1
|
Korzun T, Moses AS, Diba P, Sattler AL, Olson B, Taratula OR, Pejovic T, Marks DL, Taratula O. Development and Perspectives: Multifunctional Nucleic Acid Nanomedicines for Treatment of Gynecological Cancers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301776. [PMID: 37518857 PMCID: PMC10827528 DOI: 10.1002/smll.202301776] [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: 02/28/2023] [Revised: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Gynecological malignancies are a significant cause of morbidity and mortality across the globe. Due to delayed presentation, gynecological cancer patients are often referred late in the disease's course, resulting in poor outcomes. A considerable number of patients ultimately succumb to chemotherapy-resistant disease, which reoccurs at advanced stages despite treatment interventions. Although efforts have been devoted to developing therapies that demonstrate reduced resistance to chemotherapy and enhanced toxicity profiles, current clinical outcomes remain unsatisfactory due to treatment resistance and unfavorable off-target effects. Consequently, innovative biological and nanotherapeutic approaches are imperative to strengthen and optimize the therapeutic arsenal for gynecological cancers. Advancements in nanotechnology-based therapies for gynecological malignancies offer significant advantages, including reduced toxicity, expanded drug circulation, and optimized therapeutic dosing, ultimately leading to enhanced treatment effectiveness. Recent advances in nucleic acid therapeutics using microRNA, small interfering RNA, and messenger RNA provide novel approaches for cancer therapeutics. Effective single-agent and combinatorial nucleic acid therapeutics for gynecological malignancies have the potential to transform cancer treatment by giving safer, more tailored approaches than conventional therapies. This review highlights current preclinical studies that effectively exploit these approaches for the treatment of gynecological malignant tumors and malignant ascites.
Collapse
Affiliation(s)
- Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue Portland, Portland, OR, 97239, USA
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
| | - Ariana L Sattler
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, Oregon, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Brennan Olson
- Mayo Clinic Department of Otolaryngology-Head and Neck Surgery, 200 First St. SW, Rochester, MN, 55905, USA
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Tanja Pejovic
- Departments of Obstetrics and Gynecology and Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, Oregon, 97201, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR, 97201, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, OR, 97201, USA
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Avenue Portland, Portland, OR, 97239, USA
| |
Collapse
|
2
|
Braatz D, Cherri M, Tully M, Dimde M, Ma G, Mohammadifar E, Reisbeck F, Ahmadi V, Schirner M, Haag R. Chemical Approaches to Synthetic Drug Delivery Systems for Systemic Applications. Angew Chem Int Ed Engl 2022; 61:e202203942. [PMID: 35575255 PMCID: PMC10091760 DOI: 10.1002/anie.202203942] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Indexed: 11/10/2022]
Abstract
Poor water solubility and low bioavailability of active pharmaceutical ingredients (APIs) are major causes of friction in the pharmaceutical industry and represent a formidable hurdle for pharmaceutical drug development. Drug delivery remains the major challenge for the application of new small-molecule drugs as well as biopharmaceuticals. The three challenges for synthetic delivery systems are: (i) controlling drug distribution and clearance in the blood; (ii) solubilizing poorly water-soluble agents, and (iii) selectively targeting specific tissues. Although several polymer-based systems have addressed the first two demands and have been translated into clinical practice, no targeted synthetic drug delivery system has reached the market. This Review is designed to provide a background on the challenges and requirements for the design and translation of new polymer-based delivery systems. This report will focus on chemical approaches to drug delivery for systemic applications.
Collapse
Affiliation(s)
- Daniel Braatz
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Mariam Cherri
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Michael Tully
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Mathias Dimde
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Guoxin Ma
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Ehsan Mohammadifar
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Felix Reisbeck
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Vahid Ahmadi
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Michael Schirner
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| |
Collapse
|
3
|
Bhadra K. A Mini Review on Molecules Inducing Caspase-Independent Cell Death: A New Route to Cancer Therapy. Molecules 2022; 27:molecules27196401. [PMID: 36234938 PMCID: PMC9572491 DOI: 10.3390/molecules27196401] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Most anticancer treatments trigger tumor cell death through apoptosis, where initiation of proteolytic action of caspase protein is a basic need. But under certain circumstances, apoptosis is prevented by the apoptosis inhibitor proteins, survivin and Hsp70. Several drugs focusing on classical programmed death of the cell have been reported to have low anti-tumorogenic potency due to mutations in proteins involved in the caspase-dependent programmed cell death with intrinsic and extrinsic pathways. This review concentrates on the role of anti-cancer drug molecules targeting alternative pathways of cancer cell death for treatment, by providing a molecular basis for the new strategies of novel anti-cancer treatment. Under these conditions, active agents targeting alternative cell death pathways can be considered as potent chemotherapeutic drugs. Many natural compounds and other small molecules, such as inorganic and synthetic compounds, including several repurposing drugs, are reported to cause caspase-independent cell death in the system. However, few molecules indicated both caspase-dependent as well caspase-free cell death in specific cancer lines. Cancer cells have alternative methods of caspase-independent programmed cell death which are equally promising for being targeted by small molecules. These small molecules may be useful leads for rational therapeutic drug design, and can be of potential interest for future cancer-preventive strategies.
Collapse
Affiliation(s)
- Kakali Bhadra
- Department of Zoology, University of Kalyani, Nadia, Kalyani 741235, India
| |
Collapse
|
4
|
Lepland A, Malfanti A, Haljasorg U, Asciutto EK, Pickholz M, Bringas M, Đorđević S, Salumäe L, Peterson P, Teesalu T, Vicent MJ, Scodeller P. Depletion of Mannose Receptor-Positive Tumor-associated Macrophages via a Peptide-targeted Star-shaped Polyglutamate Inhibits Breast Cancer Progression in Mice. CANCER RESEARCH COMMUNICATIONS 2022; 2:533-551. [PMID: 36923553 PMCID: PMC10010335 DOI: 10.1158/2767-9764.crc-22-0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/03/2022] [Accepted: 06/03/2022] [Indexed: 12/31/2022]
Abstract
Although many studies have explored the depletion of tumor-associated macrophages (TAM) as a therapeutic strategy for solid tumors, currently available compounds suffer from poor efficacy and dose-limiting side effects. Here, we developed a novel TAM-depleting agent ("OximUNO") that specifically targets CD206+ TAMs and demonstrated efficacy in a triple-negative breast cancer (TNBC) mouse model. OximUNO comprises a star-shaped polyglutamate (St-PGA) decorated with the CD206-targeting peptide mUNO that carries the chemotherapeutic drug doxorubicin (DOX). In the TNBC model, a fluorescently labeled mUNO-decorated St-PGA homed to CD206+ TAMs within primary lesions and metastases. OximUNO exhibited no acute liver or kidney toxicity in vivo. Treatment with OximUNO reduced the progression of primary tumor lesions and pulmonary metastases, significantly diminished the number of CD206+ TAMs and increased the CD8/FOXP3 expression ratio (indicating immunomodulation). Our findings suggest the potential benefit of OximUNO as a TAM-depleting agent for TNBC treatment. Importantly, our studies also represent a novel design of a peptide-targeted St-PGA as a targeted therapeutic nanoconjugate. Significance A peptide-targeted nanoformulation of DOX exclusively eliminates mannose receptor+ TAMs in breast cancer models, generating response without off-target effects (a drawback of many TAM-depleting agents under clinical study).
Collapse
Affiliation(s)
- Anni Lepland
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Alessio Malfanti
- Polymer Therapeutics Laboratory, Prince Felipe Research Centre, Valencia, Spain
| | - Uku Haljasorg
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Eliana K. Asciutto
- School of Science and Technology, National University of San Martin (UNSAM) ICIFI and CONICET, Buenos Aires, Argentina
| | - Monica Pickholz
- Departamento de Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Física de Buenos Aires (IFIBA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mauro Bringas
- Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA-CONICET), C1405BWE Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Snežana Đorđević
- Polymer Therapeutics Laboratory, Prince Felipe Research Centre, Valencia, Spain
| | - Liis Salumäe
- Pathology Department, Tartu University Hospital, Tartu, Estonia
| | - Pärt Peterson
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Centre for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, California
| | - María J. Vicent
- Polymer Therapeutics Laboratory, Prince Felipe Research Centre, Valencia, Spain
| | - Pablo Scodeller
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
5
|
Đorđević S, Gonzalez MM, Conejos-Sánchez I, Carreira B, Pozzi S, Acúrcio RC, Satchi-Fainaro R, Florindo HF, Vicent MJ. Current hurdles to the translation of nanomedicines from bench to the clinic. Drug Deliv Transl Res 2022; 12:500-525. [PMID: 34302274 PMCID: PMC8300981 DOI: 10.1007/s13346-021-01024-2] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
The field of nanomedicine has significantly influenced research areas such as drug delivery, diagnostics, theranostics, and regenerative medicine; however, the further development of this field will face significant challenges at the regulatory level if related guidance remains unclear and unconsolidated. This review describes those features and pathways crucial to the clinical translation of nanomedicine and highlights considerations for early-stage product development. These include identifying those critical quality attributes of the drug product essential for activity and safety, appropriate analytical methods (physical, chemical, biological) for characterization, important process parameters, and adequate pre-clinical models. Additional concerns include the evaluation of batch-to-batch consistency and considerations regarding scaling up that will ensure a successful reproducible manufacturing process. Furthermore, we advise close collaboration with regulatory agencies from the early stages of development to assure an aligned position to accelerate the development of future nanomedicines.
Collapse
Affiliation(s)
- Snežana Đorđević
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - María Medel Gonzalez
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain
| | - Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel.
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal.
| | - María J Vicent
- Polymer Therapeutics Laboratory, Prince Felipe Research Center (CIPF), Eduardo Primo Yúfera 3, 46012, Valencia, Av, Spain.
| |
Collapse
|
6
|
Tejedor S, Dolz‐Pérez I, Decker CG, Hernándiz A, Diez JL, Álvarez R, Castellano D, García NA, Ontoria‐Oviedo I, Nebot VJ, González‐King H, Igual B, Sepúlveda P, Vicent MJ. Polymer Conjugation of Docosahexaenoic Acid Potentiates Cardioprotective Therapy in Preclinical Models of Myocardial Ischemia/Reperfusion Injury. Adv Healthc Mater 2021; 10:e2002121. [PMID: 33720548 DOI: 10.1002/adhm.202002121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/16/2021] [Indexed: 01/16/2023]
Abstract
While coronary angioplasty represents an effective treatment option following acute myocardial infarction, the reperfusion of the occluded coronary artery can prompt ischemia-reperfusion (I/R) injury that significantly impacts patient outcomes. As ω-3 polyunsaturated fatty acids (PUFAs) have proven, yet limited cardioprotective abilities, an optimized polymer-conjugation approach is reported that improves PUFAs bioavailability to enhance cardioprotection and recovery in animal models of I/R-induced injury. Poly-l-glutamic acid (PGA) conjugation improves the solubility and stability of di-docosahexaenoic acid (diDHA) under physiological conditions and protects rat neonatal ventricular myocytes from I/R injury by reducing apoptosis, attenuating autophagy, inhibiting reactive oxygen species generation, and restoring mitochondrial membrane potential. Enhanced protective abilities are associated with optimized diDHA loading and evidence is provided for the inherent cardioprotective potential of PGA itself. Pretreatment with PGA-diDHA before reperfusion in a small animal I/R model provides for cardioprotection and limits area at risk (AAR). Furthermore, the preliminary findings suggest that PGA-diDHA administration in a swine I/R model may provide cardioprotection, limit edema and decrease AAR. Overall, the evaluation of PGA-diDHA in relevant preclinical models provides evidence for the potential of polymer-conjugated PUFAs in the mitigation of I/R injury associated with coronary angioplasty.
Collapse
Affiliation(s)
- Sandra Tejedor
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Irene Dolz‐Pérez
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Caitlin G. Decker
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| | - Amparo Hernándiz
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Jose L. Diez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Raquel Álvarez
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Delia Castellano
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Nahuel A. García
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Imelda Ontoria‐Oviedo
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Vicent J. Nebot
- Polypeptide Therapeutic Solutions S.L. Av. Benjamin Franklin 19, Paterna Valencia 46980 Spain
| | - Hernán González‐King
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Begoña Igual
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit Instituto de Investigación Sanitaria La Fe Avda. Fernando Abril Martorell 106 Valencia 46026 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Av. Eduardo Primo Yúfera 3 Valencia E‐46012 Spain
| |
Collapse
|
7
|
Duro-Castano A, Borrás C, Herranz-Pérez V, Blanco-Gandía MC, Conejos-Sánchez I, Armiñán A, Mas-Bargues C, Inglés M, Miñarro J, Rodríguez-Arias M, García-Verdugo JM, Viña J, Vicent MJ. Targeting Alzheimer's disease with multimodal polypeptide-based nanoconjugates. SCIENCE ADVANCES 2021; 7:7/13/eabf9180. [PMID: 33771874 PMCID: PMC7997513 DOI: 10.1126/sciadv.abf9180] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/08/2021] [Indexed: 05/04/2023]
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, remains incurable mainly due to our failings in the search for effective pharmacological strategies. Here, we describe the development of targeted multimodal polypeptide-based nanoconjugates as potential AD treatments. Treatment with polypeptide nanoconjugates bearing propargylamine moieties and bisdemethoxycurcumin or genistein afforded neuroprotection and displayed neurotrophic effects, as evidenced by an increase in dendritic density of pyramidal neurons in organotypic hippocampal culture. The additional conjugation of the Angiopep-2 targeting moiety enhanced nanoconjugate passage through the blood-brain barrier and modulated brain distribution with nanoconjugate accumulation in neurogenic areas, including the olfactory bulb. Nanoconjugate treatment effectively reduced neurotoxic β amyloid aggregate levels and rescued impairments to olfactory memory and object recognition in APP/PS1 transgenic AD model mice. Overall, this study provides a description of a targeted multimodal polyglutamate-based nanoconjugate with neuroprotective and neurotrophic potential for AD treatment.
Collapse
Affiliation(s)
- A Duro-Castano
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - C Borrás
- Grupo de Investigación FRESHAGE, Departamento de Fisiología, Facultad de Medicina, Univ.. Valencia, CIBERFES-ISCIII, INCLIVA, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - V Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, Univ. València, CIBERNED, 46980 Valencia, Spain
- Predepartamental Unit of Medicine, Faculty of Health Sciences, Univ. Jaume I, 12071 Castelló de la Plana, Spain
| | - M C Blanco-Gandía
- Departamento de Psicología y Sociología, Facultad de Ciencias Sociales y Humanas, Univ. Zaragoza, Teruel, Spain
| | - I Conejos-Sánchez
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - A Armiñán
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain
| | - C Mas-Bargues
- Grupo de Investigación FRESHAGE, Departamento de Fisiología, Facultad de Medicina, Univ.. Valencia, CIBERFES-ISCIII, INCLIVA, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - M Inglés
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Univ. Valencia, Valencia, Spain
| | - J Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Univ. Valencia, Valencia, Spain
| | - M Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Univ. Valencia, Valencia, Spain
| | - J M García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, Univ. València, CIBERNED, 46980 Valencia, Spain
| | - J Viña
- Grupo de Investigación FRESHAGE, Departamento de Fisiología, Facultad de Medicina, Univ.. Valencia, CIBERFES-ISCIII, INCLIVA, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - M J Vicent
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe (CIPF), Av. Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| |
Collapse
|
8
|
Polyglutamic acid-based crosslinked doxorubicin nanogels as an anti-metastatic treatment for triple negative breast cancer. J Control Release 2021; 332:10-20. [PMID: 33587988 DOI: 10.1016/j.jconrel.2021.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Treatment of triple negative breast cancer (TNBC)-associated metastasis represents an unmet clinical need, and we lack effective therapeutics for a disease that exhibits high relapse rates and associates with poor patient outcomes. Advanced nanosized drug delivery systems may enhance the efficacy of first-line chemotherapeutics by altering drug pharmacokinetics and enhancing tumor/metastasis targeting to significantly improve efficacy and safety. Herein, we propose the application of injectable poly-amino acid-based nanogels (NGs) as a versatile hydrophilic drug delivery platform for the treatment of TNBC lung metastasis. We prepared biocompatible and biodegradable cross-linked NGs from polyglutamic acid (PGA) loaded with the chemotherapeutic agent doxorubicin (DOX). Our optimized synthetic procedures generated NGs of ~100 nm in size and 25 wt% drug loading content that became rapidly internalized in TNBC cell lines and displayed IC50 values comparable to the free form of DOX. Importantly, PGA-DOX NGs significantly inhibited lung metastases and almost completely suppressed lymph node metastases in a spontaneously metastatic orthotopic mouse TNBC model. Overall, our newly developed PGA-DOX NGs represent a potentially effective therapeutic strategy for the treatment of TNBC metastases.
Collapse
|
9
|
Piscatelli JA, Ban J, Lucas AT, Zamboni WC. Complex Factors and Challenges that Affect the Pharmacology, Safety and Efficacy of Nanocarrier Drug Delivery Systems. Pharmaceutics 2021; 13:114. [PMID: 33477395 PMCID: PMC7830329 DOI: 10.3390/pharmaceutics13010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/01/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Major developments in nanomedicines, such as nanoparticles (NPs), nanosomes, and conjugates, have revolutionized drug delivery capabilities over the past four decades. Although nanocarrier agents provide numerous advantages (e.g., greater solubility and duration of systemic exposure) compared to their small-molecule counterparts, there is considerable inter-patient variability seen in the systemic disposition, tumor delivery and overall pharmacological effects (i.e., anti-tumor efficacy and unwanted toxicity) of NP agents. This review aims to provide a summary of fundamental factors that affect the disposition of NPs in the treatment of cancer and why they should be evaluated during preclinical and clinical development. Furthermore, this chapter will highlight some of the translational challenges associated with elements of NPs and how these issues can only be addressed by detailed and novel pharmacology studies.
Collapse
Affiliation(s)
- Joseph A. Piscatelli
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Jisun Ban
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Andrew T. Lucas
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Liu G, Lovell JF, Zhang L, Zhang Y. Stimulus-Responsive Nanomedicines for Disease Diagnosis and Treatment. Int J Mol Sci 2020; 21:E6380. [PMID: 32887466 PMCID: PMC7504550 DOI: 10.3390/ijms21176380] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Stimulus-responsive drug delivery systems generally aim to release the active pharmaceutical ingredient (API) in response to specific conditions and have recently been explored for disease treatments. These approaches can also be extended to molecular imaging to report on disease diagnosis and management. The stimuli used for activation are based on differences between the environment of the diseased or targeted sites, and normal tissues. Endogenous stimuli include pH, redox reactions, enzymatic activity, temperature and others. Exogenous site-specific stimuli include the use of magnetic fields, light, ultrasound and others. These endogenous or exogenous stimuli lead to structural changes or cleavage of the cargo carrier, leading to release of the API. A wide variety of stimulus-responsive systems have been developed-responsive to both a single stimulus or multiple stimuli-and represent a theranostic tool for disease treatment. In this review, stimuli commonly used in the development of theranostic nanoplatforms are enumerated. An emphasis on chemical structure and property relationships is provided, aiming to focus on insights for the design of stimulus-responsive delivery systems. Several examples of theranostic applications of these stimulus-responsive nanomedicines are discussed.
Collapse
Affiliation(s)
- Gengqi Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, NY 14260, USA;
| | - Lei Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| |
Collapse
|
11
|
Fathi M, Abdolahinia ED, Barar J, Omidi Y. Smart stimuli-responsive biopolymeric nanomedicines for targeted therapy of solid tumors. Nanomedicine (Lond) 2020; 15:2171-2200. [DOI: 10.2217/nnm-2020-0146] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Solid tumors form a permissive microenvironment with irregular features, including high pressured tumor interstitial fluid with acidic pH, co-adaptation of cancer cells with other cells like the immune system cells, abnormal metabolism and anomalous overexpression of various pieces of molecular machinery. The functional expressions of several oncomarkers in different solid tumors have led to the development of targeted drug-delivery systems (DDSs). As a new class of DDSs, stimuli-responsive nanomedicines (SRNMs) have been developed using advanced nanobiomaterials such as biopolymers that show excellent biocompatibility with low inherent immunogenicity. In this review, we aim to overview different types of SRNMs, present deep insights into the stimuli-responsive biopolymers and discuss the most up-to-date progress in the design and development of SRNMs used as advanced DDSs for targeted therapy of cancer.
Collapse
Affiliation(s)
- Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
12
|
Pisarevsky E, Blau R, Epshtein Y, Ben-Shushan D, Eldar-Boock A, Tiram G, Koshrovski-Michael S, Scomparin A, Pozzi S, Krivitsky A, Shenbach-Koltin G, Yeini E, Fridrich L, White R, Satchi-Fainaro R. Rational Design of Polyglutamic Acid Delivering an Optimized Combination of Drugs Targeting Mutated BRAF and MEK in Melanoma. ADVANCED THERAPEUTICS 2020; 3:2000028. [PMID: 35754977 PMCID: PMC9223483 DOI: 10.1002/adtp.202000028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Indexed: 12/17/2022]
Abstract
Targeted therapies against cancer can relieve symptoms and induce remission, however, they often present limited duration of disease control, cause side effects and often induce acquired resistance. Therefore, there is a great motivation to develop a unique delivery system, targeted to the tumor, in which we can combine several active entities, increase the therapeutic index by reducing systemic exposure, and enhance their synergistic activity. To meet these goals, we chose the biocompatible and biodegradable poly(α,L-glutamic acid) (PGA) as a nanocarrier that facilitates extravasation-dependent tumor targeting delivery. The RAS/RAF/MEK/ERK pathway when aberrantly activated in melanoma, can lead to uncontrolled cell proliferation, induced invasion, and reduced apoptosis. Here, we selected two drugs targeting this pathway; a MEK1/2 inhibitor (selumetinib; SLM) and a modified BRAF inhibitor (modified dabrafenib; mDBF), that exhibited synergism in vitro. We synthesized and characterized our nanomedicine of PGA conjugated to SLM and mDBF (PGA-SLM-mDBF). PGA-SLM-mDBF inhibited the proliferation of melanoma cells and decreased their migratory and sprouting abilities without inducing a hemolytic effect. Moreover, the polymer-2-drugs conjugate exhibited superior anti-tumor activity in comparison with the two separate polymer-drug conjugates in vitro and with free drugs in a mouse model of primary melanoma and prolonged survival at a lower dose.
Collapse
Affiliation(s)
- Evgeni Pisarevsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rachel Blau
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yana Epshtein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gal Shenbach-Koltin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lidar Fridrich
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Richard White
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
13
|
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
|
14
|
McDowell SH, Gallaher SA, Burden RE, Scott CJ. Leading the invasion: The role of Cathepsin S in the tumour microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118781. [PMID: 32544418 DOI: 10.1016/j.bbamcr.2020.118781] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
Elevated expression of the cysteine protease Cathepsin S has been correlated with a number of different cancer types in recent years. As tools have been developed to enable more accurate examination of individual cathepsin species, our knowledge and appreciation of the role that this protease plays in facilitating cancer has increased exponentially. This review focuses on our current understanding of the role of Cathepsin S within tumours and the surrounding microenvironment. While various publications have shown that Cathepsin S can be derived from tumour cells themselves, a plethora of more recent studies have identified that Cathepsin S can also be derived from other cell types within the tumour microenvironment including endothelial cells, macrophages and T cells. Furthermore, specific proteolytic substrates cleaved by Cathepsin S have also been identified which have reinforced our hypothesis that this protease facilitates key steps within tumours leading to their invasion, angiogenesis and metastasis.
Collapse
Affiliation(s)
- Sara H McDowell
- The Patrick G Johnston Centre for Cancer Research, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK.
| | - Samantha A Gallaher
- The Patrick G Johnston Centre for Cancer Research, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK.
| | - Roberta E Burden
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | - Christopher J Scott
- The Patrick G Johnston Centre for Cancer Research, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK.
| |
Collapse
|
15
|
Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J Control Release 2020; 322:566-592. [DOI: 10.1016/j.jconrel.2020.03.051] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022]
|
16
|
Maso K, Grigoletto A, Raccagni L, Bellini M, Marigo I, Ingangi V, Suzuki A, Hirai M, Kamiya M, Yoshioka H, Pasut G. Poly(L-glutamic acid)-co-poly(ethylene glycol) block copolymers for protein conjugation. J Control Release 2020; 324:228-237. [PMID: 32413454 DOI: 10.1016/j.jconrel.2020.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 01/04/2023]
Abstract
Poly(L-glutamic acid)-co-poly(ethylene glycol) block copolymers (PLE-PEG) are here investigated as polymers for conjugation to therapeutic proteins such as granulocyte colony stimulating factor (G-CSF) and human growth hormone (hGH). PLE-PEG block copolymers are able to stabilize and protect proteins from degradation and to prolong their residence time in the blood stream, features that are made possible thanks to PEG's intrinsic properties and the simultaneous presence of the biodegradable anionic PLE moiety. When PLE-PEG copolymers are selectively tethered to the N-terminus of G-CSF and hGH, they yield homogeneous monoconjugates that preserve the protein's secondary structure. During the current study the pharmacokinetics of PLE10-PEG20k-G-CSF and PLE20-PEG20k-G-CSF derivatives and their ability to induce granulopoiesis were, respectively, assessed in Sprague-Dawley rats and in C57BL6 mice. Our results show that the bioavailability and bioactivity of the derivatives are comparable to or better than those of PEG20k-Nter-G-CSF (commercially known as Pegfilgrastim). The therapeutic effects of PLE10-PEG20k-hGH and PLE20-PEG20k-hGH derivatives tested in hypophysectomized rats demonstrate that the presence of a negatively charged PLE block enhances the biological properties of the conjugates additionally with respect to PEG20k-Nter-hGH.
Collapse
Affiliation(s)
- Katia Maso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, via F. Marzolo 5, 35131 Padua, Italy
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, via F. Marzolo 5, 35131 Padua, Italy
| | - Lucia Raccagni
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, via F. Marzolo 5, 35131 Padua, Italy
| | - Marino Bellini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, via F. Marzolo 5, 35131 Padua, Italy
| | - Ilaria Marigo
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | | | - Akira Suzuki
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Midori Hirai
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Masaki Kamiya
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Hiroki Yoshioka
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, via F. Marzolo 5, 35131 Padua, Italy.
| |
Collapse
|
17
|
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
|
18
|
Tumor heterogeneity and nanoparticle-mediated tumor targeting: the importance of delivery system personalization. Drug Deliv Transl Res 2018; 8:1508-1526. [PMID: 30128797 DOI: 10.1007/s13346-018-0578-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
After the discovery of the enhanced permeability and retention effect in 1986, it was envisioned that nanoparticle-mediated tumor-targeted delivery of chemotherapeutics would make a radical change in cancer therapy. However, after three decades of extensive research, only a few nanotherapeutics have been approved for clinical use. Although significant advantages of nanomedicines have been demonstrated in pre-clinical studies, clinical outcome was found to be variable. Advanced research has revealed that significant biochemical and structural variations exist between (and among) different tumors. These variations can considerably affect the tumor delivery and efficacy of nanomedicines. Tumor penetration is an important determining factor for positive therapeutic outcome and same nanomedicine can show diverse efficacy against different tumors depending on the extent of tumor accumulation and penetration. Recent research has started shading light on how the tumor variations can influence nanoparticle tumor delivery. These findings indicate that there is no "ideal" design of nanoparticles for exhibiting equally high efficacy against a broad spectrum of tumors. For achieving maximum benefit of the nanotherapeutics, it is necessary to analyze the tumor microenvironment for understanding the biological and structural characteristics of the tumor. Designing of the nanomedicine should be done according to the tumor characteristics. In this comprehensive review, we have first given a brief overview of the design characteristics of nanomedicine which impact their tumor delivery. Then we discussed about the variability in the tumor architecture and how it influences nanomedicine delivery. Finally, we have discussed the possibility of delivery system personalization based on the tumor characteristics.
Collapse
|
19
|
El-Sawy HS, Al-Abd AM, Ahmed TA, El-Say KM, Torchilin VP. Stimuli-Responsive Nano-Architecture Drug-Delivery Systems to Solid Tumor Micromilieu: Past, Present, and Future Perspectives. ACS NANO 2018; 12:10636-10664. [PMID: 30335963 DOI: 10.1021/acsnano.8b06104] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The microenvironment characteristics of solid tumors, renowned as barriers that harshly impeded many drug-delivery approaches, were precisely studied, investigated, categorized, divided, and subdivided into a complex diverse of barriers. These categories were further studied with a particular perspective, which makes all barriers found in solid-tumor micromilieu turn into different types of stimuli, and were considered triggers that can increase and hasten drug-release targeting efficacy. This review gathers data concerning the nature of solid-tumor micromilieu. Past research focused on the treatment of such tumors, the recent efforts employed for engineering smart nanoarchitectures with the utilization of the specified stimuli categories, the possibility of combining more than one stimuli for much-greater targeting enhancement, examples of the approved nanoarchitectures that already translated clinically as well as the obstacles faced by the use of these nanostructures, and, finally, an overview of the possible future implementations of smart-chemical engineering for the design of more-efficient drug delivery and theranostic systems and for making nanosystems with a much-higher level of specificity and penetrability features.
Collapse
Affiliation(s)
- Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy , Egyptian Russian University , Badr City , Cairo 63514 , Egypt
| | - Ahmed M Al-Abd
- Department of Pharmaceutical Sciences, College of Pharmacy , Gulf Medical University , Ajman , United Arab Emirates
- Pharmacology Department, Medical Division , National Research Centre , Giza 12622 , Egypt
| | - Tarek A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Khalid M El-Say
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , 140 The Fenway, Room 211/214, 360 Huntington Aveue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
20
|
Rao NV, Ko H, Lee J, Park JH. Recent Progress and Advances in Stimuli-Responsive Polymers for Cancer Therapy. Front Bioeng Biotechnol 2018; 6:110. [PMID: 30159310 PMCID: PMC6104418 DOI: 10.3389/fbioe.2018.00110] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
The conventional chemotherapeutic agents, used for cancer chemotherapy, have major limitations including non-specificity, ubiquitous biodistribution, low concentration in tumor tissue, and systemic toxicity. In recent years, owing to their unique features, polymeric nanoparticles have been widely used for the target-specific delivery of drugs in the body. Although polymeric nanoparticles have addressed a number of important issues, the bioavailability of drugs at the disease site, and especially upon cellular internalization, remains a challenge. A polymer nanocarrier system with a stimuli-responsive property (e.g., pH, temperature, or redox potential), for example, would be amenable to address the intracellular delivery barriers by taking advantage of pH, temperature, or redox potentials. With a greater understanding of the difference between normal and pathological tissues, there is a highly promising role of stimuli-responsive nanocarriers for drug delivery in the future. In this review, we highlighted the recent advances in different types of stimuli-responsive polymers for drug delivery.
Collapse
Affiliation(s)
- N. Vijayakameswara Rao
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Hyewon Ko
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, South Korea
| | - Jeongjin Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, South Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, South Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, South Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
21
|
Rautio J, Meanwell NA, Di L, Hageman MJ. The expanding role of prodrugs in contemporary drug design and development. Nat Rev Drug Discov 2018; 17:559-587. [DOI: 10.1038/nrd.2018.46] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
Morey M, Pandit A. Responsive triggering systems for delivery in chronic wound healing. Adv Drug Deliv Rev 2018; 129:169-193. [PMID: 29501700 DOI: 10.1016/j.addr.2018.02.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/27/2018] [Accepted: 02/26/2018] [Indexed: 12/31/2022]
Abstract
Non-communicable diseases including cancer, cardiovascular disease, diabetes, and neuropathy are chronic in nature. Treatment of these diseases with traditional delivery systems is limited due to lack of site-specificity, non-spatiotemporal release and insufficient doses. Numerous responsive delivery systems which respond to both physiological and external stimuli have been reported in the literature. However, effective strategies incorporating a multifactorial approach are required to control these complex wounds. This can be achieved by fabricating spatiotemporal release systems, multimodal systems or dual/multi-stimuli responsive delivery systems loaded with one or more bioactive components. Critically, these next generation stimuli responsive delivery systems that are at present not feasible are required to treat chronic wounds. This review provides a critical assessment of recent developments in the field of responsive delivery systems, highlighting their limitations and providing a perspective on how these challenges can be overcome.
Collapse
Affiliation(s)
- Mangesh Morey
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
23
|
Synthetic Poly(L-Glutamic Acid)-conjugated CpG Exhibits Antitumor Efficacy With Increased Retention in Tumor and Draining Lymph Nodes After Intratumoral Injection in a Mouse Model of Melanoma. J Immunother 2018; 40:11-20. [PMID: 27681378 DOI: 10.1097/cji.0000000000000145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There is an urgent need for new clinically applicable drug-delivery methods to enhance accumulation of immune-activating drugs in tumors. We synthesized a poly(L-glutamic acid)-CpG ODN2216 conjugate (PG-CpG) and injected it intratumorally into C57BL/6 mice bearing subcutaneous B16-ovalbumin melanoma. PG-CpG elicited the same potent antitumoral activity as CpG with respect to reducing tumor growth and triggering antigen-specific CD8 T-cell responses in this well-established solid tumor model. Moreover, PG-CpG was retained significantly longer in both tumor and draining lymph nodes than was free CpG after intratumoral injection. Specifically, 48 hours after injection, 26.5%±16.9% of the injected PG-CpG dose versus 4.72%±2.61% of free CpG remained at the tumor, and 1.53%±1.22% of the injected PG-CpG versus 0.37%±0.33% of free CpG was retained in the draining inguinal lymph nodes. These findings indicate that PG is an effective synthetic polymeric carrier for delivery of immunostimulatory agents to tumors and lymph nodes.
Collapse
|
24
|
Wang Z, Wu Z, Liu J, Zhang W. Particle morphology: an important factor affecting drug delivery by nanocarriers into solid tumors. Expert Opin Drug Deliv 2017; 15:379-395. [PMID: 29264946 DOI: 10.1080/17425247.2018.1420051] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Efficient delivery of drugs by nanoparticles deep into solid tumors is the precondition of valid cancer therapy. Despite profound understanding of the delivery of spherical nanoparticles into solid tumor attained, insufficient attention was paid to anisotropic particles. Actually, owing to their structural asymmetry, some non-spherical particles exhibit significant advantages over their spherical counterparts. AREAS COVERED This review will focus on particles with different shapes (discoidal particle, nanorod, filamentous particle, single-walled carbon nanotube) and the influence of their morphological characteristics (size, aspect ratio, rigidity) on the process of drug delivery to solid tumor in view of systemic circulation, transport from circulation system to tumor tissue, intratumoral transport and uptake by tumor cells, on the basis of introduction of challenges for drug delivery to solid tumor. In addition, the morphological characteristics will be briefly introduced to provide an understanding of anisotropic particle morphology. EXPERT OPINION Anisotropic particles exhibit desirable properties such as enhanced circulation time and efficient tumor penetration that could serve as an enlightenment in the exploitation of novel non-spherical nanocarriers to clinical therapy. Yet, current understanding of how anisotropic particles interact with organism is insufficient, which restricts the biomedical application of anisotropic particles. Further work is desired for the development of practical fabrication of anisotropic particles, quantitative analysis of particle morphology, as well as profound understanding of new targeting mechanism and intratumoral penetration of anisotropic particles.
Collapse
Affiliation(s)
- Zhen Wang
- a Department of Pharmaceutics , China Pharmaceutical University , Nanjing , PR China.,b Drug Discovery Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Zimei Wu
- c School of Pharmacy , University of Auckland , Auckland , New Zealand
| | - Jianping Liu
- a Department of Pharmaceutics , China Pharmaceutical University , Nanjing , PR China
| | - Wenli Zhang
- a Department of Pharmaceutics , China Pharmaceutical University , Nanjing , PR China
| |
Collapse
|
25
|
Ekladious I, Liu R, Zhang H, Foil DH, Todd DA, Graf TN, Padera RF, Oberlies NH, Colson YL, Grinstaff MW. Synthesis of poly(1,2-glycerol carbonate)-paclitaxel conjugates and their utility as a single high-dose replacement for multi-dose treatment regimens in peritoneal cancer. Chem Sci 2017; 8:8443-8450. [PMID: 29619192 PMCID: PMC5863611 DOI: 10.1039/c7sc03501b] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022] Open
Abstract
A high drug-density, biodegradable polymeric nanocarrier replaces multi-dose paclitaxel treatment regimens.
Current chemotherapeutic dosing strategies are limited by the toxicity of anticancer agents and therefore rely on multiple low-dose administrations. As an alternative, we describe a novel sustained-release, biodegradable polymeric nanocarrier as a single administration replacement of multi-dose paclitaxel (PTX) treatment regimens. The first synthesis of poly(1,2-glycerol carbonate)-graft-succinic acid-paclitaxel (PGC–PTX) is described, and its use enables high, controlled PTX loadings of up to 74 wt%. Moreover, the polymer backbone is composed of biocompatible building blocks—glycerol and carbon dioxide. When formulated as nanoparticles (NPs), PGC–PTX NPs exhibit PTX concentrations >15 mg mL–1, sub-100 nm diameters, narrow dispersity, storage stability for up to 6 months, and sustained and controlled PTX release kinetics over an extended period of 70 days. A safely administered single dose of PGC–PTX NPs contains more PTX than the median lethal dose of standard PTX. In murine models of peritoneal carcinomatosis, in which the clinical implementation of multi-dose intraperitoneal (IP) treatment regimens is limited by catheter-related complications, PGC–PTX NPs exhibit improved safety at high doses, tumor localization, and efficacy even after a single IP injection, with comparable curative effect to PTX administered as a multi-dose IP treatment regimen.
Collapse
Affiliation(s)
- Iriny Ekladious
- Departments of Biomedical Engineering and Chemistry , Boston University , Boston , MA 02215 , USA .
| | - Rong Liu
- Department of Surgery , Brigham and Women's Hospital , Boston , MA 02215 , USA .
| | - Heng Zhang
- Departments of Biomedical Engineering and Chemistry , Boston University , Boston , MA 02215 , USA .
| | - Daniel H Foil
- Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC 27402 , USA
| | - Daniel A Todd
- Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC 27402 , USA
| | - Tyler N Graf
- Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC 27402 , USA
| | - Robert F Padera
- Department of Pathology , Brigham and Women's Hospital , Boston , MA 02215 , USA
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry , University of North Carolina at Greensboro , Greensboro , NC 27402 , USA
| | - Yolonda L Colson
- Department of Surgery , Brigham and Women's Hospital , Boston , MA 02215 , USA .
| | - Mark W Grinstaff
- Departments of Biomedical Engineering and Chemistry , Boston University , Boston , MA 02215 , USA .
| |
Collapse
|
26
|
Odiba A, Ottah V, Ottah C, Anunobi O, Ukegbu C, Edeke A, Uroko R, Omeje K. Therapeutic nanomedicine surmounts the limitations of pharmacotherapy. Open Med (Wars) 2017. [DOI: 10.1515/med-2017-0041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AbstractScience always strives to find an improved way of doing things and nanoscience is one such approach. Nanomaterials are suitable for pharmaceutical applications mostly because of their size which facilitates absorption, distribution, metabolism and excretion of the nanoparticles. Whether labile or insoluble nanoparticles, their cytotoxic effect on malignant cells has moved the use of nanomedicine into focus. Since nanomedicine can be described as the science and technology of diagnosing, treating and preventing diseases towards ultimately improving human health, a lot of nanotechnology options have received approval by various regulatory agencies. Nanodrugs also have been discovered to be more precise in targeting the desired site, hence maximizing the therapeutic effects, while minimizing side-effects on the rest of the body. This unique property and more has made nanomedicine popular in therapeutic medicine employing nanotechnology in genetic therapy, drug encapsulation, enzyme manipulation and control, tissue engineering, target drug delivery, pharmacogenomics, stem cell and cloning, and even virus-based hybrids. This review highlights nanoproducts that are in development and have gained approval through one clinical trial stage or the other.
Collapse
Affiliation(s)
- Arome Odiba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - Victoria Ottah
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - Comfort Ottah
- 4Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Usman Danfodio University, Sokoto, Nigeria
| | - Ogechukwu Anunobi
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
- Department of Biochemistry, Faculty of Science and Technology, Bingham University Karu, Nasarawa State, Nigeria
| | - Chimere Ukegbu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - Affiong Edeke
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - Robert Uroko
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
- Department of Biochemistry, Faculty of Science, Michael Okpara University of Agriculture, Umudike, Nigeria
| | - Kingsley Omeje
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
27
|
Zhao J, Koay EJ, Li T, Wen X, Li C. A hindsight reflection on the clinical studies of poly(l-glutamic acid)-paclitaxel. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10:e1497. [PMID: 28895304 DOI: 10.1002/wnan.1497] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 11/06/2022]
Abstract
Chemotherapy for cancer treatment is limited by the excessive toxicity to normal tissues. The design of chemodrug-loaded nanoformulations provides a unique approach to improve the treatment efficacy while minimizing toxicity. Despite the numerous publications of nanomedicine for the last several decades, however, only a small fraction of the developed nanoformulations have entered clinical trials, with even fewer being approved for clinical application. Poly(l-glutamic acid)-paclitaxel (PG-TXL) belongs to the few formulations that reached phase III clinical trials. Unfortunately, the development of PG-TXL stopped in 2016 due to the inability to show significant improvement over current standard care. This review will provide an overview of the preclinical and clinical evaluations of PG-TXL, and discuss lessons to be learned from this ordeal. The precise identification of suitable patients for clinical trial studies, deep understanding of the mechanisms of action, and an effective academic-industry partnership throughout all phases of drug development are important for the successful bench-to-bedside translation of new nanoformulations. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Peptide-Based Structures.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tingting Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoxia Wen
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
28
|
Scomparin A, Florindo HF, Tiram G, Ferguson EL, Satchi-Fainaro R. Two-step polymer- and liposome-enzyme prodrug therapies for cancer: PDEPT and PELT concepts and future perspectives. Adv Drug Deliv Rev 2017; 118:52-64. [PMID: 28916497 DOI: 10.1016/j.addr.2017.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/17/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022]
Abstract
Polymer-directed enzyme prodrug therapy (PDEPT) and polymer enzyme liposome therapy (PELT) are two-step therapies developed to provide anticancer drugs site-selective intratumoral accumulation and release. Nanomedicines, such as polymer-drug conjugates and liposomal drugs, accumulate in the tumor site due to extravasation-dependent mechanism (enhanced permeability and retention - EPR - effect), and further need to cross the cellular membrane and release their payload in the intracellular compartment. The subsequent administration of a polymer-enzyme conjugate able to accumulate in the tumor tissue and to trigger the extracellular release of the active drug showed promising preclinical results. The development of polymer-enzyme, polymer-drug conjugates and liposomal drugs had undergone a vast advancement over the past decades. Several examples of enzyme mimics for in vivo therapy can be found in the literature. Moreover, polymer therapeutics often present an enzyme-sensitive mechanism of drug release. These nanomedicines can thus be optimal substrates for PDEPT and this review aims to provide new insights and stimuli toward the future perspectives of this promising combination.
Collapse
Affiliation(s)
- Anna Scomparin
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Elaine L Ferguson
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
29
|
Duncan R. Polymer therapeutics at a crossroads? Finding the path for improved translation in the twenty-first century. J Drug Target 2017; 25:759-780. [PMID: 28783978 DOI: 10.1080/1061186x.2017.1358729] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Despite the relatively small early investment, first generation 'polymer therapeutics' have been remarkably successful with more than 25 products licenced for human use as polymeric drugs, sequestrants, conjugates, and as an imaging agent. Many exhibit both clinical and commercial success with new concepts already in clinical trials. Nevertheless after four decades of evolution, this field is arriving at an important crossroads. Over the last decade, the landscape has changed rapidly. There are an increasing number of failed clinical trials, the number of 'copy' and 'generic' products is growing (danger of ignoring the biological rationale for design and suppression of innovation), potential drawbacks of PEG are becoming more evident, and the 'nanomedicine' boom has brought danger of loss of scientific focus/hype. Grasping opportunities provided by advances in understanding of the patho-physiology and molecular basis of diseases, new polymer/conjugate synthetic and analytical methods, as well as the large database of clinical experience will surely ensure a successful future for innovative polymer therapeutics. Progress will, however, be in jeopardy if polymer safety is overlooked in respect of the specific route of administration/clinical use, poorly characterised materials/formulations are used to define biological or early clinical properties, and if clinical trial protocols fail to select patients most likely to benefit from these macromolecular therapeutics. Opportunities to improve clinical trial design for polymer-anticancer drug conjugates are discussed. This short personal perspective summarises some of the important challenges facing polymer therapeutics in R&D today, and future opportunities to improve successful translation.
Collapse
Affiliation(s)
- Ruth Duncan
- a Polymer Therapeutics Laboratory , Centro de Investigación Príncipe Felipe , Valencia , Spain.,b Intracellular Delivery Solutions Laboratory, Faculty of Engineering and Science , University of Greenwich , Kent , UK
| |
Collapse
|
30
|
Lucas AT, Price LS, Schorzman A, Zamboni WC. Complex effects of tumor microenvironment on the tumor disposition of carrier-mediated agents. Nanomedicine (Lond) 2017; 12:2021-2042. [PMID: 28745129 DOI: 10.2217/nnm-2017-0101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Major advances in carrier-mediated agents, including nanoparticle, conjugates and antibody-drug conjugates, have created revolutionary drug delivery systems in cancer over the past two decades. While these agents provide several advantages, such as greater duration of exposure and solubility, compared with their small-molecule counterparts, there is substantial variability in delivery of these agents to tissues and especially tumors. This review provides an overview of tumor microenvironment factors that affect the pharmacokinetics and pharmacodynamics of carrier-mediated agents observed in preclinical models and patients.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lauren Sl Price
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Allison Schorzman
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C Zamboni
- Division of Pharmacotherapy & Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Carolina Institute for Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
31
|
Kramer L, Turk D, Turk B. The Future of Cysteine Cathepsins in Disease Management. Trends Pharmacol Sci 2017; 38:873-898. [PMID: 28668224 DOI: 10.1016/j.tips.2017.06.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/23/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023]
Abstract
Since the discovery of the key role of cathepsin K in bone resorption, cysteine cathepsins have been investigated by pharmaceutical companies as drug targets. The first clinical results from targeting cathepsins by activity-based probes and substrates are paving the way for the next generation of molecular diagnostic imaging, whereas the majority of antibody-drug conjugates currently in clinical trials depend on activation by cathepsins. Finally, cathepsins have emerged as suitable vehicles for targeted drug delivery. It is therefore timely to review the future of cathepsins in drug discovery. We focus here on inflammation-associated diseases because dysregulation of the immune system accompanied by elevated cathepsin activity is a common feature of these conditions.
Collapse
Affiliation(s)
- Lovro Kramer
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, 1000 Ljubljana, Slovenia
| | - Dušan Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKEBIP, Jamova 39, 1000 Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKEBIP, Jamova 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia.
| |
Collapse
|
32
|
El-Sherbiny I, Khalil I, Ali I, Yacoub M. Updates on smart polymeric carrier systems for protein delivery. Drug Dev Ind Pharm 2017; 43:1567-1583. [DOI: 10.1080/03639045.2017.1338723] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ibrahim El-Sherbiny
- Center for Materials Science, University of Science and Technology (UST), Zewail City of Science and Technology, Cairo, Egypt
| | - Islam Khalil
- Center for Materials Science, University of Science and Technology (UST), Zewail City of Science and Technology, Cairo, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Misr University of Science and Technology (MUST), Cairo, Egypt
| | - Isra Ali
- Center for Materials Science, University of Science and Technology (UST), Zewail City of Science and Technology, Cairo, Egypt
| | - Magdi Yacoub
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
33
|
Paclitaxel: What has been done and the challenges remain ahead. Int J Pharm 2017; 526:474-495. [DOI: 10.1016/j.ijpharm.2017.05.016] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/05/2017] [Accepted: 05/06/2017] [Indexed: 12/17/2022]
|
34
|
Howard D, Garcia-Parra J, Healey GD, Amakiri C, Margarit L, Francis LW, Gonzalez D, Conlan RS. Antibody-drug conjugates and other nanomedicines: the frontier of gynaecological cancer treatment. Interface Focus 2016; 6:20160054. [PMID: 27920893 PMCID: PMC5071815 DOI: 10.1098/rsfs.2016.0054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Gynaecological cancers: malignancies of the cervix, uterus, ovaries, vagina and vulva, are responsible for over 1.1 million new cancer cases and almost half a million deaths annually. Ovarian cancer in particular is difficult to treat due to often being diagnosed at a late stage, and the incidence of uterine and vulvar malignancies are both on the rise. The field of nanomedicine is beginning to introduce drugs into the clinic for oncological applications exemplified by the liposomal drugs, Doxil and Myocet, the nanoparticle, Abraxane and antibody-drug conjugates (ADCs), Kadcyla and Adcetris. With many more agents currently undergoing clinical trials, the field of nanomedicine promises to have a significant impact on cancer therapy. This review considers the state of the art for nanomedicines currently on the market and those being clinically evaluated for the treatment of gynaecological cancers. In particular, it focuses on ADCs and presents a methodology for their rational design and evaluation.
Collapse
Affiliation(s)
- David Howard
- Swansea University Medical School, Swansea SA2 8PP, UK
| | | | | | | | - Lavinia Margarit
- Abertawe Bro Morannwg University Health Board, Obstetrics & Gynecology Department Princess of Wales Hospital, Bridgend, CF31 1RQ, UK
| | | | | | | |
Collapse
|
35
|
Zagorodko O, Arroyo-Crespo JJ, Nebot VJ, Vicent MJ. Polypeptide-Based Conjugates as Therapeutics: Opportunities and Challenges. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600316] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/02/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Oleksandr Zagorodko
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| | - Juan José Arroyo-Crespo
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| | - Vicent J. Nebot
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
- Polypeptide Therapeutic Solutions SL; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| |
Collapse
|
36
|
Nanocarriers in cancer clinical practice: a pharmacokinetic issue. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:583-599. [PMID: 27520727 DOI: 10.1016/j.nano.2016.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/22/2016] [Accepted: 07/23/2016] [Indexed: 01/24/2023]
Abstract
The advent of nanocarriers for drug delivery has given rise to new intriguing scenarios in the cancer field. Nanocarriers indeed partly overcome the limits of traditional cytotoxic drugs principally changing the pharmacokinetic behavior of the parental drug. The peculiar characteristics of these systems strongly minimize the adverse reactions and ensure a more precise release of the compound to the tumor site. Several nanocarriers have been developed for the delivery of cytotoxic drugs such as paclitaxel and doxorubicin in order to improve both the outcome and the patients' quality of life. The aims of this review are to describe in detail the pharmacokinetics of nanocarriers, already marketed or in advanced clinical phases, for paclitaxel and doxorubicin, to highlight the main differences with the parental drugs, and to underline, in a critical manner, benefits and disadvantages related to the use of these new drug delivery systems.
Collapse
|
37
|
Krivitsky A, Polyak D, Scomparin A, Eliyahu S, Ori A, Avkin-Nachum S, Krivitsky V, Satchi-Fainaro R. Structure–Function Correlation of Aminated Poly(α)glutamate as siRNA Nanocarriers. Biomacromolecules 2016; 17:2787-800. [DOI: 10.1021/acs.biomac.6b00555] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Adva Krivitsky
- Department
of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dina Polyak
- Department
of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department
of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shay Eliyahu
- Department
of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Asaf Ori
- QBI Enterprise, Ltd., Ness-Ziona 70400, Israel
| | | | - Vadim Krivitsky
- School of Chemistry, the Raymond and Beverly Sackler Faculty of Exact Sciences, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department
of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
38
|
Liu D, Yang F, Xiong F, Gu N. The Smart Drug Delivery System and Its Clinical Potential. Theranostics 2016; 6:1306-23. [PMID: 27375781 PMCID: PMC4924501 DOI: 10.7150/thno.14858] [Citation(s) in RCA: 604] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/22/2016] [Indexed: 12/22/2022] Open
Abstract
With the unprecedented progresses of biomedical nanotechnology during the past few decades, conventional drug delivery systems (DDSs) have been involved into smart DDSs with stimuli-responsive characteristics. Benefiting from the response to specific internal or external triggers, those well-defined nanoplatforms can increase the drug targeting efficacy, in the meantime, reduce side effects/toxicities of payloads, which are key factors for improving patient compliance. In academic field, variety of smart DDSs have been abundantly demonstrated for various intriguing systems, such as stimuli-responsive polymeric nanoparticles, liposomes, metals/metal oxides, and exosomes. However, these nanoplatforms are lack of standardized manufacturing method, toxicity assessment experience, and clear relevance between the pre-clinical and clinical studies, resulting in the huge difficulties to obtain regulatory and ethics approval. Therefore, such relatively complex stimulus-sensitive nano-DDSs are not currently approved for clinical use. In this review, we highlight the recent advances of smart nanoplatforms for targeting drug delivery. Furthermore, the clinical translation obstacles faced by these smart nanoplatforms have been reviewed and discussed. We also present the future directions and perspectives of stimuli-sensitive DDS in clinical applications.
Collapse
Affiliation(s)
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210009, China
| | | | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210009, China
| |
Collapse
|
39
|
Ulbrich K, Holá K, Šubr V, Bakandritsos A, Tuček J, Zbořil R. Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies. Chem Rev 2016; 116:5338-431. [DOI: 10.1021/acs.chemrev.5b00589] [Citation(s) in RCA: 1120] [Impact Index Per Article: 124.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Karel Ulbrich
- Institute
of Macromolecular Chemistry, The Czech Academy of Sciences, v.v.i., Heyrovsky Square 2, 162 06 Prague 6, Czech Republic
| | - Kateřina Holá
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Vladimir Šubr
- Institute
of Macromolecular Chemistry, The Czech Academy of Sciences, v.v.i., Heyrovsky Square 2, 162 06 Prague 6, Czech Republic
| | - Aristides Bakandritsos
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Jiří Tuček
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Radek Zbořil
- Regional
Centre of Advanced Technologies and Materials, Department of Physical
Chemistry, Faculty of Science, Palacky University, 17 Listopadu 1192/12, 771 46 Olomouc, Czech Republic
| |
Collapse
|
40
|
Hung CC, Huang WC, Lin YW, Yu TW, Chen HH, Lin SC, Chiang WH, Chiu HC. Active Tumor Permeation and Uptake of Surface Charge-Switchable Theranostic Nanoparticles for Imaging-Guided Photothermal/Chemo Combinatorial Therapy. Theranostics 2016; 6:302-17. [PMID: 26909107 PMCID: PMC4737719 DOI: 10.7150/thno.13686] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/29/2015] [Indexed: 12/13/2022] Open
Abstract
To significantly promote tumor uptake and penetration of therapeutics, a nanovehicle system comprising poly(lactic-co-glycolic acid) (PLGA) as the hydrophobic cores coated with pH-responsive N-acetyl histidine modified D-α-tocopheryl polyethylene glycol succinate (NAcHis-TPGS) is developed in this work. The nanocarriers with switchable surface charges in response to tumor extracellular acidity (pHe) were capable of selectively co-delivering indocyanine green (ICG), a photothermal agent, and doxorubicin (DOX), a chemotherapy drug, to tumor sites. The in vitro cellular uptake of ICG/DOX-loaded nanoparticles by cancer cells and macrophages was significantly promoted in weak acidic environments due to the increased protonation of the NAcHis moieties. The results of in vivo and ex vivo biodistribution studies demonstrated that upon intravenous injection the theranostic nanoparticles were substantially accumulated in TRAMP-C1 solid tumor of tumor-bearing mice. Immunohistochemical examination of tumor sections confirmed the active permeation of the nanoparticles into deep tumor hypoxia due to their small size, pHe-induced near neutral surface, and the additional hitchhiking transport via tumor-associated macrophages. The prominent imaging-guided photothermal therapy of ICG/DOX-loaded nanoparticles after tumor accumulation induced extensive tumor tissue/vessel ablation, which further promoted their extravasation and DOX tumor permeation, thus effectively suppressing tumor growth.
Collapse
|
41
|
Shirbin SJ, Ladewig K, Fu Q, Klimak M, Zhang X, Duan W, Qiao GG. Cisplatin-Induced Formation of Biocompatible and Biodegradable Polypeptide-Based Vesicles for Targeted Anticancer Drug Delivery. Biomacromolecules 2015; 16:2463-74. [PMID: 26166192 DOI: 10.1021/acs.biomac.5b00692] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Novel cisplatin (CDDP)-loaded, polypeptide-based vesicles for the targeted delivery of cisplatin to cancer cells have been prepared. These vesicles were formed from biocompatible and biodegradable maleimide-poly(ethylene oxide)114-b-poly(L-glutamic acid)12 (Mal-PEG114-b-PLG12) block copolymers upon conjugation with the drug itself. CDDP conjugation forms a short, rigid, cross-linked, drug-loaded, hydrophobic block in the copolymer, and subsequently induces self-assembly into hollow vesicle structures with average hydrodynamic diameters (Dh) of ∼ 270 nm. CDDP conjugation is critical to the formation of the vesicles. The reactive maleimide-PEG moieties that form the corona and inner layer of the vesicles were protected via formation of a reversible Diels-Alder (DA) adduct throughout the block copolymer synthesis so as to maintain their integrity. Drug release studies demonstrated a low and sustained drug release profile in systemic conditions (pH = 7.4, [Cl(-)] = 140 mM) with a higher "burst-like" release rate being observed under late endosomal/lysosomal conditions (pH = 5.2, [Cl(-)] = 35 mM). Further, the peripheral maleimide functionalities on the vesicle corona were conjugated to thiol-functionalized folic acid (FA) (via in situ reduction of a novel bis-FA disulfide, FA-SS-FA) to form an active targeting drug delivery system. These targeting vesicles exhibited significantly higher cellular binding/uptake into and dose-dependent cytotoxicity toward cancer cells (HeLa) compared to noncancerous cells (NIH-3T3), which show high and low folic acid receptor (FR) expression, respectively. This work thus demonstrates a novel approach to polypeptide-based vesicle assembly and a promising strategy for targeted, effective CDDP anticancer drug delivery.
Collapse
Affiliation(s)
- Steven J Shirbin
- †Polymer Science Group, Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Katharina Ladewig
- †Polymer Science Group, Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Qiang Fu
- †Polymer Science Group, Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Molly Klimak
- †Polymer Science Group, Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Xiaoqing Zhang
- ‡CSIRO Manufacturing Flagship, Clayton South, Victoria 3169, Australia
| | - Wei Duan
- §School of Medicine, Deakin University, Geelong, Victoria 3216, Australia
| | - Greg G Qiao
- †Polymer Science Group, Department of Chemical and Biomolecular Engineering, University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| |
Collapse
|
42
|
Friberg S, Nyström AM. Nanotechnology in the war against cancer: new arms against an old enemy – a clinical view. Future Oncol 2015; 11:1961-75. [DOI: 10.2217/fon.15.91] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
ABSTRACT Clinical oncology is facing a paradigm shift. A new treatment philosophy is emerging and new targets are appearing that require new active agents. The medical use of nanotechnology – nanomedicine – holds several promising possibilities in the war against cancer. Some of these include: new formats for old drugs, that is, increasing efficacy while diminishing side effects; and new administration routes – that is, dermal, oral and pulmonary. In this overview, we describe some nanoparticles and their medical uses as well as highlight advantages of nanoparticles compared with conventional pharmaceuticals. We also point to some of the many technical challenges and potential risks with using nanotechnology for oncological applications.
Collapse
Affiliation(s)
- Sten Friberg
- Swedish Medical Nanoscience Center, Department of Neuroscience, Retzius väg 8, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Andreas M Nyström
- Institute of Environmental Medicine, Nobels väg 13, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
43
|
|
44
|
Li Q, Lv S, Tang Z, Liu M, Zhang D, Yang Y, Chen X. A co-delivery system based on paclitaxel grafted mPEG-b-PLG loaded with doxorubicin: Preparation, in vitro and in vivo evaluation. Int J Pharm 2014; 471:412-20. [DOI: 10.1016/j.ijpharm.2014.05.065] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/30/2014] [Accepted: 05/31/2014] [Indexed: 01/26/2023]
|
45
|
Peng L, Schorzman AN, Ma P, Madden AJ, Zamboni WC, Benhabbour SR, Mumper RJ. 2'-(2-bromohexadecanoyl)-paclitaxel conjugate nanoparticles for the treatment of non-small cell lung cancer in an orthotopic xenograft mouse model. Int J Nanomedicine 2014; 9:3601-10. [PMID: 25114529 PMCID: PMC4124067 DOI: 10.2147/ijn.s66040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A nanoparticle (NP) formulation with 2'-(2-bromohexadecanoyl)-paclitaxel (Br-16-PX) conjugate was developed in these studies for the treatment of non-small cell lung cancer (NSCLC). The lipophilic paclitaxel conjugate Br-C16-PX was synthesized and incorporated into lipid NPs where the 16-carbon chain enhanced drug entrapment in the drug delivery system and improved in vivo pharmacokinetics. The electron-withdrawing bromine group was used to facilitate the conversion of Br-C16-PX to paclitaxel at the tumor site. The developed system was evaluated in luciferase-expressing A549 cells in vitro and in an orthotopic NSCLC mouse model. The results demonstrated that the Br-C16-PX NPs had a higher maximum tolerated dose (75 mg/kg) than Taxol (19 mg/kg) and provided significantly longer median survival (88 days versus 70 days, P<0.05) in the orthotopic NSCLC model. An improved pharmacokinetic profile was observed for the Br-C16-PX NPs at 75 mg/kg compared to Taxol at 19 mg/kg. The area under the concentration versus time curve (AUC)₀₋₉₆ h of Br-C16-PX from the NPs was 91.7-fold and 49.6-fold greater than Taxol in plasma and tumor-bearing lungs, respectively, which provided sustained drug exposure and higher antitumor efficacy in the NP-treated group.
Collapse
Affiliation(s)
- Lei Peng
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Ping Ma
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Andrew J Madden
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Soumya Rahima Benhabbour
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| | - Russell J Mumper
- Center for Nanotechnology in Drug Delivery, Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, NC, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina, NC, USA
| |
Collapse
|
46
|
Ahmad Z, Tang Z, Shah A, Lv S, Zhang D, Zhang Y, Chen X. Cisplatin Loaded Methoxy Poly (ethylene glycol)-block
-Poly (L
-glutamic acid-co
-L
-Phenylalanine) Nanoparticles against Human Breast Cancer Cell. Macromol Biosci 2014; 14:1337-45. [DOI: 10.1002/mabi.201400109] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/19/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Zaheer Ahmad
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
- Department of Chemistry; Quaid-I-Azam University; Islamabad 45320 Pakistan
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Afzal Shah
- Department of Chemistry; Quaid-I-Azam University; Islamabad 45320 Pakistan
| | - Shixian Lv
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Ying Zhang
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 P. R. China
| |
Collapse
|
47
|
Reddy LH, Bazile D. Drug delivery design for intravenous route with integrated physicochemistry, pharmacokinetics and pharmacodynamics: illustration with the case of taxane therapeutics. Adv Drug Deliv Rev 2014; 71:34-57. [PMID: 24184489 DOI: 10.1016/j.addr.2013.10.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/22/2013] [Accepted: 10/24/2013] [Indexed: 12/12/2022]
Abstract
This review is aimed at combining the published data on taxane formulations into a generalized Drug Delivery approach, starting from the physicochemistry and assessing its relationships with the pharmacokinetics, the biodistribution and the pharmacodynamics. Owing to the number and variety of taxane formulation designs, we considered this class of cytotoxic anticancer agents of particular interest to illustrate the concepts attached to this approach. According to the history of taxane development, we propose a classification as (i) "surfactant-based formulations" first generation, (ii) "surfactant-free formulations" second generation and (iii) "modulated pharmacokinetics drug delivery systems" third generation. Since our objective was to make the link between (i) the physicochemistry of the drug and carrier and (ii) the efficacy and safety of the drug in preclinical animal models and (iii) in human, we focused on the drug delivery technologies that were tested in clinic.
Collapse
Affiliation(s)
- L Harivardhan Reddy
- Drug Delivery Technologies and Innovation, Pharmaceutical Sciences Department, Sanofi Research and Development, 13 Quai Jules-Guesde, 94403 Vitry-sur-Seine, France.
| | - Didier Bazile
- Drug Delivery Technologies and Innovation, Pharmaceutical Sciences Department, Sanofi Research and Development, 13 Quai Jules-Guesde, 94403 Vitry-sur-Seine, France
| |
Collapse
|
48
|
|
49
|
Wang Y, Liu J, Zhang J, Wang L, Chan J, Wang H, Jin Y, Yu L, Grainger DW, Ying W. A cell-based pharmacokinetics assay for evaluating tubulin-binding drugs. Int J Med Sci 2014; 11:479-87. [PMID: 24688312 PMCID: PMC3970101 DOI: 10.7150/ijms.8340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/27/2014] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence reveals that traditional pharmacokinetics parameters based on plasma drug concentrations are insufficient to reliably demonstrate accurate pharmacological effects of drugs in target organs or cells in vivo. This underscores the increasing need to improve the types and qualities of cellular pharmacokinetic information for drug preclinical screening and clinical efficacy assessments. Here we report a whole cell-based method to assess drugs that disturb microtubule dynamics to better understand different formulation-mediated intracellular drug release profiles. As proof of concept for this approach, we compared the well-known taxane class of anti-microtubule drugs based on paclitaxel (PTX), including clinically familiar albumin nanoparticle-based Abraxane™, and a polymer nanoparticle-based degradable paclitaxel carrier, poly(L-glutamic acid)-paclitaxel conjugate (PGA-PTX, also known as CT-2103) versus control PTX. This in vitro cell-based evaluation of PTX efficacy includes determining the cellular kinetics of tubulin polymerization, relative populations of cells under G2 mitotic arrest, cell proliferation and total cell viability. For these taxane tubulin-binding compounds, the kinetics of cell microtubule stabilization directly correlate with G2 arrest and cell proliferation, reflecting the kinetics and amounts of intracellular PTX release. Each individual cell-based dose-response experiment correlates with published, key therapeutic parameters and taken together, provide a comprehensive understanding of drug intracellular pharmacokinetics at both cellular and molecular levels. This whole cell-based evaluating method is convenient, quantitative and cost-effective for evaluating new formulations designed to optimize cellular pharmacokinetics for drugs perturbing tubulin polymerization as well as assisting in explaining drug mechanisms of action at cellular levels.
Collapse
Affiliation(s)
- Yuwei Wang
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Jihua Liu
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Jun Zhang
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Liping Wang
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Jonathon Chan
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Hai Wang
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Yi Jin
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - Lei Yu
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| | - David W Grainger
- 2. Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, UT 84112, USA
| | - Wenbin Ying
- 1. Molecular Therapeutics Department, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92054, USA
| |
Collapse
|
50
|
Mishra D, Hubenak JR, Mathur AB. Nanoparticle systems as tools to improve drug delivery and therapeutic efficacy. J Biomed Mater Res A 2013; 101:3646-60. [PMID: 23878102 DOI: 10.1002/jbm.a.34642] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 02/04/2013] [Accepted: 02/05/2013] [Indexed: 02/03/2023]
Abstract
Nanoparticle-based drug delivery systems are appealing because, among other properties, they are easily manufactured and have the capacity to encapsulate a wide variety of drugs, many of which are not directly miscible with water. This review classifies nanoparticles into three broad categories based upon material composition: bio-inspired systems, synthetic systems, and inorganic systems. Each has distinct properties suitable for drug delivery applications, including their structure, composition, and pharmacokinetics (including clearance and uptake mechanisms), making each uniquely suitable for certain types of drugs. Furthermore, nanoparticles can be customized, making them ideal for a variety of applications. Advantages and disadvantages of the different systems are discussed. Strategies for improving nanoparticle efficacy include adding targeting agents on the nanoparticle surface, altering the degradation profile to control drug release, or PEGylating the surface to increase circulation times and reduce immediate clearance by the kidneys. The future of nanoparticle systems seems to be focused on further improving overall patient outcome by increasing delivery accuracy to the target area.
Collapse
Affiliation(s)
- Deepa Mishra
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 602, Houston, Texas, 77030
| | | | | |
Collapse
|