1
|
Theivendren P, Kunjiappan S, Pavadai P, Ravi K, Murugavel A, Dayalan A, Santhana Krishna Kumar A. Revolutionizing Cancer Immunotherapy: Emerging Nanotechnology-Driven Drug Delivery Systems for Enhanced Therapeutic Efficacy. ACS MEASUREMENT SCIENCE AU 2025; 5:31-55. [PMID: 39991031 PMCID: PMC11843507 DOI: 10.1021/acsmeasuresciau.4c00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 02/25/2025]
Abstract
Cancer immunotherapy is an innovative way of treating cancer by stimulating individual cells to overcome cancer. Widespread biomedical studies were carried out with the aim of exploring immunotherapy cancer therapeutics, and this review spotlights some mechanisms in which it was developed, namely immune checkpoint inhibitors (E.G PD-1/PD-L1, CTLA-4), adoptive cell therapy (e.g., CAR T-cell therapy), and cancer vaccines. Although it has shown clinical benefit in a number of cancer types, including melanoma and non-small-cell lung cancer, several challenges have dampened enthusiasm for this approach, from the differing patient response rates to toxicities. Nanotechnology in drug delivery systems must play a role in overcoming the same. Nanotechnology enables increased specificity and controlled drug release, improved solubility and bioavailability, can treat the tumor specifically, and localized drug delivery at the disease site decreases systemic toxicity. The review also features advances in the construction of lipid-based, polymeric, and inorganic nanoparticles that improve drug stability and allow the delivery of cotherapeutic agents. Nanotechnology-based delivery systems can be used alone or in combination with immunotherapy to assist in improving the immune response, gaining access to the tumor microenvironment, and overcoming biological barriers. Thus, the nano-DDS were both safe and effective in preclinical studies, and ongoing clinical trials have shown that they are capable of increasing the therapeutic index of anticancer drugs. Lastly, the review also discusses current challenges and regulatory issues in advancing these technologies and highlights the importance of further research to devise appropriate methodology for efficient functionalization of nanotechnology for individualized cancer solutions in cancer treatment.
Collapse
Affiliation(s)
- Panneerselvam Theivendren
- Department
of Pharmaceutical Chemistry, Swamy Vivekanandha
College of Pharmacy, Elayampalayam 637205, Namakkal, Tamil Nadu, India
| | - Selvaraj Kunjiappan
- Department
of Biotechnology, Kalasalingam Academy of
Research and Education, Krishnankoil 626126, Tamil Nadu, India
| | - Parasuraman Pavadai
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M. S. R. Nagar, Bengaluru 560054, Karnataka, India
| | - Kaveena Ravi
- Department
of Pharmaceutics, Swamy Vivekananda College
of Pharmacy, Elayampalayam 637205, Namakkal, Tamil Nadu, India
| | - Anusuya Murugavel
- Center
for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Avinash Dayalan
- Center
for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - A. Santhana Krishna Kumar
- Department
of Chemistry, National Sun Yat-sen University, No. 70, Lien-hai Road, Gushan District, Kaohsiung City 80424, Taiwan
- Department
of Chemistry, Saveetha School of Engineering, Saveetha Institute of
Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 602105, Tamil Nadu, India
| |
Collapse
|
2
|
Taha E, Nour SA, Mamdouh W, Naguib MJ. Investigating the potential of highly porous zopiclone-loaded 3D electrospun nanofibers for brain targeting via the intranasal route. Int J Pharm 2024; 660:124230. [PMID: 38782156 DOI: 10.1016/j.ijpharm.2024.124230] [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: 01/27/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Nanofibers (NFs) have proven to be very attractive tool as drug delivery plateform among the different plethora of nanosystems, owing to their unique features. They exhibit two- and three-dimensional structures some of which mimic structural environment of the body tissues, in addition to being safe, efficacious, and biocompatible drug delivery platform. Thus, this study embarked on fabricating polyvinyl alcohol/chitosan (PVA/CS) electrospun NFs encapsulating zopiclone (ZP) drug for intranasal brain targeted drug delivery. Electrospun NFs were optimized by adopting a three factor-two level full factorial design. The independent variables were: PVA/CS ratio (X1), flow rate (X2), and applied voltage (X3). The measured responses were: fiber diameter (Y1,nm), pore size (Y2,nm) and ultimate tensile strength (UTS,Y3,MPa). The selected optimum formula had resulted in NFs diameter of 215.90 ± 15.46 nm, pore size 7.12 ± 0.27 nm, and tensile strength around 6.64 ± 0.95 MPa. In-vitro biodegradability testing confirmed proper degradation of the NFs within 8 h. Moreover, swellability and breathability assessment revealed good hydrophilicity and permeability of the prepared NFs. Ex-vivo permeability study declared boosted ex-vivo permeation with an enhancement factor of 2.73 compared to ZP suspension. In addition, optimized NFs formula significantly reduced sleep latency and prolonged sleep duration in rats compared to IV ZP drug solution. These findings demonstrate the feasibility of employing the designed NFs as an effective safe platform for intranasal delivery of ZP for insomnia management.
Collapse
Affiliation(s)
- Esraa Taha
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Samia A Nour
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Wael Mamdouh
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo 11835, Egypt.
| | - Marianne J Naguib
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
3
|
Baranwal J, Barse B, Di Petrillo A, Gatto G, Pilia L, Kumar A. Nanoparticles in Cancer Diagnosis and Treatment. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5354. [PMID: 37570057 PMCID: PMC10420054 DOI: 10.3390/ma16155354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
The use of tailored medication delivery in cancer treatment has the potential to increase efficacy while decreasing unfavourable side effects. For researchers looking to improve clinical outcomes, chemotherapy for cancer continues to be the most challenging topic. Cancer is one of the worst illnesses despite the limits of current cancer therapies. New anticancer medications are therefore required to treat cancer. Nanotechnology has revolutionized medical research with new and improved materials for biomedical applications, with a particular focus on therapy and diagnostics. In cancer research, the application of metal nanoparticles as substitute chemotherapy drugs is growing. Metals exhibit inherent or surface-induced anticancer properties, making metallic nanoparticles extremely useful. The development of metal nanoparticles is proceeding rapidly and in many directions, offering alternative therapeutic strategies and improving outcomes for many cancer treatments. This review aimed to present the most commonly used nanoparticles for cancer applications.
Collapse
Affiliation(s)
- Jaya Baranwal
- DBT-ICGEB Centre for Advanced Bioenergy Research, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Brajesh Barse
- US India Business Council|US Chamber of Commerce, DLF Centre, Sansad Marg, New Delhi 110001, India
| | - Amalia Di Petrillo
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042 Cagliari, Italy;
| | - Gianluca Gatto
- Department of Electrical and Electronic Engineering, University of Cagliari, Via Marengo 2, 09123 Cagliari, Italy;
| | - Luca Pilia
- Department of Mechanical, Chemical and Material Engineering, University of Cagliari, Via Marengo 2, 09123 Cagliari, Italy
| | - Amit Kumar
- Department of Electrical and Electronic Engineering, University of Cagliari, Via Marengo 2, 09123 Cagliari, Italy;
| |
Collapse
|
4
|
Verkhovskii RA, Ivanov AN, Lengert EV, Tulyakova KA, Shilyagina NY, Ermakov AV. Current Principles, Challenges, and New Metrics in pH-Responsive Drug Delivery Systems for Systemic Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051566. [PMID: 37242807 DOI: 10.3390/pharmaceutics15051566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 05/28/2023] Open
Abstract
The paradigm of drug delivery via particulate formulations is one of the leading ideas that enable overcoming limitations of traditional chemotherapeutic agents. The trend toward more complex multifunctional drug carriers is well-traced in the literature. Nowadays, the prospectiveness of stimuli-responsive systems capable of controlled cargo release in the lesion nidus is widely accepted. Both endogenous and exogenous stimuli are employed for this purpose; however, endogenous pH is the most common trigger. Unfortunately, scientists encounter multiple challenges on the way to the implementation of this idea related to the vehicles' accumulation in off-target tissues, their immunogenicity, the complexity of drug delivery to intracellular targets, and finally, the difficulties in the fabrication of carriers matching all imposed requirements. Here, we discuss fundamental strategies for pH-responsive drug delivery, as well as limitations related to such carriers' application, and reveal the main problems, weaknesses, and reasons for poor clinical results. Moreover, we attempted to formulate the profiles of an "ideal" drug carrier in the frame of different strategies drawing on the example of metal-comprising materials and considered recently published studies through the lens of these profiles. We believe that this approach will facilitate the formulation of the main challenges facing researchers and the identification of the most promising trends in technology development.
Collapse
Affiliation(s)
- Roman A Verkhovskii
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia
| | - Alexey N Ivanov
- Central Research Laboratory, Saratov State Medical University of V. I. Razumovsky, Ministry of Health of the Russian Federation, 410012 Saratov, Russia
| | - Ekaterina V Lengert
- Central Research Laboratory, Saratov State Medical University of V. I. Razumovsky, Ministry of Health of the Russian Federation, 410012 Saratov, Russia
- Institute of Molecular Theranostics, I. M. Sechenov First Moscow State Medical University, 8 Trubetskaya Str., 119991 Moscow, Russia
| | - Ksenia A Tulyakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Natalia Yu Shilyagina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Alexey V Ermakov
- Central Research Laboratory, Saratov State Medical University of V. I. Razumovsky, Ministry of Health of the Russian Federation, 410012 Saratov, Russia
- Institute of Molecular Theranostics, I. M. Sechenov First Moscow State Medical University, 8 Trubetskaya Str., 119991 Moscow, Russia
| |
Collapse
|
5
|
Exploring uterine targeting potential of 99mTc-Paclitaxel loaded ultradeformable vesicles designed for endometrial cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
6
|
Petrazzuolo A, Maiuri MC, Zitvogel L, Kroemer G, Kepp O. Trial Watch: combination of tyrosine kinase inhibitors (TKIs) and immunotherapy. Oncoimmunology 2022; 11:2077898. [PMID: 35655707 PMCID: PMC9154809 DOI: 10.1080/2162402x.2022.2077898] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The past decades witnessed the clinical employment of targeted therapies including but not limited to tyrosine kinase inhibitors (TKIs) that restrain a broad variety of pro-tumorigenic signals. TKIs can be categorized into (i) agents that directly target cancer cells, (ii) normalize angiogenesis or (iii) affect cells of the hematologic lineage. However, a clear distinction of TKIs based on this definition is limited by the fact that many TKIs designed to inhibit cancer cells have also effects on immune cells that are being discovered. Additionally, TKIs originally designed to target hematological cancers exhibit bioactivities on healthy cells of the same hematological lineage. TKIs have been described to improve immune recognition and cancer immunosurveillance, providing the scientific basis to combine TKIs with immunotherapy. Indeed, combination of TKIs with immunotherapy showed synergistic effects in preclinical models and clinical trials and some combinations of TKIs normalizing angiogenesis with immune checkpoint blocking antibodies have already been approved by the FDA for cancer therapy. However, the identification of appropriate drug combinations as well as optimal dosing and scheduling needs to be improved in order to obtain tangible progress in cancer care. This Trial Watch summarizes active clinical trials combining TKIs with various immunotherapeutic strategies to treat cancer patients.
Collapse
Affiliation(s)
- Adriana Petrazzuolo
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - M. Chiara Maiuri
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Faculty of Medicine, University Paris Saclay, Kremlin Bicêtre, France
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Institut National de la Santé et de la Recherche Medicale (INSERM) U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) Biotheris 1428, Villejuif, France
| | - Guido Kroemer
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Oliver Kepp
- Team “Metabolism, Cancer & Immunity”, Centre de Recherche des Cordeliers, INSERM UMRS1138, Université Paris Cité, Sorbonne Université, Paris, France
- Cell Biology and Metabolomics platforms, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
7
|
Alshahrani SM. A judicious review on the applications of chemotherapeutic loaded nanoemulsions in cancer management. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
8
|
Jose J, Kanniyappan H, Muthuvijayan V. A novel, rapid and cost-effective method for separating drug-loaded liposomes prepared from egg yolk phospholipids. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
9
|
Giraldo E, Nebot VJ, Đorđević S, Requejo-Aguilar R, Alastrue-Agudo A, Zagorodko O, Armiñan A, Martinez-Rojas B, Vicent MJ, Moreno-Manzano V. A rationally designed self-immolative linker enhances the synergism between a polymer-rock inhibitor conjugate and neural progenitor cells in the treatment of spinal cord injury. Biomaterials 2021; 276:121052. [PMID: 34388362 DOI: 10.1016/j.biomaterials.2021.121052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/04/2021] [Accepted: 07/24/2021] [Indexed: 10/24/2022]
Abstract
Rho/ROCK signaling induced after spinal cord injury (SCI) contributes to secondary damage by promoting apoptosis, inflammation, and axon growth inhibition. The specific Rho-kinase inhibitor fasudil can contribute to functional regeneration after SCI, although inherent low stability has hampered its use. To improve the therapeutic potential of fasudil, we now describe a family of rationally-designed bioresponsive polymer-fasudil conjugates based on an understanding of the conditions after SCI, such as low pH, enhanced expression of specific proteases, and a reductive environment. Fasudil conjugated to poly-l-glutamate via a self-immolative redox-sensitive linker (PGA-SS-F) displays optimal release kinetics and, consequently, treatment with PGA-SS-F significantly induces neurite elongation and axon growth in dorsal root ganglia explants, spinal cord organotypic cultures, and neural precursor cells (NPCs). The intrathecal administration of PGA-SS-F after SCI in a rat model prevents early apoptosis and induces the expression of axonal growth- and neuroplasticity-associated markers to a higher extent than the free form of fasudil. Moreover, a combination treatment comprising the acute transplantation of NPCs pre-treated with PGA-SS-F leads to enhanced cell engraftment and reduced cyst formation after SCI. In chronic SCI, combinatory treatment increases the preservation of neuronal fibers. Overall, this synergistic combinatorial strategy may represent a potentially efficient clinical approach to SCI treatment.
Collapse
Affiliation(s)
- E Giraldo
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain; Department of Biotechnology. Universitat Politècnica de València, Valencia, Spain
| | - V J Nebot
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain; PTS S.L., Valencia, Spain
| | - S Đorđević
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - R Requejo-Aguilar
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain; Dept. Biochemistry and Molecular Biology, University of Cordoba, Cordoba, Spain. Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Cordoba, Spain
| | - A Alastrue-Agudo
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain
| | - O Zagorodko
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - A Armiñan
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain
| | - B Martinez-Rojas
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain
| | - M J Vicent
- Polymer Therapeutics Lab. Prince Felipe Research Institute, Valencia, Spain.
| | - V Moreno-Manzano
- Neuronal and Tissue Regeneration Lab. Prince Felipe Research Institute, Valencia, Spain.
| |
Collapse
|
10
|
Butowska K, Woziwodzka A, Borowik A, Piosik J. Polymeric Nanocarriers: A Transformation in Doxorubicin Therapies. MATERIALS (BASEL, SWITZERLAND) 2021; 14:2135. [PMID: 33922291 PMCID: PMC8122860 DOI: 10.3390/ma14092135] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
Doxorubicin, a member of the anthracycline family, is a common anticancer agent often used as a first line treatment for the wide spectrum of cancers. Doxorubicin-based chemotherapy, although effective, is associated with serious side effects, such as irreversible cardiotoxicity or nephrotoxicity. Those often life-threatening adverse risks, responsible for the elongation of the patients' recuperation period and increasing medical expenses, have prompted the need for creating novel and safer drug delivery systems. Among many proposed concepts, polymeric nanocarriers are shown to be a promising approach, allowing for controlled and selective drug delivery, simultaneously enhancing its activity towards cancerous cells and reducing toxic effects on healthy tissues. This article is a chronological examination of the history of the work progress on polymeric nanostructures, designed as efficient doxorubicin nanocarriers, with the emphasis on the main achievements of 2010-2020. Numerous publications have been reviewed to provide an essential summation of the nanopolymer types and their essential properties, mechanisms towards efficient drug delivery, as well as active targeting stimuli-responsive strategies that are currently utilized in the doxorubicin transportation field.
Collapse
Affiliation(s)
- Kamila Butowska
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland; (K.B.); (A.W.); (A.B.)
| | - Anna Woziwodzka
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland; (K.B.); (A.W.); (A.B.)
| | - Agnieszka Borowik
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland; (K.B.); (A.W.); (A.B.)
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK 73104, USA
| | - Jacek Piosik
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland; (K.B.); (A.W.); (A.B.)
| |
Collapse
|
11
|
Brossard C, Vlach M, Vène E, Ribault C, Dorcet V, Noiret N, Loyer P, Lepareur N, Cammas-Marion S. Synthesis of Poly(Malic Acid) Derivatives End-Functionalized with Peptides and Preparation of Biocompatible Nanoparticles to Target Hepatoma Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:958. [PMID: 33918663 PMCID: PMC8070460 DOI: 10.3390/nano11040958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Recently, short synthetic peptides have gained interest as targeting agents in the design of site-specific nanomedicines. In this context, our work aimed at developing new tools for the diagnosis and/or therapy of hepatocellular carcinoma (HCC) by grafting the hepatotropic George Baker (GB) virus A (GBVA10-9) and Plasmodium circumsporozoite protein (CPB)-derived peptides to the biocompatible poly(benzyl malate), PMLABe. We successfully synthesized PMLABe derivatives end-functionalized with peptides GBVA10-9, CPB, and their corresponding scrambled peptides through a thiol/maleimide reaction. The corresponding nanoparticles (NPs), varying by the nature of the peptide (GBVA10-9, CPB, and their scrambled peptides) and the absence or presence of poly(ethylene glycol) were also successfully formulated using nanoprecipitation technique. NPs were further characterized by dynamic light scattering (DLS), electrophoretic light scattering (ELS) and transmission electron microscopy (TEM), highlighting a diameter lower than 150 nm, a negative surface charge, and a more or less spherical shape. Moreover, a fluorescent probe (DiD Oil) has been encapsulated during the nanoprecipitation process. Finally, preliminary in vitro internalisation assays using HepaRG hepatoma cells demonstrated that CPB peptide-functionalized PMLABe NPs were efficiently internalized by endocytosis, and that such nanoobjects may be promising drug delivery systems for the theranostics of HCC.
Collapse
Affiliation(s)
- Clarisse Brossard
- University Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR, UMR 6226, ScanMAT, UMS2001, F-35000 Rennes, France; (C.B.); (V.D.); (N.N.)
| | - Manuel Vlach
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, University Rennes, F-35000 Rennes, France; (M.V.); (E.V.); (C.R.)
| | - Elise Vène
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, University Rennes, F-35000 Rennes, France; (M.V.); (E.V.); (C.R.)
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU Rennes, F-35033 Rennes, France
| | - Catherine Ribault
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, University Rennes, F-35000 Rennes, France; (M.V.); (E.V.); (C.R.)
| | - Vincent Dorcet
- University Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR, UMR 6226, ScanMAT, UMS2001, F-35000 Rennes, France; (C.B.); (V.D.); (N.N.)
| | - Nicolas Noiret
- University Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR, UMR 6226, ScanMAT, UMS2001, F-35000 Rennes, France; (C.B.); (V.D.); (N.N.)
| | - Pascal Loyer
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, University Rennes, F-35000 Rennes, France; (M.V.); (E.V.); (C.R.)
| | - Nicolas Lepareur
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, University Rennes, F-35000 Rennes, France; (M.V.); (E.V.); (C.R.)
- Comprehensive Cancer Center Eugène Marquis, F-35000 Rennes, France
| | - Sandrine Cammas-Marion
- University Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR, UMR 6226, ScanMAT, UMS2001, F-35000 Rennes, France; (C.B.); (V.D.); (N.N.)
- INSERM, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, University Rennes, F-35000 Rennes, France; (M.V.); (E.V.); (C.R.)
| |
Collapse
|
12
|
Vitale I, Yamazaki T, Wennerberg E, Sveinbjørnsson B, Rekdal Ø, Demaria S, Galluzzi L. Targeting Cancer Heterogeneity with Immune Responses Driven by Oncolytic Peptides. Trends Cancer 2021; 7:557-572. [PMID: 33446447 DOI: 10.1016/j.trecan.2020.12.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
Accumulating preclinical and clinical evidence indicates that high degrees of heterogeneity among malignant cells constitute a considerable obstacle to the success of cancer therapy. This calls for the development of approaches that operate - or enable established treatments to operate - despite such intratumoral heterogeneity (ITH). In this context, oncolytic peptides stand out as promising therapeutic tools based on their ability to drive immunogenic cell death associated with robust anticancer immune responses independently of ITH. We review the main molecular and immunological pathways engaged by oncolytic peptides, and discuss potential approaches to combine these agents with modern immunotherapeutics in support of superior tumor-targeting immunity and efficacy in patients with cancer.
Collapse
Affiliation(s)
- Ilio Vitale
- Italian Institute for Genomic Medicine (IIGM), Istituto Di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO)-IRCCS, Candiolo, Italy
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Erik Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Baldur Sveinbjørnsson
- Lytix Biopharma, Oslo, Norway; Department of Medical Biology, University of Tromsø, Tromsø, Norway; Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway; Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| |
Collapse
|
13
|
Weyemi U, Galluzzi L. Chromatin and genomic instability in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 364:ix-xvii. [PMID: 34507786 DOI: 10.1016/s1937-6448(21)00116-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Urbain Weyemi
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| |
Collapse
|
14
|
Grigoletto A, Tedeschini T, Canato E, Pasut G. The evolution of polymer conjugation and drug targeting for the delivery of proteins and bioactive molecules. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1689. [PMID: 33314717 DOI: 10.1002/wnan.1689] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022]
Abstract
Polymer conjugation can be considered one of the leading approaches within the vast field of nanotechnology-based drug delivery systems. In fact, such technology can be exploited for delivering an active molecule, such as a small drug, a protein, or genetic material, or it can be applied to other drug delivery systems as a strategy to improve their in vivo behavior or pharmacokinetic activities such as prolonging the half-life of a drug, conferring stealth properties, providing external stimuli responsiveness, and so on. If on the one hand, polymer conjugation with biotech drug is considered the linchpin of the protein delivery field boasting several products in clinical use, on the other, despite dedicated research, conjugation with low molecular weight drugs has not yet achieved the milestone of the first clinical approval. Some of the primary reasons for this debacle are the difficulties connected to achieving selective targeting to diseased tissue, organs, or cells, which is the main goal not only of polymer conjugation but of all delivery systems of small drugs. In light of the need to achieve better drug targeting, researchers are striving to identify more sophisticated, biocompatible delivery approaches and to open new horizons for drug targeting methodologies leading to successful clinical applications. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Tommaso Tedeschini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Elena Canato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
15
|
Abstract
Exosomes are extracellular vesicles secreted by cells and involved in intercellular communications among close and distant cells. Exosomes encapsulate and carry biomolecules as cargo to the recipient cells. They contain nucleic acids (DNA, RNA, microRNA) proteins and lipids. Each exosomal components may be isolated and be studied by specific techniques. In this chapter, different methods will be described to isolate DNA from exosomes, since it is important in shaping the response of the recipient cells following the exosome uptake in multiple scenarios, including physiological and pathological conditions. Moreover, the exosomal DNA may be a novel biomarker for diagnosis, disease progression and patient's treatment response.
Collapse
Affiliation(s)
- Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
16
|
Three-dimensional cell models for extracellular vesicles production, isolation, and characterization. Methods Enzymol 2020; 645:209-230. [PMID: 33565973 DOI: 10.1016/bs.mie.2020.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) play a pivotal role in cancer progression. However, the majority of functional studies performed so far relies on data acquired in traditional 2D cultures. Because the spatial architecture of tissue is decisive for the cell fate, new cell models to study EV functions in the 3D environment must approximate in vitro models to the physiological conditions. Several models were developed during the last years, which may be suitable to serve as 3D models to study EVs; among them are hydrogels, solid scaffolds, bioreactors, and 3D CoSeedis™ inserts. We present in this chapter a protocol for a 3D cell model based on the 3D CoSeedis™ agarose inserts, allowing for a long-term culture of cells of different origins under serum-free conditions and easy EV recovery. Additionally, information on individual culture conditions in 3D CoSeedis™ for different cell lines, protocols for model evaluation, and quality controls are included. We hope that our suggestions and experience will be useful to carry out EV study under more physiological conditions and contribute to the EV research field's progress.
Collapse
|
17
|
Kopeček J, Yang J. Polymer nanomedicines. Adv Drug Deliv Rev 2020; 156:40-64. [PMID: 32735811 PMCID: PMC7736172 DOI: 10.1016/j.addr.2020.07.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
Polymer nanomedicines (macromolecular therapeutics, polymer-drug conjugates, drug-free macromolecular therapeutics) are a group of biologically active compounds that are characterized by their large molecular weight. This review focuses on bioconjugates of water-soluble macromolecules with low molecular weight drugs and selected proteins. After analyzing the design principles, different structures of polymer carriers are discussed followed by the examination of the efficacy of the conjugates in animal models and challenges for their translation into the clinic. Two innovative directions in macromolecular therapeutics that depend on receptor crosslinking are highlighted: a) Combination chemotherapy of backbone degradable polymer-drug conjugates with immune checkpoint blockade by multivalent polymer peptide antagonists; and b) Drug-free macromolecular therapeutics, a new paradigm in drug delivery.
Collapse
Affiliation(s)
- Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
18
|
Novoselova MV, Loh HM, Trushina DB, Ketkar A, Abakumova TO, Zatsepin TS, Kakran M, Brzozowska AM, Lau HH, Gorin DA, Antipina MN, Brichkina AI. Biodegradable Polymeric Multilayer Capsules for Therapy of Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:5610-5623. [PMID: 31942802 DOI: 10.1021/acsami.9b21381] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Formulated forms of cancer therapeutics enhance the efficacy of treatment by more precise targeting, increased bioavailability of drugs, and an aptitude of some delivery systems to overcome multiple drug resistance of tumors. Drug carriers acquire importance for anti-cancer interventions via targeting tumor-associated macrophages with active molecules capable to either eliminate them or change their polarity. Although several packaged drug forms have reached the market, there is still a high demand for novel carrier systems to hurdle limitations of existing drugs on active molecules, toxicity, bioeffect, and stability. Here, we report a facile assembly and delivery methodology for biodegradable polymeric multilayer capsules (PMC) with the purpose of further use in injectable drug formulations for lung cancer therapy via direct erosion of tumors and suppression of the tumor-promoting function of macrophages in the tumor microenvironment. We demonstrate delivery of low-molecular-weight drug molecules to lung cancer cells and macrophages and provide details on in vivo distribution, cellular uptake, and disintegration of the developed PMC. Poly-l-arginine and dextran sulfate alternately adsorb on a ∼500 nm CaCO3 sacrificial template followed by removal of the inorganic core to obtain hollow capsules for consequent loading with drug molecules, gemcitabine or clodronate. The capsules further compacted upon loading down to ∼250 nm in diameter via heat treatment. A comparative study of the capsule internalization rate in vitro and in vivo reveals the benefits of a diminished carrier size. We show that macrophages and epithelial cells of the lungs and liver internalize capsules with efficacy higher than 75%. Using an in vivo mouse model of lung cancer, we also confirm that tumor lungs better retain smaller capsules than the healthy lung tissue. The pronounced cytotoxic effect of the encapsulated gemcitabine on lung cancer cells and the ability of the encapsulated clodronate to block the tumor-promoting function of macrophages prove the efficacy of the developed capsule loading method in vitro. Our study taken as a whole demonstrates the great potential of the developed PMC for in vivo treatment of cancer via transporting active molecules, including those that are water-soluble with low molecular weight, to both cancer cells and macrophages through the bloodstream.
Collapse
Affiliation(s)
- Marina V Novoselova
- Institute of Materials Research and Engineering, A*STAR , 2 Fusionopolis Way , Innovis, #08-03, Singapore , 138634 , Singapore
- Skolkovo Institute of Science and Technology , Bolshoy Boulevard 30, bld. 1 , Moscow 121205 , Russia
| | - Hui Mun Loh
- Institute of Molecular and Cell Biology, A*STAR , 61 Biopolis Drive , Proteos, Singapore 138673 , Singapore
| | - Daria B Trushina
- Institute of Materials Research and Engineering, A*STAR , 2 Fusionopolis Way , Innovis, #08-03, Singapore , 138634 , Singapore
- I.M. Sechenov First Moscow State Medical University , Bol'shaya Pirogovskaya Ulitsa 19c1 Moscow 119146 , Russia
- A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences , Leninskiy Prospekt, 59 , Moscow 119333 , Russia
| | - Avanee Ketkar
- Institute of Molecular Oncology , Philipps University of Marburg , member of the German Center for Lung Research (DZL), Hans-Meerwein-Str. 3 35043 Marburg , Germany
| | - Tatiana O Abakumova
- Skolkovo Institute of Science and Technology , Bolshoy Boulevard 30, bld. 1 , Moscow 121205 , Russia
| | - Timofei S Zatsepin
- Skolkovo Institute of Science and Technology , Bolshoy Boulevard 30, bld. 1 , Moscow 121205 , Russia
| | - Mitali Kakran
- Institute of Materials Research and Engineering, A*STAR , 2 Fusionopolis Way , Innovis, #08-03, Singapore , 138634 , Singapore
| | - Agata Maria Brzozowska
- Institute of Materials Research and Engineering, A*STAR , 2 Fusionopolis Way , Innovis, #08-03, Singapore , 138634 , Singapore
| | - Hooi Hong Lau
- Institute of Materials Research and Engineering, A*STAR , 2 Fusionopolis Way , Innovis, #08-03, Singapore , 138634 , Singapore
| | - Dmitry A Gorin
- Skolkovo Institute of Science and Technology , Bolshoy Boulevard 30, bld. 1 , Moscow 121205 , Russia
| | - Maria N Antipina
- Institute of Materials Research and Engineering, A*STAR , 2 Fusionopolis Way , Innovis, #08-03, Singapore , 138634 , Singapore
| | - Anna I Brichkina
- Institute of Molecular and Cell Biology, A*STAR , 61 Biopolis Drive , Proteos, Singapore 138673 , Singapore
- Institute of Molecular Oncology , Philipps University of Marburg , member of the German Center for Lung Research (DZL), Hans-Meerwein-Str. 3 35043 Marburg , Germany
| |
Collapse
|
19
|
Melnyk T, Đorđević S, Conejos-Sánchez I, Vicent MJ. Therapeutic potential of polypeptide-based conjugates: Rational design and analytical tools that can boost clinical translation. Adv Drug Deliv Rev 2020; 160:136-169. [PMID: 33091502 DOI: 10.1016/j.addr.2020.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022]
Abstract
The clinical success of polypeptides as polymeric drugs, covered by the umbrella term "polymer therapeutics," combined with related scientific and technological breakthroughs, explain their exponential growth in the development of polypeptide-drug conjugates as therapeutic agents. A deeper understanding of the biology at relevant pathological sites and the critical biological barriers faced, combined with advances regarding controlled polymerization techniques, material bioresponsiveness, analytical methods, and scale up-manufacture processes, have fostered the development of these nature-mimicking entities. Now, engineered polypeptides have the potential to combat current challenges in the advanced drug delivery field. In this review, we will discuss examples of polypeptide-drug conjugates as single or combination therapies in both preclinical and clinical studies as therapeutics and molecular imaging tools. Importantly, we will critically discuss relevant examples to highlight those parameters relevant to their rational design, such as linking chemistry, the analytical strategies employed, and their physicochemical and biological characterization, that will foster their rapid clinical translation.
Collapse
Affiliation(s)
- Tetiana Melnyk
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Snežana Đorđević
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
20
|
|
21
|
Pasut G. Grand Challenges in Nano-Based Drug Delivery. FRONTIERS IN MEDICAL TECHNOLOGY 2019; 1:1. [PMID: 35047870 PMCID: PMC8757891 DOI: 10.3389/fmedt.2019.00001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|