36291832 PMCID: PMC9600270 DOI: 10.3390/cancers14205048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Active drug delivery by tumor-targeting peptides is a promising approach to improve existing therapies for rhabdomyosarcoma (RMS), by increasing the therapeutic effect and decreasing the systemic toxicity, e.g., by drug-loaded peptide-targeted nanoparticles. Here, we tested 20 different tumor-targeting peptides for their ability to bind to two RMS cell lines, Rh30 and RD, using quantum dots Streptavidin and biotin-peptides conjugates as a model for nanoparticles. Four peptides revealed a very strong binding to RMS cells: NCAM-1-targeting NTP peptide, nucleolin-targeting F3 peptide, and two Furin-targeting peptides, TmR and shTmR. F3 peptide showed the strongest binding to all RMS cell lines tested, low binding to normal control myoblasts and fibroblasts, and efficient internalization into RMS cells demonstrated by the cytoplasmic delivery of the Saporin toxin. The expression of the nucleophosphoprotein nucleolin, the target of F3, on the surface of RMS cell lines was validated by competition with the natural ligand lactoferrin, by colocalization with the nucleolin-binding aptamer AS1411, and by the marked sensitivity of RMS cell lines to the growth inhibitory nucleolin-binding N6L pseudopeptide. Taken together, our results indicate that nucleolin-targeting by F3 peptide represents a potential therapeutic approach for RMS.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Safa Ali
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Andrea J. De Micheli
- Department of Oncology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| | - Kamel Mamchaoui
- Centre de Recherche en Myologie, Institut de Myologie, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Ilaria Cascone
- IMRB, INSERM, University Paris Est Creteil, 94010 Creteil, France
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biothérapie, 94010 Créteil, France
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| |
Collapse
|
5
|
van der Koog L, Gandek TB, Nagelkerke A. Liposomes and Extracellular Vesicles as Drug Delivery Systems: A Comparison of Composition, Pharmacokinetics, and Functionalization. Adv Healthc Mater 2022; 11:e2100639. [PMID: 34165909 PMCID: PMC11468589 DOI: 10.1002/adhm.202100639] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Over the past decades, lipid-based nanoparticle drug delivery systems (DDS) have caught the attention of researchers worldwide, encouraging the field to rapidly develop improved ways for effective drug delivery. One of the most prominent examples is liposomes, which are spherical shaped artificial vesicles composed of lipid bilayers and able to encapsulate both hydrophilic and hydrophobic materials. At the same time, biological nanoparticles naturally secreted by cells, called extracellular vesicles (EVs), have emerged as promising more complex biocompatible DDS. In this review paper, the differences and similarities in the composition of both vesicles are evaluated, and critical mediators that affect their pharmacokinetics are elucidate. Different strategies that have been assessed to tweak the pharmacokinetics of both liposomes and EVs are explored, detailing the effects on circulation time, targeting capacity, and cytoplasmic delivery of therapeutic cargo. Finally, whether a hybrid system, consisting of a combination of only the critical constituents of both vesicles, could offer the best of both worlds is discussed. Through these topics, novel leads for further research are provided and, more importantly, gain insight in what the liposome field and the EV field can learn from each other.
Collapse
Affiliation(s)
- Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyGRIAC Research Institute, University Medical Center GroningenUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
6
|
Protease-triggered bioresponsive drug delivery for the targeted theranostics of malignancy. Acta Pharm Sin B 2021; 11:2220-2242. [PMID: 34522585 PMCID: PMC8424222 DOI: 10.1016/j.apsb.2021.01.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Proteases have a fundamental role in maintaining physiological homeostasis, but their dysregulation results in severe activity imbalance and pathological conditions, including cancer onset, progression, invasion, and metastasis. This striking importance plus superior biological recognition and catalytic performance of proteases, combining with the excellent physicochemical characteristics of nanomaterials, results in enzyme-activated nano-drug delivery systems (nanoDDS) that perform theranostic functions in highly specific response to the tumor phenotype stimulus. In the tutorial review, the key advances of protease-responsive nanoDDS in the specific diagnosis and targeted treatment for malignancies are emphatically classified according to the effector biomolecule types, on the premise of summarizing the structure and function of each protease. Subsequently, the incomplete matching and recognition between enzyme and substrate, structural design complexity, volume production, and toxicological issues related to the nanocomposites are highlighted to clarify the direction of efforts in nanotheranostics. This will facilitate the promotion of nanotechnology in the management of malignant tumors.
Collapse
|
7
|
Weber F, Casalini T, Valentino G, Brülisauer L, Andreas N, Koeberle A, Kamradt T, Contini A, Luciani P. Targeting transdifferentiated hepatic stellate cells and monitoring the hepatic fibrogenic process by means of IGF2R-specific peptides designed in silico. J Mater Chem B 2021; 9:2092-2106. [PMID: 33595041 DOI: 10.1039/d0tb02372h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The lack of accurate and easily applicable methods for the diagnosis of liver fibrosis, a disease characterized by an accumulation of the extracellular matrix released by activated hepatic stellate cells (HSCs), has been a major limitation for the clinical management of liver diseases. The identification of biomarkers specific to liver microstructure alterations, combined with a non-invasive optical imaging modality, could guide clinicians towards a therapeutic strategy. In this study, structural information of the insulin-like growth factor 2 receptor (IGF2R), an overexpressed protein on activated HSCs, was used for in silico screening of novel IGF2R-specific peptide ligands. Molecular dynamics simulations, followed by computational alanine scanning of the IGF2R/IGF2 complex, led to the identification of a putative peptide sequence containing the most relevant amino acids for the receptor-ligand interaction (IGF2 E12-C21). The Residue Scan tool, implemented in the MOE software, was then used to optimize the binding affinity of this sequence by amino acid mutations. The designed peptides and their associated scrambled sequences were fluorescently labelled and their binding affinity to LX-2 cells (model for activated human HSCs) was tested using flow cytometry and confocal microscopy. In vitro binding was verified for all sequences (KD ≤ 13.2 μM). With respect to the putative binding sequence, most mutations led to an increased affinity. All sequences have shown superior binding compared to their associated scrambled sequences. Using HPLC, all peptides were tested in vitro for their proteolytic resistance and showed a stability of ≥60% intact after 24 h at 37 °C in 50% v/v FBS. In view of their prospective diagnostic application, a comparison of binding affinity was performed in perpetuated and quiescent-like LX-2 cells. Furthermore, the IGF2R expression for different cell phenotypes was analysed by a quantitative mass spectrometric approach. Our peptides showed increased binding to the perpetuated cell state, indicating their good selectivity for the diagnostically relevant phenotype. In summary, the increased binding affinity of our peptides towards perpetuated LX-2 cells, as well as the satisfactory proteolytic stability, proves that the in silico designed sequences offer a new potential strategy for the targeting of hepatic fibrosis.
Collapse
Affiliation(s)
- Florian Weber
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland. and Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Tommaso Casalini
- Institute of Mechanical Engineering and Material Technology, Department of Innovative Technology, SUPSI, Manno, Switzerland and Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Gina Valentino
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland. and Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Lorine Brülisauer
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital, Jena, Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria and Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Thomas Kamradt
- Institute of Immunology, Jena University Hospital, Jena, Germany
| | - Alessandro Contini
- Dipartimento di Scienze Farmaceutiche-Sezione di Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milano, Italy
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland. and Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
8
|
Zhang X, Shao X, Cai Z, Yan X, Zong W. The fabrication of phospholipid vesicle-based artificial cells and their functions. NEW J CHEM 2021. [DOI: 10.1039/d0nj05538g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Phospholipid vesicles as artificial cells are used to simulate the cellular structure and function.
Collapse
Affiliation(s)
- Xunan Zhang
- College of Chemistry and Chemical Engineering
- Qiqihar University
- Qiqihar
- China
| | - Xiaotong Shao
- College of Chemistry and Chemical Engineering
- Qiqihar University
- Qiqihar
- China
| | - Zhenzhen Cai
- College of Chemistry and Chemical Engineering
- Qiqihar University
- Qiqihar
- China
| | - Xinyu Yan
- College of Chemistry and Chemical Engineering
- Qiqihar University
- Qiqihar
- China
| | - Wei Zong
- College of Chemistry and Chemical Engineering
- Qiqihar University
- Qiqihar
- China
| |
Collapse
|
9
|
Drescher S, van Hoogevest P. The Phospholipid Research Center: Current Research in Phospholipids and Their Use in Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12121235. [PMID: 33353254 PMCID: PMC7766331 DOI: 10.3390/pharmaceutics12121235] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the research on phospholipids and their use for drug delivery related to the Phospholipid Research Center Heidelberg (PRC). The focus is on projects that have been approved by the PRC since 2017 and are currently still ongoing or have recently been completed. The different projects cover all facets of phospholipid research, from basic to applied research, including the use of phospholipids in different administration forms such as liposomes, mixed micelles, emulsions, and extrudates, up to industrial application-oriented research. These projects also include all routes of administration, namely parenteral, oral, and topical. With this review we would like to highlight possible future research directions, including a short introduction into the world of phospholipids.
Collapse
|
10
|
Alijaj N, Moutel S, Gouveia ZL, Gray M, Roveri M, Dzhumashev D, Weber F, Meier G, Luciani P, Rössler JK, Schäfer BW, Perez F, Bernasconi M. Novel FGFR4-Targeting Single-Domain Antibodies for Multiple Targeted Therapies against Rhabdomyosarcoma. Cancers (Basel) 2020; 12:cancers12113313. [PMID: 33182650 PMCID: PMC7696840 DOI: 10.3390/cancers12113313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Rhabdomyosarcoma (RMS) accounts for more than 50% of all soft tissue sarcomas in childhood and adolescence. Despite progress and intensified multimodality treatment, prognoses are extremely poor with an overall survival rate of approximately 20% in the advanced stage. Therefore, there is an urgent need for targeted treatment options to improve overall survival rates, and to limit long-term side effects. The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in RMS and other tumors as well. The goal of this work was to select FGFR4 specific single-domain antibodies (sdAb) and to develop FGFR4-targeted therapies. We could show that FGFR4-targeted liposomes have the potential to deliver drugs specifically to FGFR4-positive tumor cells and that chimeric antigen receptor T cells built with the selected antibodies can kill specifically FGFR4-expressing RMS cells. Abstract The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in rhabdomyosarcoma (RMS) and represents a promising target for treatments based on specific and efficient antibodies. Despite progress, there is an urgent need for targeted treatment options to improve survival rates, and to limit long-term side effects. From phage display libraries we selected FGFR4-specific single-domain antibodies (sdAb) binding to recombinant FGFR4 and validated them by flow cytometry, surface plasmon resonance, and fluorescence microscopy. The specificity of the selected sdAb was verified on FGFR4-wild type and FGFR4-knock out cells. FGFR4-sdAb were used to decorate vincristine-loaded liposomes and to generate chimeric antigen receptor (CAR) T cells. First, incubation of RMS cells with FGFR4-sdAb revealed that FGFR4-sdAb can block FGF19-FGFR4 signaling via the MAPK pathway and could therefore serve as therapeutics for FGFR4-dependent cancers. Second, FGFR4-targeted vincristine-loaded liposomes bound specifically to RMS cells and were internalized by the receptor, demonstrating the potential for active drug delivery to the tumor. Third, FGFR4-CAR T cells, generated with one sdAb candidate, demonstrated strong and specific cytotoxicity against FGFR4 expressing RMS cells. We selected novel FGFR4-sdAb with high specificity and nano- to picomolar affinities for FGFR4 which have the potential to enable multiple FGFR4-targeted cancer therapy approaches.
Collapse
Affiliation(s)
- Nagjie Alijaj
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Sandrine Moutel
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Recombinant Antibody Platform (TAb-IP), Institut Curie, 75248 Paris, France
| | - Zelia L. Gouveia
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Honing Biosciences, 75004 Paris, France
| | - Maxim Gray
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Maurizio Roveri
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Florian Weber
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland; (F.W.); (P.L.)
| | - Gianmarco Meier
- Institute of Medical Microbiology, University of Zurich, 8006 Zurich, Switzerland;
| | - Paola Luciani
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland; (F.W.); (P.L.)
| | - Jochen K. Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Beat W. Schäfer
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Franck Perez
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Correspondence: (F.P.); (M.B.)
| | - Michele Bernasconi
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Correspondence: (F.P.); (M.B.)
| |
Collapse
|
11
|
Daurat M, Nguyen C, Dominguez Gil S, Sol V, Chaleix V, Charnay C, Raehm L, El Cheikh K, Morère A, Bernasconi M, Timpanaro A, Garcia M, Cunin F, Roessler J, Durand JO, Gary-Bobo M. The mannose 6-phosphate receptor targeted with porphyrin-based periodic mesoporous organosilica nanoparticles for rhabdomyosarcoma theranostics. Biomater Sci 2020; 8:3678-3684. [DOI: 10.1039/d0bm00586j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Periodic mesoporous organosilica nanoparticles targeted to the mannose 6-phosphate receptor for rhabdomyosarcoma photodynamic therapy using a pulsed laser.
Collapse
Affiliation(s)
- Morgane Daurat
- NanoMedSyn Avenue Charles Flahault
- Montpellier Cedex 05
- France
| | | | | | - Vincent Sol
- Laboratoire PEIRENE
- EA 7500
- Université de Limoges
- Faculté des Sciences et Techniques
- 87060 Limoges
| | - Vincent Chaleix
- Laboratoire PEIRENE
- EA 7500
- Université de Limoges
- Faculté des Sciences et Techniques
- 87060 Limoges
| | | | | | | | - Alain Morère
- IBMM
- UMR 5247 CNRS
- UM-Faculté de Pharmacie
- Montpellier Cedex 05
- France
| | - Michele Bernasconi
- Division of Pediatric Hematology/Oncology
- Department of Pediatrics
- Inselspital
- Bern University Hospital
- Department for BioMedical Research (DBMR)
| | - Andrea Timpanaro
- Division of Pediatric Hematology/Oncology
- Department of Pediatrics
- Inselspital
- Bern University Hospital
- Department for BioMedical Research (DBMR)
| | - Marcel Garcia
- NanoMedSyn Avenue Charles Flahault
- Montpellier Cedex 05
- France
| | | | - Jochen Roessler
- Division of Pediatric Hematology/Oncology
- Department of Pediatrics
- Inselspital
- Bern University Hospital
- Department for BioMedical Research (DBMR)
| | | | - Magali Gary-Bobo
- IBMM
- UMR 5247 CNRS
- UM-Faculté de Pharmacie
- Montpellier Cedex 05
- France
| |
Collapse
|
12
|
Wang D, Sun Y, Liu Y, Meng F, Lee RJ. Clinical translation of immunoliposomes for cancer therapy: recent perspectives. Expert Opin Drug Deliv 2018; 15:893-903. [PMID: 30169978 DOI: 10.1080/17425247.2018.1517747] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Liposomes have been extensively investigated as drug delivery vehicles. Immunoliposomes (ILs) are antibody-conjugated liposomes designed to selectively target antigen-expressing cells. ILs can be used to deliver drugs to tumor cells for improving efficacy and reducing toxicity. In addition, ILs can be used in immunoassays, immunotherapy, and imaging. Although there has been extensive coverage on ILs in the literature, only a limited number of clinical trials have been reported and no IL drug has been approved by the FDA. AREAS COVERED Factors to consider in developing ILs are discussed, including the choice of antibody or antibody fragment, the formulation of liposomes, and the conjugation chemistry. In addition, challenges and opportunities in clinical development of ILs are discussed. The purpose of this review is to provide an overview on the state of the art of ILs and to discuss potential future developments. EXPERT OPINION IL research has had a lengthy history and numerous preclinical studies have yielded encouraging results. However, there are a number of obstacles to clinical translation of ILs. Given the unique capabilities of ILs, its potential for clinical application is underexplored. There is great potential for expanded role for ILs in the clinic and further efforts to this end are warranted. ABBREVIATIONS Ab: antibody; ADCs: antibody-drug conjugates; API: active pharmaceutical ingredient; ADCC: antibody-dependent cellular cytotoxicity; CR: complete remission; cGMP: current good manufacturing practice; DSPE: distearoyl phosphatidylethanolamine; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; EPR: enhanced permeability and retention; Fc: fragment crystalline; Tf: transferrin; HACA: human-anti-chimeric antibody; HAHA: human-anti-human antibody; HAMA: human-anti-mouse antibody; HER2: human epidermal growth factor 2; IL: immunoliposome; LNPs: lipid nanoparticles; MRI: magnetic resonance imaging; MTD: maximum tolerated dose; PEG: polyethylene glycol; PET: positron emission tomography; PR: partial response; PSMA: prostate-specific membrane antigen; scFv: single-chain variable fragment; SPECT: single photon emission computed tomography; TTR: transthyretin.
Collapse
Affiliation(s)
- Di Wang
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yating Sun
- a School of Life Sciences , Jilin University , Changchun , China
| | - Yange Liu
- a School of Life Sciences , Jilin University , Changchun , China
| | - Fanchao Meng
- a School of Life Sciences , Jilin University , Changchun , China
| | - Robert J Lee
- a School of Life Sciences , Jilin University , Changchun , China.,b Division of Pharmaceutics and Pharmaceutical Chemistry , The Ohio State University , Columbus , OH , USA
| |
Collapse
|