1
|
Shen Y, Wang J, Li Y, Yang CT, Zhou X. Modified Bacteriophage for Tumor Detection and Targeted Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13040665. [PMID: 36839030 PMCID: PMC9963578 DOI: 10.3390/nano13040665] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 05/07/2023]
Abstract
Malignant tumor is one of the leading causes of death in human beings. In recent years, bacteriophages (phages), a natural bacterial virus, have been genetically engineered for use as a probe for the detection of antigens that are highly expressed in tumor cells and as an anti-tumor reagent. Furthermore, phages can also be chemically modified and assembled with a variety of nanoparticles to form a new organic/inorganic composite, thus extending the application of phages in biological detection and tumor therapeutic. This review summarizes the studies on genetically engineered and chemically modified phages in the diagnosis and targeting therapy of tumors in recent years. We discuss the advantages and limitations of modified phages in practical applications and propose suitable application scenarios based on these modified phages.
Collapse
Affiliation(s)
- Yuanzhao Shen
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jingyu Wang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yuting Li
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
| | - Chih-Tsung Yang
- Future Industries Institute, Mawson Lakes Campus, University of South Australia, Adelaide, SA 5095, Australia
- Correspondence: (X.Z.); (C.-T.Y.)
| | - Xin Zhou
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (X.Z.); (C.-T.Y.)
| |
Collapse
|
2
|
Mayorga C, Perez‐Inestrosa E, Rojo J, Ferrer M, Montañez MI. Role of nanostructures in allergy: Diagnostics, treatments and safety. Allergy 2021; 76:3292-3306. [PMID: 33559903 DOI: 10.1111/all.14764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 01/08/2023]
Abstract
Nanotechnology is science, engineering and technology conducted at the nanoscale, which is about 1-100 nm. It has led to the development of nanomaterials, which behave very differently from materials with larger scales and can have a wide range of applications in biomedicine. The physical and chemical properties of materials of such small compounds depend mainly on the size, shape, composition and functionalization of the system. Nanoparticles, carbon nanotubes, liposomes, polymers, dendrimers and nanogels, among others, can be nanoengineeried for controlling all parameters, including their functionalization with ligands, which provide the desired interaction with the immunological system, that is dendritic cell receptors to activate and/or modulate the response, as well as specific IgE, or effector cell receptors. However, undesired issues related to toxicity and hypersensitivity responses can also happen and would need evaluation. There are wide panels of accessible structures, and controlling their physico-chemical properties would permit obtaining safer and more efficient compounds for clinical applications goals, either in diagnosis or treatment. The application of dendrimeric antigens, nanoallergens and nanoparticles in allergy diagnosis is very promising since it can improve sensitivity by increasing specific IgE binding, mimicking carrier proteins or enhancing signal detection. Additionally, in the case of immunotherapy, glycodendrimers, liposomes, polymers and nanoparticles have shown interest, behaving as platforms of allergenic structures, adjuvants or protectors of allergen from degradation or having a depot capacity. Taken together, the application of nanotechnology to allergy shows promising facts facing important goals related to the improvement of diagnosis as well as specific immunotherapy.
Collapse
Affiliation(s)
- Cristobalina Mayorga
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA Málaga Spain
- Allergy Unit Hospital Regional Universitario de Málaga Málaga Spain
- Andalusian Centre for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
| | - Ezequiel Perez‐Inestrosa
- Andalusian Centre for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
- Departamento de Química Orgánica, and the Biomimetic Dendrimers and Photonic Laboratory Instituto de Investigación Biomédica de Málaga‐IBIMAUniversidad de Málaga Málaga Spain
| | - Javier Rojo
- Glycosystems Laboratory Instituto de Investigaciones Químicas (IIQ)CSIC—Universidad de Sevilla Sevilla Spain
| | - Marta Ferrer
- Department of Allergy and Clinical Immunology Clínica Universidad de NavarraInstituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona Spain
| | - Maria Isabel Montañez
- Allergy Research Group Instituto de Investigación Biomédica de Málaga‐IBIMA Málaga Spain
- Andalusian Centre for Nanomedicine and Biotechnology‐BIONAND Málaga Spain
| |
Collapse
|
3
|
Ulldemolins A, Seras-Franzoso J, Andrade F, Rafael D, Abasolo I, Gener P, Schwartz S. Perspectives of nano-carrier drug delivery systems to overcome cancer drug resistance in the clinics. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:44-68. [PMID: 35582007 PMCID: PMC9019183 DOI: 10.20517/cdr.2020.59] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/21/2022]
Abstract
Advanced cancer is still considered an incurable disease because of its metastatic spread to distal organs and progressive gain of chemoresistance. Even though considerable treatment progress and more effective therapies have been achieved over the past years, recurrence in the long-term and undesired side effects are still the main drawbacks of current clinical protocols. Moreover, a majority of chemotherapeutic drugs are highly hydrophobic and need to be diluted in organic solvents, which cause high toxicity, in order to reach effective therapeutic dose. These limitations of conventional cancer therapies prompted the use of nanomedicine, the medical application of nanotechnology, to provide more effective and safer cancer treatment. Potential of nanomedicines to overcome resistance, ameliorate solubility, improve pharmacological profile, and reduce adverse effects of chemotherapeutical drugs is thus highly regarded. Their use in the clinical setting has increased over the last decade. Among the various existing nanosystems, nanoparticles have the ability to transform conventional medicine by reducing the adverse effects and providing a controlled release of therapeutic agents. Also, their small size facilitates the intracellular uptake. Here, we provide a closer review of clinical prospects and mechanisms of action of nanomedicines to overcome drug resistance. The significance of specific targeting towards cancer cells is debated as well.
Collapse
Affiliation(s)
- Anna Ulldemolins
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Joaquin Seras-Franzoso
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| | - Fernanda Andrade
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain
| | - Diana Rafael
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain
| | - Ibane Abasolo
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain.,Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain
| | - Petra Gener
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain.,Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain
| | - Simo Schwartz
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona 08035, Spain.,Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain
| |
Collapse
|
4
|
Gabriel EM, Kim M, Fisher DT, Mangum C, Attwood K, Ji W, Mukhopadhyay D, Bagaria SP, Robertson MW, Dinh TA, Knutson KL, Skitzki JJ, Wallace MB. A pilot trial of intravital microscopy in the study of the tumor vasculature of patients with peritoneal carcinomatosis. Sci Rep 2021; 11:4946. [PMID: 33654117 PMCID: PMC7925603 DOI: 10.1038/s41598-021-84430-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
Aberrancies in the tumor microvasculature limit the systemic delivery of anticancer agents, which impedes tumor response. Using human intravital microscopy (HIVM), we hypothesized that HIVM would be feasible in patients with peritoneal carcinomatosis (PC). During cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for PC, HIVM was performed in both tumor and non-tumor areas. The primary outcome was HIVM feasibility to measure vessel characteristics. We secondarily evaluated associations between HIVM vessel characteristics and oncologic outcomes (RECIST response to neoadjuvant therapy and disease-specific survival). Thirty patients with PC were enrolled. Nineteen patients (63.3%) received neoadjuvant therapy. HIVM was feasible in all patients. Compared to non-tumor (control) areas, PC areas had a lower density of functional vessels, higher proportion of non-functional vessels, smaller lumenal diameters, and lower blood flow velocity. Qualitative differences in these vessel characteristics were observed among patients who had partial response, stable disease, or progressive disease after receiving neoadjuvant therapy. However, no statistically significant relationships were found between HIVM vessel characteristics and oncologic outcomes. These novel findings comprise the first-in-human, real-time evidence of the microscopic differences between normal and tumor-associated vessels and form the basis for our larger, ongoing clinical trial appropriately powered to determine the clinical utility of HIVM (NCT03823144).
Collapse
Affiliation(s)
- Emmanuel M. Gabriel
- grid.417467.70000 0004 0443 9942Department of Surgery, Section of Surgical Oncology, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Minhyung Kim
- grid.240614.50000 0001 2181 8635Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Daniel T. Fisher
- grid.240614.50000 0001 2181 8635Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Catherine Mangum
- grid.417467.70000 0004 0443 9942Department of Surgery, Section of Surgical Oncology, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Kristopher Attwood
- grid.240614.50000 0001 2181 8635Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Wenyan Ji
- grid.240614.50000 0001 2181 8635Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Debabrata Mukhopadhyay
- grid.417467.70000 0004 0443 9942Department of Molecular Biology, Mayo Clinic, Jacksonville, FL USA
| | - Sanjay P. Bagaria
- grid.417467.70000 0004 0443 9942Department of Surgery, Section of Surgical Oncology, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Matthew W. Robertson
- grid.417467.70000 0004 0443 9942Department of Gynecological Oncology, Mayo Clinic, Jacksonville, FL USA
| | - Tri A. Dinh
- grid.417467.70000 0004 0443 9942Department of Gynecological Oncology, Mayo Clinic, Jacksonville, FL USA
| | - Keith L. Knutson
- grid.417467.70000 0004 0443 9942Department of Immunology, Mayo Clinic, Jacksonville, FL USA
| | - Joseph J. Skitzki
- grid.240614.50000 0001 2181 8635Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Michael B. Wallace
- grid.417467.70000 0004 0443 9942Department of Gastroenterology, Mayo Clinic, Jacksonville, FL USA
| |
Collapse
|
5
|
Gabriel EM, Kim M, Fisher DT, Powers C, Attwood K, Bagaria SP, Knutson KL, Skitzki JJ. Dynamic control of tumor vasculature improves antitumor responses in a regional model of melanoma. Sci Rep 2020; 10:13245. [PMID: 32764623 PMCID: PMC7413248 DOI: 10.1038/s41598-020-70233-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/24/2020] [Indexed: 11/09/2022] Open
Abstract
Despite advances in therapy for melanoma, heterogeneous responses with limited durability represent a major gap in treatment outcomes. The purpose of this study was to determine whether alteration in tumor blood flow could augment drug delivery and improve antitumor responses in a regional model of melanoma. This approach to altering tumor blood flow was termed "dynamic control." Dynamic control of tumor vessels in C57BL/6 mice bearing B16 melanoma was performed using volume expansion (saline bolus) followed by phenylephrine. Intravital microscopy (IVM) was used to observe changes directly in real time. Our approach restored blood flow in non-functional tumor vessels. It also resulted in increased chemotherapy (melphalan) activity, as measured by formation of DNA adducts. The combination of dynamic control and melphalan resulted in superior outcomes compared to melphalan alone (median time to event 40.0 vs 25.0 days, respectively, p = 0.041). Moreover, 25% (3/12) of the mice treated with the combination approach showed complete tumor response. Importantly, dynamic control plus melphalan did not result in increased adverse events. In summary, we showed that dynamic control was feasible, directly observable, and augmented antitumor responses in a regional model of melanoma. Early clinical trials to determine the translational feasibility of dynamic control are ongoing.
Collapse
Affiliation(s)
- Emmanuel M Gabriel
- Department of Surgery, Section of Surgical Oncology, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Minhyung Kim
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Daniel T Fisher
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Colin Powers
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher Attwood
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sanjay P Bagaria
- Department of Surgery, Section of Surgical Oncology, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
| | - Joseph J Skitzki
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
6
|
Jiang P, Gao W, Ma T, Wang R, Piao Y, Dong X, Wang P, Zhang X, Liu Y, Su W, Xiang R, Zhang J, Li N. CD137 promotes bone metastasis of breast cancer by enhancing the migration and osteoclast differentiation of monocytes/macrophages. Theranostics 2019; 9:2950-2966. [PMID: 31244935 PMCID: PMC6568184 DOI: 10.7150/thno.29617] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/25/2019] [Indexed: 12/13/2022] Open
Abstract
Rationale: Bone is one of the most common metastatic sites of breast cancer. CD137 (4-1BB), a member of the tumor necrosis factor (TNF) receptor superfamily, is mainly expressed in activated leukocytes. Previous study demonstrates the effect of CD137-CD137L bidirectional signaling pathway on RANKL-mediated osteoclastogenesis. However, the role of CD137 in bone metastasis of breast cancer needs further study. Methods: Stable monocyte/macrophage cell lines with Cd137 overexpression and silencing were established. Western blot, real-time PCR, transwell and tartrate-resistant acid phosphatase staining were used to detect the regulatory effect of CD137 on migration and osteoclastogenesis of monocytes/macrophages in vitro. Spontaneous bone metastasis mouse model was established, bioluminescent images, immunohistochemistry and histology assay were performed to detect the function of CD137 in bone metastasis in vivo. Results: We found that CD137 promotes the migration of monocytes/macrophages to tumor microenvironment by upregulating the expression of Fra1. It also promoted the differentiation of monocytes/macrophages into osteoclasts at the same time, thus providing a favorable microenvironment for the colonization and growth of breast cancer cells in bone. Based on these findings, a novel F4/80-targeted liposomal nanoparticle encapsulating the anti-CD137 blocking antibody (NP-αCD137 Ab-F4/80) was synthesized. This nanoparticle could inhibit both bone and lung metastases of 4T1 breast cancer cells with high efficacy in vivo. In addition, it increased the therapeutic efficacy of Fra1 inhibitor on tumor metastasis. Conclusions: Taken together, these findings reveal the promotion effect of macrophage/monocyte CD137 on bone metastases and provide a promising therapeutic strategy for metastasis of breast cancer.
Collapse
|
7
|
Rationally designed peptide nanosponges for cell-based cancer therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2555-2564. [DOI: 10.1016/j.nano.2017.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/07/2017] [Accepted: 07/12/2017] [Indexed: 11/20/2022]
|
8
|
Guo C, Chen Y, Gao W, Chang A, Ye Y, Shen W, Luo Y, Yang S, Sun P, Xiang R, Li N. Liposomal Nanoparticles Carrying anti-IL6R Antibody to the Tumour Microenvironment Inhibit Metastasis in Two Molecular Subtypes of Breast Cancer Mouse Models. Am J Cancer Res 2017; 7:775-788. [PMID: 28255366 PMCID: PMC5327649 DOI: 10.7150/thno.17237] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/04/2016] [Indexed: 12/17/2022] Open
Abstract
Tumour microenvironment (TME) contributes significantly towards potentiating the stemness and metastasis properties of cancer cells. IL6-Stat3 is one of the important cell signaling pathways in mediating the communication between tumour and immune cells. Here, we have systematically developed a novel anti-CD44 antibody-mediated liposomal nanoparticle delivery system loaded with anti-IL6R antibody, which could specifically target the TME of CD44+ breast cancer cells in different mouse models for triple negative and luminal breast cancer. This nanoparticle had an enhanced and specific tumour targeting efficacy with dramatic anti-tumour metastasis effects in syngeneic BALB/c mice bearing 4T1 cells as was in the syngeneic MMTV-PyMT mice. It inhibited IL6R-Stat3 signaling and moderated the TME, characterized by the reduced expression of genes encoding Stat3, Sox2, VEGFA, MMP-9 and CD206 in the breast tissues. Furthermore, this nanoparticle reduced the subgroups of Sox2+ and CD206+ cells in the lung metastatic foci, demonstrating its inhibitory effect on the lung metastatic niche for breast cancer stem cells. Taken together, the CD44 targeted liposomal nanoparticles encapsulating anti-IL6R antibody achieved a significant effect to inhibit the metastasis of breast cancer in different molecular subtypes of breast cancer mouse models. Our results shed light on the application of nanoparticle mediated cancer immune-therapy through targeting TME.
Collapse
|
9
|
Merkle HP. Drug delivery's quest for polymers: Where are the frontiers? Eur J Pharm Biopharm 2016; 97:293-303. [PMID: 26614554 DOI: 10.1016/j.ejpb.2015.04.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/03/2015] [Accepted: 04/22/2015] [Indexed: 12/20/2022]
Abstract
Since the legendary 1964 article of Folkman and Long entitled "The use of silicone rubber as a carrier for prolonged drug therapy" the role of polymers in controlled drug delivery has come a long way. Today it is evident that polymers play a crucial if not the prime role in this field. The latest boost owes to the interest in drug delivery for the purpose of tissue engineering in regenerative medicine. The focus of this commentary is on a selection of general and personal observations that are characteristic for the current state of polymer therapeutics and carriers. It briefly highlights selected examples for the long march of synthetic polymer-drug conjugates from bench to bedside, comments on the ambivalence of selected polymers as inert excipients versus biological response modifiers, and on the yet unsolved dilemma of cationic polymers for the delivery of nucleic acid therapeutics. Further subjects are the complex design of multifunctional polymeric carriers including recent concepts towards functional supramolecular polymers, as well as observations on stimuli-sensitive polymers and the currently ongoing trend towards natural and naturally-derived biopolymers. The final topic is the discovery and early development of a novel type of biodegradable polyesters for parenteral use. Altogether, it is not the basic and applied research in polymer therapeutics and carriers, but the translational process that is the key hurdle to proceed towards an authoritative approval of new polymer therapeutics and carriers.
Collapse
Affiliation(s)
- Hans P Merkle
- Institute of Pharmaceutical Sciences, ETH Zurich, Campus Hönggerberg, Vladimir-Prelog-Weg 1-5/10, CH-8093 Zürich, Switzerland.
| |
Collapse
|
10
|
Sarko D. Kidney-Specific Drug Delivery: Review of Opportunities, Achievements, and Challenges. ACTA ACUST UNITED AC 2016. [DOI: 10.15406/japlr.2016.02.00033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
11
|
Dubey RD, Saneja A, Qayum A, Singh A, Mahajan G, Chashoo G, Kumar A, Andotra SS, Singh SK, Singh G, Koul S, Mondhe DM, Gupta PN. PLGA nanoparticles augmented the anticancer potential of pentacyclic triterpenediol in vivo in mice. RSC Adv 2016. [DOI: 10.1039/c6ra14929d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A novel pentacyclic triterpenediol (TPD), an anticancer lead fromBoswellia serrata, was encapsulated into PLGA nanoparticles, leading to enhancement in anticancer potential in EAT bearing mice model.
Collapse
|
12
|
Wibroe PP, Ahmadvand D, Oghabian MA, Yaghmur A, Moghimi SM. An integrated assessment of morphology, size, and complement activation of the PEGylated liposomal doxorubicin products Doxil®, Caelyx®, DOXOrubicin, and SinaDoxosome. J Control Release 2015; 221:1-8. [PMID: 26608877 DOI: 10.1016/j.jconrel.2015.11.021] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 11/12/2015] [Accepted: 11/18/2015] [Indexed: 11/15/2022]
Abstract
In order to improve patient's benefit and safety, comprehensive regulatory guidelines on specificities of Non-Biological Complex Drugs (NBCDs), such as doxorubicin-encapsulated liposomes, and their follow-on versions are needed. Here, we compare Doxil® and its European analog Caelyx® with the two follow-on products DOXOrubicin (approved by the US Food and Drug Administration) and SinaDoxosome (produced in Iran) by cryogenic transmission electron microscopy, dynamic light scattering and Nanoparticle Tracking Analysis, and assess their potential in activating the complement system in human sera. We found subtle physicochemical differences between the tested liposomal products and even between the tested batches of Doxil® and Caelyx®. Notably, these included differences in vesicular population aspect ratios and particle number. Among the tested products, only SinaDoxosome, in addition to the presence of unilamellar vesicles with entrapped doxorubicin crystals, contained empty circular disks. Differences were also found in complement responses, which may be related to some morphological differences. This study has demonstrated an integrated biophysical and immunological toolbox for improved analysis and detection of physical differences among vesicular populations that may modulate their clinical performance. Combined, these approaches may help better product selection for infusion to the patients as well as for improved design and characterization of future vesicular NBCDs with enhanced clinical performance and safety.
Collapse
Affiliation(s)
- Peter P Wibroe
- Nanomedicine Laboratory, Centre for Pharmaceutical Nanotechnology and Nanotoxicology, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Davoud Ahmadvand
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oghabian
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Anan Yaghmur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - S Moein Moghimi
- Nanomedicine Laboratory, Centre for Pharmaceutical Nanotechnology and Nanotoxicology, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark; NanoScience Centre, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen Ø, Denmark.
| |
Collapse
|
13
|
Glück S. nab-Paclitaxel for the Treatment of Aggressive Metastatic Breast Cancer. Clin Breast Cancer 2014; 14:221-7. [DOI: 10.1016/j.clbc.2014.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 10/25/2022]
|
14
|
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: 61] [Impact Index Per Article: 6.1] [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
|
15
|
Cirauqui BC, García VQ, Rubio CL, Miguel MIP, Riera LC, Aranda NP, Martín SV, Martínez AM, Caruncho CR, Vila MM. Nanoparticle albumin-bound paclitaxel in a patient with locally advanced breast cancer and taxane-induced skin toxicity: a case report. J Med Case Rep 2014; 8:6. [PMID: 24386978 PMCID: PMC3917539 DOI: 10.1186/1752-1947-8-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/19/2013] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Taxanes have demonstrated effectiveness in the treatment of breast cancer, the most common type of cancer in women. The toxicity profile of taxanes (including skin toxicities) induces dose adjustment, delay, or discontinuation, which prevents a sufficient dose intensity to achieve a response. Nanoparticle albumin-bound paclitaxel, a solvent-free form of paclitaxel, prevents toxicities and reduces the pharmacokinetic interferences between paclitaxel and other drugs. CASE PRESENTATION We describe the case of a 55-year-old Caucasian woman with locally advanced breast cancer treated with neoadjuvant therapy who developed secondary skin toxicity due to delayed hypersensitivity to taxanes. She received Adriamycin® (doxorubicin), cyclophosphamide and docetaxel and developed toxicity that promoted treatment delay and a switch to weekly paclitaxel. After the third and fourth weeks of treatment, paclitaxel toxicities also induced treatment delay and paclitaxel was switched to nanoparticle albumin-bound paclitaxel. She completed the five planned nanoparticle albumin-bound paclitaxel cycles with acceptable tolerability (including persistent grade 2 neuropathy) and without dose delay or adjustments. Clinical response was achieved although pathological response was not good. CONCLUSIONS Nanoparticle albumin-bound paclitaxel treatment is a good option for patients with breast cancer with taxanes-related skin toxicity. This drug allows the treatment to be completed with acceptable tolerance in our case.
Collapse
Affiliation(s)
- Beatriz Cirauqui Cirauqui
- Department of Medical Oncology, Catalonian Institute of Oncology, Germans Trias i Pujol Hospital, Badalona, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Dayani Y, Malmstadt N. Liposomes with double-stranded DNA anchoring the bilayer to a hydrogel core. Biomacromolecules 2013; 14:3380-5. [PMID: 24083513 PMCID: PMC3874235 DOI: 10.1021/bm401155a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Liposomes are important biomolecular nanostructures for handling membrane-associated molecules in the lab and delivering drugs in the clinic. In addition to their biomedical applications, they have been widely used as model cell membranes in biophysical studies. Here we present a liposome-based model membrane that mimics the attachment of membrane-resident molecules to the cytoskeleton. To facilitate this attachment, we have developed a lipid-based hybrid nanostructure in which the liposome bilayer membrane is covalently anchored to a biocompatible poly(ethylene) glycol (PEG) hydrogel core using short double-stranded DNA (dsDNA) linkers. The dsDNA linkers connect cholesterol groups that reside in the bilayer to vinyl groups that are incorporated in the cross-linked hydrogel backbone. Size exclusion chromatography (SEC) of intact and surfactant-treated nanoparticles confirms the formation of anchored hydrogel structures. Transmission electron microscopy (TEM) shows ~100 nm nanoparticles even after removal of unanchored phospholipids. The location of dsDNA groups at the hydrogel-bilayer interface is confirmed with a fluorescence assay. Using DNA as a linker between the bilayer and a hydrogel core allows for temperature-dependent release of the anchoring interaction, produces polymer nanogels with addressible hybridization sites on their surface, and provides a prototype structure for potential future oligonucleotide drug delivery applications.
Collapse
Affiliation(s)
- Yasaman Dayani
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Noah Malmstadt
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
17
|
Golla K, Cherukuvada B, Ahmed F, Kondapi AK. Efficacy, safety and anticancer activity of protein nanoparticle-based delivery of doxorubicin through intravenous administration in rats. PLoS One 2012; 7:e51960. [PMID: 23284832 PMCID: PMC3528733 DOI: 10.1371/journal.pone.0051960] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 11/09/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIMS Doxorubicin is a potent anticancer drug and a major limiting factor that hinders therapeutic use as its high levels of systemic circulation often associated with various off-target effects, particularly cardiotoxicity. The present study focuses on evaluation of the efficacy of doxorubicin when it is loaded into the protein nanoparticles and delivered intravenously in rats bearing Hepatocellular carcinoma (HCC). The proteins selected as carrier were Apotransferrin and Lactoferrin, since the receptors for these two proteins are known to be over expressed on cancer cells due to their iron transport capacity. METHODS Doxorubicin loaded apotransferrin (Apodoxonano) and lactoferrin nanoparticles (Lactodoxonano) were prepared by sol-oil chemistry. HCC in the rats was induced by 100 mg/l of diethylnitrosamine (DENA) in drinking water for 8 weeks. Rats received 5 doses of 2 mg/kg drug equivalent nanoparticles through intravenous administration. Pharmacokinetics and toxicity of nanoformulations was evaluated in healthy rats and anticancer activity was studied in DENA treated rats. The anticancer activity was evaluated through counting of the liver nodules, H & E analysis and by estimating the expression levels of angiogenic and antitumor markers. RESULTS In rats treated with nanoformulations, the numbers of liver nodules were found to be significantly reduced. They showed highest drug accumulation in liver (22.4 and 19.5 µg/g). Both nanoformulations showed higher localization compared to doxorubicin (Doxo) when delivered in the absence of a carrier. Higher amounts of Doxo (195 µg/g) were removed through kidney, while Apodoxonano and Lactodoxonano showed only a minimal amount of removal (<40 µg/g), suggesting the extended bioavailability of Doxo when delivered through nanoformulation. Safety analysis shows minimal cardiotoxicity due to lower drug accumulation in heart in the case of nanoformulation. CONCLUSION Drug delivery through nanoformulations not only minimizes the cardiotoxicity of doxorubicin but also enhances the efficacy and bioavailability of the drug in a target-specific manner.
Collapse
Affiliation(s)
- Kishore Golla
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
- Centre for Nanotechnology, University of Hyderabad, Hyderabad, India
| | | | - Farhan Ahmed
- Department of Biotechnology, University of Hyderabad, Hyderabad, India
| | - Anand K. Kondapi
- Department of Biotechnology, University of Hyderabad, Hyderabad, India
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
- Centre for Nanotechnology, University of Hyderabad, Hyderabad, India
| |
Collapse
|
18
|
Abstract
Amphiphilic polymers represented by block copolymers self-assemble into well-defined nanostructures capable of incorporating therapeutics. Polymer nanoassemblies currently developed for cancer treatment and imaging are reviewed in this article. Particular attention is paid to three representative polymer nanoassemblies: polymer micelles, polymer micellar aggregates and polymer vesicles. Rationales, design and performance of these polymer nanoassemblies are addressed, focusing on increasing the solubility and chemical stability of drugs. Also discussed are polymer nanoassembly formation, the distribution of polymer materials in the human body and applications of polymer nanoassemblies for combined therapy and imaging of cancer. Updates on tumor-targeting approaches, based on preclinical and clinical results are provided, as well as solutions for current issues that drug-delivery systems have, such as in vivo stability, tissue penetration and therapeutic efficacy. These are discussed to provide insights on the future development of more effective polymer nanoassemblies for the delivery of therapeutics in the body.
Collapse
|
19
|
Jackisch C, Lück HJ, Untch M, Bischoff J, Müller V, Schmidt M, Thill M, Kiechle M. Weekly nab-Paclitaxel in Metastatic Breast Cancer - Summary and Results of an Expert Panel Discussion. ACTA ACUST UNITED AC 2012; 7:137-143. [PMID: 22740801 DOI: 10.1159/000338273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Taxanes are regarded as the most effective single agents in the treatment of metastatic breast cancer (MBC). For conventional taxanes, crucial toxicities and impairments in clinical efficacy are related to solvents necessary because of the agents' hydrophobicity. The mandatory premedication with corticosteroids causes additional side effects. Nab-paclitaxel is a solvent-free colloidal suspension of paclitaxel and human serum albumin that exploits the physiological transport properties of albumin. It is registered as monotherapy with a recommended dose of 260 mg/m(2) every 3 weeks for the treatment of patients with MBC, who have failed a first-line treatment of metastatic disease and for whom a standard anthracycline treatment is not indicated. Clinical evidence is available for the registered 3-weekly administration and for alternative weekly schedules in first and further lines of therapy of patients with MBC. During an advisory board meeting, a group of 8 German breast cancer experts reviewed the clinical data of nab-paclitaxel in MBC and discussed how nab-paclitaxel could be used in clinical practice on the basis of the current data.
Collapse
|
20
|
"Targeting" nanoparticles: the constraints of physical laws and physical barriers. J Control Release 2012; 164:115-24. [PMID: 22484196 DOI: 10.1016/j.jconrel.2012.03.022] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 03/10/2012] [Accepted: 03/13/2012] [Indexed: 11/20/2022]
Abstract
In comparison to the complexities of the body, its organs, its normal and aberrant cells, many nanoparticles will appear to be relatively simple objects. This view is deceptive because the physicochemical properties of nanosystems, although quite well understood on the basis of material science, surface science and colloid theory, are far from simple in practice. While their properties are largely controllable in vitro, often purportedly "designed", their administration by any route changing environments conspires to produce additional layers of complexity. Some of the key physical laws and physicochemical parameters governing the fate of nanoparticles on their journey from point of intravenous administration to desired destinations such as tumors are discussed. Much of the science relevant to nanocarrier based targeting has been elaborated in studying purely physical phenomena, but there can be found therein many analogies with biological systems. These include factors that impede quantitative targeting: diffusion in complex media, aggregation and flocculation, hindered behavior of particles in confined spaces, jamming and dispersion in flow. All of these have the ability to influence fate and destination. Most of the critical processes are particle size dependent but not always linearly so. Virtually all processes in vivo involve an element of probability. Particle size and properties can be controlled to a large extent, but stochastic processes cannot by definition. Progress has been made, but the quantitative delivery of a nanocarrier to defined sites in tumors is neither inevitable nor yet predictable.
Collapse
|
21
|
Kaminskas LM, McLeod VM, Porter CJH, Boyd BJ. Association of chemotherapeutic drugs with dendrimer nanocarriers: an assessment of the merits of covalent conjugation compared to noncovalent encapsulation. Mol Pharm 2012; 9:355-73. [PMID: 22250750 DOI: 10.1021/mp2005966] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cancer is a leading cause of death within developed nations, and part of this morbidity is due to difficulties associated with its treatment. Currently, anticancer therapy relies heavily upon the administration of small molecule cytotoxic drugs that attack both cancerous and noncancerous cells due to limited selectivity of the drugs and widespread distribution of the cytotoxic molecules throughout the body. The antitumor efficacy and systemic toxicity of existing chemotherapeutic drugs can, however, be improved by employing formulation and particle engineering approaches. Thus, drug delivery systems can be developed that more specifically target tumor tissue using both passive (such as the enhanced permeation and retention effect) and active (through the use of cancer targeting ligands) modalities. Dendrimers are one such system that can be developed with high structural monodispersity, long plasma circulation times and precise control over surface structure and biodistribution properties. Chemotherapeutic drugs can be associated with dendrimers via covalent conjugation to the surface, or via encapsulation of drugs within the structure. Each of these approaches has demonstrated therapeutic benefit relative to the administration of free drug. Thus far, however, there has not been a systematic review toward which drug association approach will provide the best outcomes in terms of antitumor efficacy and systemic toxicity. Hence, the current literature is reviewed here and recommendations are proposed as to the suggested approach to develop dendrimers as tumor targeted drug-delivery vectors.
Collapse
Affiliation(s)
- Lisa M Kaminskas
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
22
|
Moghimi SM, Hunter AC, Andresen TL. Factors controlling nanoparticle pharmacokinetics: an integrated analysis and perspective. Annu Rev Pharmacol Toxicol 2011; 52:481-503. [PMID: 22035254 DOI: 10.1146/annurev-pharmtox-010611-134623] [Citation(s) in RCA: 371] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intravenously injected nanoparticulate drug carriers provide a wide range of unique opportunities for site-specific targeting of therapeutic agents to many areas within the vasculature and beyond. Pharmacokinetics and biodistribution of these carriers are controlled by a complex array of interrelated core and interfacial physicochemical and biological factors. Pertinent to realizing therapeutic goals, definitive maps that establish the interdependency of nanoparticle size, shape, and surface characteristics in relation to interfacial forces, biodistribution, controlled drug release, excretion, and adverse effects must be outlined. These concepts are critically evaluated and an integrated perspective is provided on the basis of the recent application of nanoscience approaches to nanocarrier design and engineering. The future of this exciting field is bright; some regulatory-approved products are already on the market and many are in late-phase clinical trials. With concomitant advances in extensive computational knowledge of the genomics and epigenomics of interindividual variations in drug responses, the boundaries toward development of personalized nanomedicines can be pushed further.
Collapse
Affiliation(s)
- S M Moghimi
- Centre for Pharmaceutical Nanotechnology and Nanotoxicology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark.
| | | | | |
Collapse
|
23
|
Merisko-Liversidge E, Liversidge GG. Nanosizing for oral and parenteral drug delivery: a perspective on formulating poorly-water soluble compounds using wet media milling technology. Adv Drug Deliv Rev 2011; 63:427-40. [PMID: 21223990 DOI: 10.1016/j.addr.2010.12.007] [Citation(s) in RCA: 383] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 12/29/2010] [Indexed: 11/29/2022]
Abstract
A significant percentage of active pharmaceutical ingredients identified through discovery screening programs is poorly soluble in water. These molecules are often difficult to formulate using conventional approaches and are associated with innumerable formulation-related performance issues, e.g. poor bioavailability, lack of dose proportionality, slow onset of action and other attributes leading to poor patient compliance. In addition, for parenteral products, these molecules are generally administered with co-solvents and thus have many undesirable side effects. Wet media milling is one of the leading particle size reduction approaches that have been successfully used to formulate these problematic compounds. The approach is a water-based media milling process where micron-sized drug particles are shear-fractured into nanometer-sized particles. Nanoparticle dispersions are stable and typically have a mean diameter of less than 200 nm with 90% of the particles being less than 400 nm. The formulation consists only of water, drug and one or more GRAS excipients. Drug concentrations approaching 300-400mg/g can be targeted with the use of minimal amounts stabilizer. Typically, on average, the drug to stabilizer ratio on a weight basis ranges from 2:1 to 20:1. These liquid nanodispersions exhibit acceptable shelf-life and can be post-processed into various types of solid dosage forms. Nanoparticulate-based drug products have been shown to improve bioavailability and enhance drug exposure for oral and parenteral dosage forms. Suitable formulations for the most commonly used routes of administration can be identified with milligram quantities of drug substance providing the discovery scientist an alternate avenue for screening and identifying superior leads. In the last few years, formulating poorly water soluble compounds as nanosuspensions has evolved from a conception to a realization. The versatility and applicability of this drug delivery platform are just beginning to be realized.
Collapse
|
24
|
Basel MT, Shrestha TB, Troyer DL, Bossmann SH. Protease-sensitive, polymer-caged liposomes: a method for making highly targeted liposomes using triggered release. ACS NANO 2011; 5:2162-75. [PMID: 21314184 DOI: 10.1021/nn103362n] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Liposomes have become useful and well-known drug delivery vehicles because of their ability to entrap drugs without chemically modifying them and to deliver them somewhat selectively to tumorous tissue via the enhanced permeation and retention (EPR) effect. Although useful, liposome preparations are still less than ideal because of imperfect specificity, slow release kinetics in the tumor, and leakiness prior to reaching the tumor site. Cancer-associated proteases (CAPs), which are differentially expressed in tumors, have also gained traction recently as a method for tumor targeting and drug delivery. By combining the EPR effect with CAPs sensitivity, a much more specific liposome can be produced. The method described here creates an improved liposome system that can target more specifically, with faster release kinetics and lower general leaking, by deliberately producing a very unstable liposome (loaded with hyperosmotic vehicle) that is subsequently stabilized by a cross-linked polymer shell containing consensus sequences for cancer-associated proteases (protease-triggered, caged liposomes). A cholesterol-anchored, graft copolymer, composed of a short peptide sequence for urokinase plasminogen activator (uPA) and poly(acrylic acid), was synthesized and incorporated into liposomes prepared at high osmolarities. Upon cross-linking of the polymers, the protease-triggered, caged liposomes showed significant resistance to osmotic swelling and leaking of contents. Protease-triggered, caged liposomes also showed significant and substantial differential release of contents in the presence of uPA, while bare liposomes showed no differential effect in the presence of uPA. Thus a protease-sensitive liposome system with fast release kinetics was developed that could be used for more specific targeting to tumors.
Collapse
Affiliation(s)
- Matthew T Basel
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, KS 66506, USA.
| | | | | | | |
Collapse
|
25
|
Drug delivery systems using polymer nanoassemblies for cancer treatment. Ther Deliv 2010; 1:361-3. [DOI: 10.4155/tde.10.28] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
26
|
Resorcinolic lipids improve the properties of sphingomyelin–cholesterol liposomes. Chem Phys Lipids 2010; 163:648-54. [DOI: 10.1016/j.chemphyslip.2010.05.202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 05/07/2010] [Accepted: 05/21/2010] [Indexed: 11/17/2022]
|
27
|
Krishna ADS, Mandraju RK, Kishore G, Kondapi AK. An efficient targeted drug delivery through apotransferrin loaded nanoparticles. PLoS One 2009; 4:e7240. [PMID: 19806207 PMCID: PMC2752169 DOI: 10.1371/journal.pone.0007240] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 08/31/2009] [Indexed: 02/07/2023] Open
Abstract
Background Cancerous state is a highly stimulated environment of metabolically active cells. The cells under these conditions over express selective receptors for assimilation of factors essential for growth and transformation. Such receptors would serve as potential targets for the specific ligand mediated transport of pharmaceutically active molecules. The present study demonstrates the specificity and efficacy of protein nanoparticle of apotransferrin for targeted delivery of doxorubicin. Methodology/Principal Findings Apotransferrin nanoparticles were developed by sol-oil chemistry. A comparative analysis of efficiency of drug delivery in conjugated and non-conjugated forms of doxorubicin to apotransferrin nanoparticle is presented. The spherical shaped apotransferrin nanoparticles (nano) have diameters of 25–50 ηm, which increase to 60–80 ηm upon direct loading of drug (direct-nano), and showed further increase in dimension (75–95 ηm) in conjugated nanoparticles (conj-nano). The competitive experiments with the transferrin receptor specific antibody showed the entry of both conj-nano and direct-nano into the cells through transferrin receptor mediated endocytosis. Results of various studies conducted clearly establish the superiority of the direct-nano over conj-nano viz. (a) localization studies showed complete release of drug very early, even as early as 30 min after treatment, with the drug localizing in the target organelle (nucleus) (b) pharmacokinetic studies showed enhanced drug concentrations, in circulation with sustainable half-life (c) the studies also demonstrated efficient drug delivery, and an enhanced inhibition of proliferation in cancer cells. Tissue distribution analysis showed intravenous administration of direct nano lead to higher drug localization in liver, and blood as compared to relatively lesser localization in heart, kidney and spleen. Experiments using rat cancer model confirmed the efficacy of the formulation in regression of hepatocellular carcinoma with negligible toxicity to kidney and liver. Conclusions The present study thus demonstrates that the direct-nano is highly efficacious in delivery of drug in a target specific manner with lower toxicity to heart, liver and kidney.
Collapse
Affiliation(s)
| | | | - Golla Kishore
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
- Centre for Nanotechnology, University of Hyderabad, Hyderabad, India
| | - Anand Kumar Kondapi
- Department of Biochemistry, University of Hyderabad, Hyderabad, India
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, India
- Centre for Nanotechnology, University of Hyderabad, Hyderabad, India
- * E-mail:
| |
Collapse
|
28
|
Moghimi S, Andresen T. Complement-mediated tumour growth: Implications for cancer nanotechnology and nanomedicines. Mol Immunol 2009; 46:1571-2. [DOI: 10.1016/j.molimm.2009.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 02/13/2009] [Accepted: 02/14/2009] [Indexed: 11/24/2022]
|
29
|
Hamad I, Christy Hunter A, Rutt KJ, Liu Z, Dai H, Moein Moghimi S. Complement activation by PEGylated single-walled carbon nanotubes is independent of C1q and alternative pathway turnover. Mol Immunol 2008; 45:3797-803. [PMID: 18602161 DOI: 10.1016/j.molimm.2008.05.020] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 05/23/2008] [Accepted: 05/25/2008] [Indexed: 11/28/2022]
Abstract
We have investigated the interaction between long circulating poly(ethylene glycol)-stabilized single-walled carbon nanotubes (SWNTs) and the complement system. Aminopoly(ethylene glycol)(5000)-distearoylphosphatidylethanolamine (aminoPEG(5000)-DSPE) and methoxyPEG(5000)-DSPE coated as-grown HIPco SWNTs activated complement in undiluted normal human serum as reflected in significant rises in C4d and SC5b-9 levels, but not the alternative pathway split-product Bb, thus indicating activation exclusively through C4 cleavage. Studies in C2-depleted serum confirmed that PEGylated nanotube-mediated elevation of SC5b-9 was C4b2a convertase-dependent. With the aid of monoclonal antibodies against C1s and human serum depleted from C1q, nanotube-mediated complement activation in C1q-depleted serum was also shown to be independent of classical pathway. Nanotube-mediated C4d elevation in C1q-depleted serum, however, was inhibited by N-acetylglucosamine, Futhan (a broad-spectrum serine protease inhibitor capable of preventing complement activation through all three pathways) and anti-MASP-2 antibodies; this strongly suggests a role for activation of MASP-2 in subsequent C4 cleavage and assembly of C4b2a covertases. Intravenous injection of PEGylated nanotubes in some rats was associated with a significant rise in plasma thromboxane B2 levels, indicative of in vivo nanotube-mediated complement activation. The clinical implications of these observations are discussed.
Collapse
Affiliation(s)
- Islam Hamad
- Molecular Targeting and Polymer Toxicology Group, School of Pharmacy, University of Brighton, Brighton BN2 4GJ, UK
| | | | | | | | | | | |
Collapse
|