51
|
Merino M, Zalba S, Garrido MJ. Immunoliposomes in clinical oncology: State of the art and future perspectives. J Control Release 2018; 275:162-176. [DOI: 10.1016/j.jconrel.2018.02.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 02/02/2023]
|
52
|
Matsumura Y. Development of CAST therapy based on the EPR effect: lesson from clinical trials. ACTA ACUST UNITED AC 2018. [DOI: 10.2745/dds.33.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Yasuhiro Matsumura
- Division of Developmental Therapeutics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center
| |
Collapse
|
53
|
Therapeutic prospects of microRNAs in cancer treatment through nanotechnology. Drug Deliv Transl Res 2017; 8:97-110. [DOI: 10.1007/s13346-017-0440-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
54
|
Repetto O, De Re V. Coagulation and fibrinolysis in gastric cancer. Ann N Y Acad Sci 2017; 1404:27-48. [PMID: 28833193 DOI: 10.1111/nyas.13454] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/12/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022]
Abstract
Coagulation is a highly conserved process occurring after an injury to a blood vessel and resulting in hemostasis. In the thrombus microenvironment, finely orchestrated events restore vessel integrity through platelet activation, adhesion, and aggregation (primary hemostasis), followed by the coagulation cascades, thrombin generation, and fibrin clot deposition (secondary hemostasis). Several studies on cancer have provided insight into dramatic changes to coagulation-related events (i.e., fibrin clot deposition, fibrinolysis) during tumor pathogenesis, progression, and metastasis, in addition to a tumor-driven systemic activation of hemostasis and thrombosis (Trousseau's syndrome). Diverse molecular and cellular effectors participate in the cross talk between hemostasis and tumors. Here, we focus on some aspects of the interconnection between cancer biology and hemostatic components, with particular attention to some key coagulation-related proteins (e.g., tissue factor, thrombin, fibrinogen, and D-dimers) in the particular case of gastric cancer (GC). Recent advances in deciphering the complex molecular link between GC and the coagulation system are described, showing their important roles in better management of patients affected by GC.
Collapse
Affiliation(s)
- Ombretta Repetto
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, CRO Aviano National Cancer Institute, Aviano (PN), Italy
| | - Valli De Re
- Facility of Bio-Proteomics, Immunopathology and Cancer Biomarkers, CRO Aviano National Cancer Institute, Aviano (PN), Italy
| |
Collapse
|
55
|
Mu LM, Ju RJ, Liu R, Bu YZ, Zhang JY, Li XQ, Zeng F, Lu WL. Dual-functional drug liposomes in treatment of resistant cancers. Adv Drug Deliv Rev 2017; 115:46-56. [PMID: 28433739 DOI: 10.1016/j.addr.2017.04.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/26/2022]
Abstract
Efficacy of regular chemotherapy is significantly hampered by multidrug resistance (MDR) and severe systemic toxicity. The reduced toxicity has been evidenced after administration of drug liposomes, consisting of the first generation of regular drug liposomes, the second generation of long-circulation drug liposomes, and the third generation of targeting drug liposomes. However, MDR of cancers remains as an unsolved issue. The objective of this article is to review the dual-functional drug liposomes, which demonstrate the potential in overcoming MDR. Herein, dual-functional drug liposomes are referring to the drug-containing phospholipid bilayer vesicles that possess a dual-function of providing the basic efficacy of drug and the extended effect of the drug carrier. They exhibit unique roles in treatment of resistant cancer via circumventing drug efflux caused by adenosine triphosphate binding cassette (ABC) transporters, eliminating cancer stem cells, destroying mitochondria, initiating apoptosis, regulating autophagy, destroying supply channels, utilizing microenvironment, and silencing genes of the resistant cancer. As the prospect of an estimation, dual-functional drug liposomes would exhibit more strength in their extended function, hence deserving further investigation for clinical validation.
Collapse
|
56
|
Abshire C, Murad HY, Arora JS, Liu J, Mandava SH, John VT, Khismatullin DB, Lee BR. Focused Ultrasound-Triggered Release of Tyrosine Kinase Inhibitor From Thermosensitive Liposomes for Treatment of Renal Cell Carcinoma. J Pharm Sci 2017; 106:1355-1362. [PMID: 28159640 DOI: 10.1016/j.xphs.2017.01.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 12/16/2016] [Accepted: 01/24/2017] [Indexed: 12/29/2022]
Abstract
This study reports, for the first time, development of tyrosine kinase inhibitor-loaded, thermosensitive liposomes (TKI/TSLs) and their efficacy for treatment of renal cell carcinoma when triggered by focused ultrasound (FUS). Uptake of these nanoparticles into renal cancer cells was visualized with confocal and fluorescent imaging of rhodamine B-loaded liposomes. The combination of TKI/TSLs and FUS was tested in an in vitro tumor model of renal cell carcinoma. According to MTT cytotoxic assay and flow cytometric analysis, the combined treatment led to the least viability (23.4% ± 2.49%, p < 0.001), significantly lower than that observed from treatment with FUS (97.6% ± 0.67%, not significant) or TKI/TSL (71.0% ± 3.65%, p < 0.001) at 96 h compared to control. The importance of this unique, synergistic combination was demonstrated in viability experiments with non-thermosensitive liposomes (TKI/NTSL + FUS: 58.8% ± 1.5% vs. TKI/TSL + FUS: 36.2% ± 1.4%, p < 0.001) and heated water immersion (TKI/TSL + WB43°: 59.3% ± 2.91% vs. TKI/TSL + FUS: 36.4% ± 1.55%, p < 0.001). Our findings coupled with the existing use of FUS in clinical practice make the proposed combination of targeted chemotherapy, nanotechnology, and FUS a promising platform for enhanced drug delivery and cancer treatment.
Collapse
Affiliation(s)
- Caleb Abshire
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Hakm Y Murad
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana 70118; Tulane Institute for Integrative Engineering for Health and Medicine, Tulane University, New Orleans, Louisiana 70112
| | - Jaspreet S Arora
- Department of Chemical and Bimolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana 70118; Vector-Borne Infectious Disease Research Center, Tulane University, New Orleans, Louisiana 70112
| | - James Liu
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Sree Harsha Mandava
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Vijay T John
- Department of Chemical and Bimolecular Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana 70118; Vector-Borne Infectious Disease Research Center, Tulane University, New Orleans, Louisiana 70112
| | - Damir B Khismatullin
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, Louisiana 70118; Tulane Institute for Integrative Engineering for Health and Medicine, Tulane University, New Orleans, Louisiana 70112; Division of Urology, University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Benjamin R Lee
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana 70112; Division of Urology, University of Arizona College of Medicine, Tucson, Arizona 85724.
| |
Collapse
|
57
|
Abstract
Targeted cancer nanotherapeutics offers numerous opportunities for the selective uptake of toxic chemotherapies within tumors and cancer cells. The unique properties of nanoparticles, such as their small size, large surface-to-volume ratios, and the ability to achieve multivalency of targeting ligands on their surface, provide superior advantages for nanoparticle-based drug delivery to a variety of cancers. This review highlights various key concepts in the design of targeted nanotherapeutics for cancer therapy, and discusses physicochemical parameters affecting nanoparticle targeting, along with recent developments for cancer-targeted nanomedicines.
Collapse
Affiliation(s)
| | | | - Joseph Kaplinsky
- Department of Micro and Nanotechnology, DTU Nanotech, Technical University of Denmark, Produktionstorvet, 2800, Kongens Lyngby, Denmark
| | - Nazila Kamaly
- Department of Micro and Nanotechnology, DTU Nanotech, Technical University of Denmark, Produktionstorvet, 2800, Kongens Lyngby, Denmark.
| |
Collapse
|
58
|
Maranhão RC, Vital CG, Tavoni TM, Graziani SR. Clinical experience with drug delivery systems as tools to decrease the toxicity of anticancer chemotherapeutic agents. Expert Opin Drug Deliv 2017; 14:1217-1226. [PMID: 28042707 DOI: 10.1080/17425247.2017.1276560] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The toxicity of chemotherapeutic agents, resulting from their low pharmacological index, introduces considerable discomfort and risk to cancer patients. Among several strategies to reduce the toxicity of chemotherapeutic agents, targeted drug delivery is the most promising one. Areas covered: Liposomes, micelles, albumin-based, polymeric, dendritic and lipid core nanoparticles have been used as carriers to concentrate anticancer drugs in neoplastic tissues, and clinical studies of those preparations are reviewed. In most clinical studies, drug delivery systems reduced drug toxicity. Lipid core nanoparticles (LDE) that bind to cell lipoprotein receptors have the ability to concentrate in neoplastic tissues and were the first artificial non-liposomal system shown in in vivo studies to possess targeting properties. The toxicity reduction achieved by LDE as vehicle of carmustine, etoposide and paclitaxel was singularly strong. Expert opinion: The reduced toxicity offered by drug delivery systems has expanded treatment population that may benefit from chemotherapy including feeble, overtreated and elderly patients that would otherwise be offered palliative therapy. Drug delivery systems may either prolong the duration of treatments or allow increases in drug dose.
Collapse
Affiliation(s)
- Raul C Maranhão
- a Heart Institute of the Medical School Hospital , University of São Paulo , São Paulo , Brazil.,b Faculty of Pharmaceutical Sciences , University of São Paulo , São Paulo , Brazil
| | - Carolina G Vital
- a Heart Institute of the Medical School Hospital , University of São Paulo , São Paulo , Brazil.,b Faculty of Pharmaceutical Sciences , University of São Paulo , São Paulo , Brazil
| | - Thauany M Tavoni
- a Heart Institute of the Medical School Hospital , University of São Paulo , São Paulo , Brazil.,b Faculty of Pharmaceutical Sciences , University of São Paulo , São Paulo , Brazil
| | - Silvia R Graziani
- a Heart Institute of the Medical School Hospital , University of São Paulo , São Paulo , Brazil
| |
Collapse
|
59
|
To exploit the tumor microenvironment: Since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release 2016; 244:108-121. [DOI: 10.1016/j.jconrel.2016.11.015] [Citation(s) in RCA: 752] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/26/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022]
|
60
|
Liposomal drug delivery systems for targeted cancer therapy: is active targeting the best choice? Future Med Chem 2016; 8:2091-2112. [PMID: 27774793 DOI: 10.4155/fmc-2016-0135] [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] [Indexed: 02/07/2023] Open
Abstract
Liposomes are biodegradable and biocompatible self-forming spherical lipid bilayer vesicles. They can encapsulate and deliver one or more hydrophobic and hydrophilic therapeutic agents with poor therapeutic indices to tumor sites. Properties such as lipid bilayer fluidity, charge, size and surface hydration can be modified to extend liposome circulation time in the bloodstream and enhance efficacy. The focus of this review is on ligand-conjugated liposomes and their potential application in tumor-targeted delivery. Ligand-conjugated liposomes are designed to target receptors which are overexpressed on tumor cells to decrease drugs side effects by enhancing their selective delivery to tumor site. Despite the extensive research in this area, no small molecule ligand-conjugated liposome has been approved up to date for cancer therapy.
Collapse
|
61
|
Trends on polymer- and lipid-based nanostructures for parenteral drug delivery to tumors. Cancer Chemother Pharmacol 2016; 79:251-265. [PMID: 27744564 DOI: 10.1007/s00280-016-3168-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/06/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE The dawn of the state-of-the-art methods of cancer treatments, nano-based delivery systems, has dispensed with the mainstream chemotherapy for being inadequate in yielding productive results and the numerous reported side effects. The popularity of this complementary approach in the course of the last two decades has been primarily attributed to its capacity to elevate the therapeutic index of anticancer drugs as well as removing the impassable delivery barriers in solid tumors with the minimal damage to the normal tissues. METHODS The PubMed database was consulted to compile this review. RESULTS A wide range of minuscule organic and inorganic nanomaterials, with dimensions not exceeding hundred nanometers, has led to hope for cancer therapy to flare-up once again due to possessing a number of exclusive traits for passive and active tumor targeting, some of which are EPR effect, high interstitial pressure of tumor, overexpressed receptors and angiogenesis. Although a limited number of liposomal and polymer-based therapeutic nanoparticles have gained applicability, a vast number of nanoparticles are still being trailed in order to be fully developed. CONCLUSIONS This study provides an overview of the advantages/disadvantages of nanocarriers for cancer drug delivery.
Collapse
|
62
|
Au M, Emeto TI, Power J, Vangaveti VN, Lai HC. Emerging Therapeutic Potential of Nanoparticles in Pancreatic Cancer: A Systematic Review of Clinical Trials. Biomedicines 2016; 4:E20. [PMID: 28536387 PMCID: PMC5344258 DOI: 10.3390/biomedicines4030020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is an aggressive disease with a five year survival rate of less than 5%, which is associated with late presentation. In recent years, research into nanomedicine and the use of nanoparticles as therapeutic agents for cancers has increased. This article describes the latest developments in the use of nanoparticles, and evaluates the risks and benefits of nanoparticles as an emerging therapy for pancreatic cancer. The Preferred Reporting Items of Systematic Reviews and Meta-Analyses checklist was used. Studies were extracted by searching the Embase, MEDLINE, SCOPUS, Web of Science, and Cochrane Library databases from inception to 18 March 2016 with no language restrictions. Clinical trials involving the use of nanoparticles as a therapeutic or prognostic option in patients with pancreatic cancer were considered. Selected studies were evaluated using the Jadad score for randomised control trials and the Therapy CA Worksheet for intervention studies. Of the 210 articles found, 10 clinical trials including one randomised control trial and nine phase I/II clinical trials met the inclusion criteria and were analysed. These studies demonstrated that nanoparticles can be used in conjunction with chemotherapeutic agents increasing their efficacy whilst reducing their toxicity. Increased efficacy of treatment with nanoparticles may improve the clinical outcomes and quality of life in patients with pancreatic cancer, although the long-term side effects are yet to be defined. The study registration number is CRD42015020009.
Collapse
Affiliation(s)
- Minnie Au
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, James Cook Drive, Douglas, Townsville QLD 4811, Australia.
- Townsville Cancer Centre, The Townsville Hospital, Townsville QLD 4814, Australia.
| | - Theophilus I Emeto
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, James Cook Drive, Douglas, Townsville QLD 4811, Australia.
| | - Jacinta Power
- Townsville Cancer Centre, The Townsville Hospital, Townsville QLD 4814, Australia.
| | - Venkat N Vangaveti
- College of Medicine and Dentistry, James Cook University, James Cook Drive, Douglas, Townsville QLD 4811, Australia.
| | - Hock C Lai
- Townsville Cancer Centre, The Townsville Hospital, Townsville QLD 4814, Australia.
| |
Collapse
|
63
|
Abstract
Targeted delivery allows drug molecules to preferentially accumulate at the sites of action and thus holds great promise to improve therapeutic index. Among various drug-targeting approaches, nanoparticle-based delivery systems offer some unique strengths and have achieved exciting preclinical and clinical results. Herein, we aim to provide a review on the recent development of cell membrane-coated nanoparticle system, a new class of biomimetic nanoparticles that combine both the functionalities of cellular membranes and the engineering flexibility of synthetic nanomaterials for effective drug delivery and novel therapeutics. This review is particularly focused on novel designs of cell membrane-coated nanoparticles as well as their underlying principles that facilitate the purpose of drug targeting. Three specific areas are highlighted, including: (i) cell membrane coating to prolong nanoparticle circulation, (ii) cell membrane coating to achieve cell-specific targeting and (iii) cell membrane coating for immune system targeting. Overall, cell membrane-coated nanoparticles have emerged as a novel class of targeted nanotherapeutics with strong potentials to improve on drug delivery and therapeutic efficacy for treatment of various diseases.
Collapse
Affiliation(s)
- Weiwei Gao
- a Department of NanoEngineering and Moores Cancer Center , University of California , San Diego , La Jolla , CA , USA
| | - Liangfang Zhang
- a Department of NanoEngineering and Moores Cancer Center , University of California , San Diego , La Jolla , CA , USA
| |
Collapse
|
64
|
Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid-Based Drug Delivery Systems in Cancer Therapy: What Is Available and What Is Yet to Come. Pharmacol Rev 2016; 68:701-87. [PMID: 27363439 PMCID: PMC4931871 DOI: 10.1124/pr.115.012070] [Citation(s) in RCA: 436] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer is a leading cause of death in many countries around the world. However, the efficacy of current standard treatments for a variety of cancers is suboptimal. First, most cancer treatments lack specificity, meaning that these treatments affect both cancer cells and their normal counterparts. Second, many anticancer agents are highly toxic, and thus, limit their use in treatment. Third, a number of cytotoxic chemotherapeutics are highly hydrophobic, which limits their utility in cancer therapy. Finally, many chemotherapeutic agents exhibit short half-lives that curtail their efficacy. As a result of these deficiencies, many current treatments lead to side effects, noncompliance, and patient inconvenience due to difficulties in administration. However, the application of nanotechnology has led to the development of effective nanosized drug delivery systems known commonly as nanoparticles. Among these delivery systems, lipid-based nanoparticles, particularly liposomes, have shown to be quite effective at exhibiting the ability to: 1) improve the selectivity of cancer chemotherapeutic agents; 2) lower the cytotoxicity of anticancer drugs to normal tissues, and thus, reduce their toxic side effects; 3) increase the solubility of hydrophobic drugs; and 4) offer a prolonged and controlled release of agents. This review will discuss the current state of lipid-based nanoparticle research, including the development of liposomes for cancer therapy, different strategies for tumor targeting, liposomal formulation of various anticancer drugs that are commercially available, recent progress in liposome technology for the treatment of cancer, and the next generation of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Phatsapong Yingchoncharoen
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, Faculty of Medicine, Bosch Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
65
|
Heat-Shock Protein 90–Targeted Nano Anticancer Therapy. J Pharm Sci 2016; 105:1454-66. [DOI: 10.1016/j.xphs.2015.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/16/2015] [Accepted: 10/12/2015] [Indexed: 11/20/2022]
|
66
|
Abstract
PURPOSE OF REVIEW Recent advances in nanotechnology have addressed some of the issues related to lack of selectivity and nonspecific toxicities associated with conventional chemotherapy. Nanoparticles are therapeutic carriers that can be fine tuned for specific application and for passive or active tumor targeting. RECENT FINDINGS Although the nanoparticle field is rapidly expanding, there are to date only six nanoparticle-based drug delivery platforms and two antibody-drug conjugates that are clinically approved for cancer therapy. Here, we review the clinical data of liposomal anthracyclines, nanoparticle formulations of paclitaxel and trastuzumab emtansine. We then briefly comment on efficacy and safety issues of nanoparticles, as well as on the next-generation nanoparticles for cancer therapy. SUMMARY The emerging development of cancer nanotechnology offers the opportunity of reinvestigating the potential of cytotoxic agents, improving tumor targeting and drug delivery, leading to better safety profile and antitumor activity. Adding specificity to nanoparticles may allow personalization of cancer therapy using chemotherapy.
Collapse
|
67
|
Brys AK, Gowda R, Loriaux DB, Robertson GP, Mosca PJ. Nanotechnology-based strategies for combating toxicity and resistance in melanoma therapy. Biotechnol Adv 2016; 34:565-577. [PMID: 26826558 DOI: 10.1016/j.biotechadv.2016.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/15/2015] [Accepted: 01/16/2016] [Indexed: 12/17/2022]
Abstract
Drug toxicity and resistance remain formidable challenges in cancer treatment and represent an area of increasing attention in the case of melanoma. Nanotechnology represents a paradigm-shifting field with the potential to mitigate drug resistance while improving drug delivery and minimizing toxicity. Recent clinical and pre-clinical studies have demonstrated how a diverse array of nanoparticles may be harnessed to circumvent known mechanisms of drug resistance in melanoma to improve therapeutic efficacy. In this review, we discuss known mechanisms of resistance to various melanoma therapies and possible nanotechnology-based strategies that could be used to overcome these barriers and improve the pharmacologic arsenal available to combat advanced stage melanoma.
Collapse
Affiliation(s)
- Adam K Brys
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States
| | - Raghavendra Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Daniel B Loriaux
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Paul J Mosca
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
68
|
van der Meel R, Vehmeijer LJC, Kok RJ, Storm G, van Gaal EVB. Ligand-targeted Particulate Nanomedicines Undergoing Clinical Evaluation: Current Status. INTRACELLULAR DELIVERY III 2016. [DOI: 10.1007/978-3-319-43525-1_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
69
|
Shargh VH, Hondermarck H, Liang M. Antibody-targeted biodegradable nanoparticles for cancer therapy. Nanomedicine (Lond) 2016; 11:63-79. [DOI: 10.2217/nnm.15.186] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanotechnology has great potentials to revolutionize the future cancer diagnosis and therapy. In this context, various nanoparticles (NPs) have been developed for targeted delivery of diagnostic/therapeutic agents to the tumor sites, which thus result in greater efficacy and much less side effects. The targeting property of NPs is often achieved by functionalizing their surface with tumor-specific ligands, such as antibodies, peptides, small molecules and oligonucleotides. In this review, we will discuss recent progress in the multifunctional design of antibody-targeted NPs with a special focus on liposomal, polymeric and protein-based delivery systems.
Collapse
Affiliation(s)
- Vahid Heravi Shargh
- School of Biomedical Sciences & Pharmacy, Faculty of Health & Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences & Pharmacy, Faculty of Health & Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Mingtao Liang
- School of Biomedical Sciences & Pharmacy, Faculty of Health & Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
70
|
Stylianopoulos T, Jain RK. Design considerations for nanotherapeutics in oncology. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2015; 11:1893-907. [PMID: 26282377 PMCID: PMC4628869 DOI: 10.1016/j.nano.2015.07.015] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/20/2015] [Accepted: 07/22/2015] [Indexed: 12/24/2022]
Abstract
Nanotherapeutics have improved the quality of life of cancer patients, primarily by reducing the adverse effects of chemotherapeutic agents, but improvements in overall survival are modest. This is in large part due to the fact that the enhanced permeability and retention effect, which is the basis for the use of nanoparticles in cancer, can be also a barrier to the delivery of nanomedicines. A careful design of nanoparticle formulations can overcome barriers posed by the tumor microenvironment and result in better treatments. In this review, we first discuss strengths and limitations of clinically-approved nanoparticles. Then, we evaluate design parameters that can be modulated to optimize delivery. The benefits of active tumor targeting and drug release rate on intratumoral delivery and treatment efficacy are also discussed. Finally, we suggest specific design strategies that should optimize delivery to most solid tumors and discuss under what conditions active targeting would be beneficial. FROM THE CLINICAL EDITOR Advances in nanotechnology have seen the introduction of new treatment modalities for cancer. The principle of action using nanocarriers for drug delivery is based mostly on the Enhanced Permeability and Retention effect. This phenomenon however, can also be a hindrance. In this article, the authors performed an in-depth review on various nanoparticle platforms in cancer therapeutics. They also suggested options to improve drug delivery, in terms of carrier design.
Collapse
Affiliation(s)
- Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
71
|
Liu D, Auguste DT. Cancer targeted therapeutics: From molecules to drug delivery vehicles. J Control Release 2015; 219:632-643. [PMID: 26342659 DOI: 10.1016/j.jconrel.2015.08.041] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The pitfall of all chemotherapeutics lies in drug resistance and the severe side effects experienced by patients. One way to reduce the off-target effects of chemotherapy on healthy tissues is to alter the biodistribution of drug. This can be achieved in two ways: Passive targeting utilizes shape, size, and surface chemistry to increase particle circulation and tumor accumulation. Active targeting employs either chemical moieties (e.g. peptides, sugars, aptamers, antibodies) to selectively bind to cell membranes or responsive elements (e.g. ultrasound, magnetism, light) to deliver its cargo within a local region. This article will focus on the systemic administration of anti-cancer agents and their ability to home to tumors and, if relevant, distant metastatic sites.
Collapse
Affiliation(s)
- Daxing Liu
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, United States
| | - Debra T Auguste
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, United States.
| |
Collapse
|
72
|
Khan DR, Webb MN, Cadotte TH, Gavette MN. Use of Targeted Liposome-based Chemotherapeutics to Treat Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:1-5. [PMID: 26309409 PMCID: PMC4533644 DOI: 10.4137/bcbcr.s29421] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 11/05/2022]
Abstract
The use of nanocarriers such as liposomes to deliver anticancer drugs to tumors can significantly enhance the therapeutic index of otherwise unencapsulated cytotoxic agents. This is in part because of the fact that the phospholipid bilayer can protect healthy sensitive tissue from the damaging effects of these types of drugs. Furthermore, the ease with which the phospholipid bilayer surface can be modified to allow for polyethylene glycol incorporation resulting in pegylated liposomes allow for increased circulation times in vivo, and thus an overall increase in the concentration of the drug delivered to the tumor site. This explains the clinical success of the liposomal-based drug Doxil, which has proven to be quite efficacious in the treatment of breast cancer. However, significant challenges remain involving poor drug transfer between the liposome and tumor cells with this type of nontargeted drug delivery system. Thus, future work involves the development of "smart" drugs, or targeted drug delivery intended for improved colocalization between the drug and cancerous cells. While it is not possible to entirely discuss such a rapidly growing field of study involving many different types of chemotherapeutics here, in this review, we discuss some of the recent advancements involving the development of targeted liposome-based chemotherapeutics to treat breast cancer.
Collapse
Affiliation(s)
- David R Khan
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| | - Maggie N Webb
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| | - Thomas H Cadotte
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| | - Madison N Gavette
- Department of Mathematics, Chemistry and Physics, West Texas A&M University, Canyon, TX, USA
| |
Collapse
|
73
|
Abstract
Theranostics is a promising field that combines therapeutics and diagnostics into single multifunctional formulations. This field is driven by advancements in nanoparticle systems capable of providing the necessary functionalities. By utilizing these powerful nanomedicines, the concept of personalized medicine can be realized by tailoring treatment strategies to the individual. This review gives a brief overview of the components of a theranostic system and the challenges that designing truly multifunctional nanoparticles present. Considerations when choosing a class of nanoparticle include the size, shape, charge, and surface chemistry, while classes of nanoparticles discussed are polymers, liposomes, dendrimers, and polymeric micelles. Targeting to disease states can be achieved either through passive or active targeting which uses specific ligands to target receptors that are overexpressed in tumors and common targeting elements are presented. To image the interactions with disease states, contrast agents are included in the nanoparticle formulation. Imaging options include optical imaging techniques, computed tomography, nuclear based, and magnetic resonance imaging. The interplay between all of these components needs to be carefully considered when designing a theranostic system.
Collapse
|
74
|
Sun L, Wu Q, Peng F, Liu L, Gong C. Strategies of polymeric nanoparticles for enhanced internalization in cancer therapy. Colloids Surf B Biointerfaces 2015; 135:56-72. [PMID: 26241917 DOI: 10.1016/j.colsurfb.2015.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/19/2015] [Accepted: 07/07/2015] [Indexed: 02/05/2023]
Abstract
In order to achieve long circulation time and high drug accumulation in the tumor sites via the EPR effects, anticancer drugs have to be protected by non-fouling polymers such as poly(ethylene glycol) (PEG), poly(ethylene oxide) (PEO), dextran, and poly(acrylic acid) (PAA). However, the dense layer of stealth polymer also prohibits efficient uptake of anticancer drugs by target cancer cells. For cancer therapy, it is often more desirable to accomplish rapid cellular uptake after anticancer drugs arriving at the pathological site, which could on one hand maximize the therapeutic efficacy and on the other hand reduce probability of drug resistance in cells. In this review, special attention will be focused on the recent potential strategies that can enable drug-loaded polymeric nanoparticles to rapidly recognize cancer cells, leading to enhanced internalization.
Collapse
Affiliation(s)
- Lu Sun
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qinjie Wu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Feng Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Changyang Gong
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
75
|
Affiliation(s)
- Bhushan S Pattni
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Vladimir V Chupin
- Laboratory for Advanced Studies of Membrane Proteins, Moscow Institute of Physics and Technology , Dolgoprudny 141700, Russia
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States.,Department of Biochemistry, Faculty of Science, King Abdulaziz University , Jeddah 21589, Saudi Arabia
| |
Collapse
|
76
|
Fernandes E, Ferreira JA, Andreia P, Luís L, Barroso S, Sarmento B, Santos LL. New trends in guided nanotherapies for digestive cancers: A systematic review. J Control Release 2015; 209:288-307. [PMID: 25957905 DOI: 10.1016/j.jconrel.2015.05.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/02/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
Digestive tract tumors are among the most common and deadliest malignancies worldwide, mainly due to late diagnosis and lack of efficient therapeutics. Current treatments essentially rely on surgery associated with (neo)adjuvant chemotherapy agents. Despite an upfront response, conventional drugs often fail to eliminate highly aggressive clones endowed with chemoresistant properties, which are responsible for tumor recurrence and disease dissemination. Synthetic drugs also present severe adverse systemic effects, hampering the administration of biologically effective dosages. Nanoencapsulation of chemotherapeutic agents within biocompatible polymeric or lipid matrices holds great potential to improve the pharmacokinetics and efficacy of conventional chemotherapy while reducing systemic toxicity. Tagging nanoparticle surfaces with specific ligands for cancer cells, namely monoclonal antibodies or antibody fragments, has provided means to target more aggressive clones, further improving the selectivity and efficacy of nanodelivery vehicles. In fact, over the past twenty years, significant research has translated into a wide array of guided nanoparticles, providing the molecular background for a new generation of intelligent and more effective anti-cancer agents. Attempting to bring awareness among the medical community to emerging targeted nanopharmaceuticals and foster advances in the field, we have conducted a systematic review about this matter. Emphasis was set on ongoing preclinical and clinical trials for liver, colorectal, gastric and pancreatic cancers. To the best of our knowledge this is the first systematic and integrated overview on this field. Using a specific query, 433 abstracts were gathered and narrowed to 47 manuscripts when matched against inclusion/exclusion criteria. All studies showed that active targeting improves the effectiveness of the nanodrugs alone, while lowering its side effects. The main focus has been on hepatocarcinomas, mainly by exploring glycans as homing molecules. Other ligands such as peptides/small proteins and antibodies/antibody fragments, with affinity to either tumor vasculature or tumor cells, have also been widely and successfully applied to guide nanodrugs to gastrointestinal carcinomas. Conversely, few solutions have been presented for pancreatic tumors. To this date only three nanocomplexes have progressed beyond pre-clinical stages: i) PK2, a galactosamine-functionalized polymeric-DOX formulation for hepatocarcinomas; ii) MCC-465, an anti-(myosin heavy chain a) immunoliposome for advanced stage metastatic solid tumors; and iii) MBP-426, a transferrin-liposome-oxaliplatin conjugate, also for advanced stage tumors. Still, none has been approved for clinical use. However, based on the high amount of pre-clinical studies showing enthusiastic results, the number of clinical trials is expected to increase in the near future. A more profound understanding about the molecular nature of chemoresistant clones and cancer stem cell biology will also contribute to boost the field of guided nanopharmacology towards more effective solutions.
Collapse
Affiliation(s)
- Elisabete Fernandes
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| | - Peixoto Andreia
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - Lima Luís
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Nucleo de Investigação em Farmácia - Centro de Investigação em Saúde e Ambiente (CISA), Health School of the Polytechnic Institute of Porto, Porto, Portugal
| | - Sérgio Barroso
- Serviço de Oncologia, Hospital de Évora, Évora, Portugal
| | - Bruno Sarmento
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal; CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRD, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal; Health School of University of Fernando Pessoa, Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| |
Collapse
|
77
|
Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm 2015; 93:52-79. [PMID: 25813885 DOI: 10.1016/j.ejpb.2015.03.018] [Citation(s) in RCA: 1035] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 02/08/2023]
Abstract
Cancer is the second worldwide cause of death, exceeded only by cardiovascular diseases. It is characterized by uncontrolled cell proliferation and an absence of cell death that, except for hematological cancers, generates an abnormal cell mass or tumor. This primary tumor grows thanks to new vascularization and, in time, acquires metastatic potential and spreads to other body sites, which causes metastasis and finally death. Cancer is caused by damage or mutations in the genetic material of the cells due to environmental or inherited factors. While surgery and radiotherapy are the primary treatment used for local and non-metastatic cancers, anti-cancer drugs (chemotherapy, hormone and biological therapies) are the choice currently used in metastatic cancers. Chemotherapy is based on the inhibition of the division of rapidly growing cells, which is a characteristic of the cancerous cells, but unfortunately, it also affects normal cells with fast proliferation rates, such as the hair follicles, bone marrow and gastrointestinal tract cells, generating the characteristic side effects of chemotherapy. The indiscriminate destruction of normal cells, the toxicity of conventional chemotherapeutic drugs, as well as the development of multidrug resistance, support the need to find new effective targeted treatments based on the changes in the molecular biology of the tumor cells. These novel targeted therapies, of increasing interest as evidenced by FDA-approved targeted cancer drugs in recent years, block biologic transduction pathways and/or specific cancer proteins to induce the death of cancer cells by means of apoptosis and stimulation of the immune system, or specifically deliver chemotherapeutic agents to cancer cells, minimizing the undesirable side effects. Although targeted therapies can be achieved directly by altering specific cell signaling by means of monoclonal antibodies or small molecules inhibitors, this review focuses on indirect targeted approaches that mainly deliver chemotherapeutic agents to molecular targets overexpressed on the surface of tumor cells. In particular, we offer a detailed description of different cytotoxic drug carriers, such as liposomes, carbon nanotubes, dendrimers, polymeric micelles, polymeric conjugates and polymeric nanoparticles, in passive and active targeted cancer therapy, by enhancing the permeability and retention or by the functionalization of the surface of the carriers, respectively, emphasizing those that have received FDA approval or are part of the most important clinical studies up to date. These drug carriers not only transport the chemotherapeutic agents to tumors, avoiding normal tissues and reducing toxicity in the rest of the body, but also protect cytotoxic drugs from degradation, increase the half-life, payload and solubility of cytotoxic agents and reduce renal clearance. Despite the many advantages of all the anticancer drug carriers analyzed, only a few of them have reached the FDA approval, in particular, two polymer-protein conjugates, five liposomal formulations and one polymeric nanoparticle are available in the market, in contrast to the sixteen FDA approval of monoclonal antibodies. However, there are numerous clinical trials in progress of polymer-protein and polymer-drug conjugates, liposomal formulations, including immunoliposomes, polymeric micelles and polymeric nanoparticles. Regarding carbon nanotubes or dendrimers, there are no FDA approvals or clinical trials in process up to date due to their unresolved toxicity. Moreover, we analyze in detail the more promising and advanced preclinical studies of the particular case of polymeric nanoparticles as carriers of different cytotoxic agents to active and passive tumor targeting published in the last 5 years, since they have a huge potential in cancer therapy, being one of the most widely studied nano-platforms in this field in the last years. The interest that these formulations have recently achieved is stressed by the fact that 90% of the papers based on cancer therapeutics with polymeric nanoparticles have been published in the last 6 years (PubMed search).
Collapse
|
78
|
Estanqueiro M, Amaral MH, Conceição J, Sousa Lobo JM. Nanotechnological carriers for cancer chemotherapy: The state of the art. Colloids Surf B Biointerfaces 2015; 126:631-48. [DOI: 10.1016/j.colsurfb.2014.12.041] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 12/15/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022]
|
79
|
Hosseini M, Haji-Fatahaliha M, Jadidi-Niaragh F, Majidi J, Yousefi M. The use of nanoparticles as a promising therapeutic approach in cancer immunotherapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1051-61. [PMID: 25612903 DOI: 10.3109/21691401.2014.998830] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
80
|
Recent advances in targeted nanoparticles drug delivery to melanoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:769-94. [PMID: 25555352 DOI: 10.1016/j.nano.2014.11.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/31/2014] [Accepted: 11/15/2014] [Indexed: 12/30/2022]
Abstract
Melanoma is one of the most aggressive skin cancers, notorious for its high multidrug resistance and low survival rate. Conventional therapies (e.g., dacarbazine, interferon-alpha-2b and interleukin-2) are limited by low response rate and demonstrate no overall survival benefit. Novel targeted therapies (e.g., vemurafenib, dabrafenib and trametinib) have higher initial response rate and clear impact on the overall survival, but relapse usually occurs within 6 to 9 months. Although immunotherapy (e.g., ipilimumab, pembrolizumab and nivolumab) can achieve long-term and durable response, rate of adverse events is extremely high. With the development of nanotechnology, the applications of nanocarriers are widely expected to change the landscape of melanoma therapy for foreseeable future. In this review, we will relate recent advances in the application of multifunctional nanocarriers for targeted drug delivery to melanoma, in melanoma nanotheranostics and combination therapy, and nanopharmaceutical associated melanoma clinical trials, followed by challenges and perspectives. From the clinical editor: The team of authors describes the current treatment regimes of malignant melanoma emphasizing the importance of achieving a better efficacy and the need to develop a better understanding of melanoma tumorigenesis.
Collapse
|
81
|
Nanomedicine in cancer therapy: challenges, opportunities, and clinical applications. J Control Release 2014; 200:138-57. [PMID: 25545217 DOI: 10.1016/j.jconrel.2014.12.030] [Citation(s) in RCA: 1191] [Impact Index Per Article: 119.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of death worldwide. Currently available therapies are inadequate and spur demand for improved technologies. Rapid growth in nanotechnology towards the development of nanomedicine products holds great promise to improve therapeutic strategies against cancer. Nanomedicine products represent an opportunity to achieve sophisticated targeting strategies and multi-functionality. They can improve the pharmacokinetic and pharmacodynamic profiles of conventional therapeutics and may thus optimize the efficacy of existing anti-cancer compounds. In this review, we discuss state-of-the-art nanoparticles and targeted systems that have been investigated in clinical studies. We emphasize the challenges faced in using nanomedicine products and translating them from a preclinical level to the clinical setting. Additionally, we cover aspects of nanocarrier engineering that may open up new opportunities for nanomedicine products in the clinic.
Collapse
|
82
|
The potential of liposomes with carbonic anhydrase IX to deliver anticancer ingredients to cancer cells in vivo. Int J Mol Sci 2014; 16:230-55. [PMID: 25547490 PMCID: PMC4307245 DOI: 10.3390/ijms16010230] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/16/2014] [Indexed: 12/19/2022] Open
Abstract
Drug delivery nanocarriers, especially targeted drug delivery by liposomes are emerging as a class of therapeutics for cancer. Early research results suggest that liposomal therapeutics enhanced efficacy, while simultaneously reducing side effects, owing to properties such as more targeted localization in tumors and active cellular uptake. Here, we highlight the features of immunoliposomes that distinguish them from previous anticancer therapies, and describe how these features provide the potential for therapeutic effects that are not achievable with other modalities. While a large number of studies has been published, the emphasis here is placed on the carbonic anhydrase IX (CA-IX) and the conjugated liposomes that are likely to open a new chapter on drug delivery system by using immunoliposomes to deliver anticancer ingredients to cancer cells in vivo.
Collapse
|
83
|
Kanapathipillai M, Brock A, Ingber DE. Nanoparticle targeting of anti-cancer drugs that alter intracellular signaling or influence the tumor microenvironment. Adv Drug Deliv Rev 2014; 79-80:107-18. [PMID: 24819216 DOI: 10.1016/j.addr.2014.05.005] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 04/22/2014] [Accepted: 05/01/2014] [Indexed: 12/13/2022]
Abstract
Nanoparticle-based therapeutics are poised to become a leading delivery strategy for cancer treatment because they potentially offer higher selectivity, reduced toxicity, longer clearance times, and increased efficacy compared to conventional systemic therapeutic approaches. This article reviews existing nanoparticle technologies and methods that are used to target drugs to treat cancer by altering signal transduction or modulating the tumor microenvironment. We also consider the implications of recent advances in the nanotherapeutics field for the future of cancer therapy.
Collapse
|
84
|
Tumor-targeted responsive nanoparticle-based systems for magnetic resonance imaging and therapy. Pharm Res 2014; 31:3487-502. [PMID: 24919932 PMCID: PMC4224753 DOI: 10.1007/s11095-014-1436-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/03/2014] [Indexed: 11/09/2022]
Abstract
Purpose Design and synthesis of a tumor responsive nanoparticle-based system for imaging and treatment of various cancers. Methods Manganese oxide nanoparticles (Mn3O4 NPs) were synthesized and modified with LHRH targeting peptide or anti-melanoma antibodies (cancer targeting moieties) and a MMP2 cleavable peptide (a possible chemotactic factor). Nanostructured lipid carriers (NLCs) were used to entrap the BRAF inhibitor, vemurafenib, and enhance cytotoxicity of the drug. Size distribution, stability, drug entrapment, cytotoxicity and genotoxicity of synthesized nanoparticles were studied in vitro. Enhancement of MRI signal by nanoparticles and their body distribution were examined in vivo on mouse models of melanoma, ovarian and lung cancers. Results Uniform, stable cancer-targeted nanoparticles (PEGylated water-soluble Mn3O4 NPs and NLCs) were synthesized. No signs of cyto-,genotoxicity and DNA damage were detected for nanoparticles that do not contain an anticancer drug. Entrapment of vemurafenib into nanoparticles significantly enhanced drug toxicity in cancer cells with targeted V600E mutation. The developed nanoparticles containing LHRH and MMP2 peptides showed preferential accumulation in primary and metastatic tumors increasing the MRI signal in mice with melanoma, lung and ovarian cancers. Conclusions The proposed nanoparticle-based systems provide the foundation for building an integrated MRI diagnostic and therapeutic approach for various types of cancer.
Collapse
|
85
|
Fonseca NA, Gregório AC, Valério-Fernandes A, Simões S, Moreira JN. Bridging cancer biology and the patients' needs with nanotechnology-based approaches. Cancer Treat Rev 2014; 40:626-35. [PMID: 24613464 DOI: 10.1016/j.ctrv.2014.02.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/06/2014] [Accepted: 02/12/2014] [Indexed: 01/27/2023]
Abstract
Cancer remains as stressful condition and a leading cause of death in the western world. Actual cornerstone treatments of cancer disease rest as an elusive alternative, offering limited efficacy with extensive secondary effects as a result of severe cytotoxic effects in healthy tissues. The advent of nanotechnology brought the promise to revolutionize many fields including oncology, proposing advanced systems for cancer treatment. Drug delivery systems rest among the most successful examples of nanotechnology. Throughout time they have been able to evolve as a function of an increased understanding from cancer biology and the tumor microenvironment. Marketing of Doxil® unleashed a remarkable impulse in the development of drug delivery systems. Since then, several nanocarriers have been introduced, with aspirations to overrule previous technologies, demonstrating increased therapeutic efficacy besides decreased toxicity. Spatial and temporal targeting to cancer cells has been explored, as well as the use of drug combinations co-encapsulated in the same particle as a mean to take advantage of synergistic interactions in vivo. Importantly, targeted delivery of siRNA for gene silencing therapy has made its way to the clinic for a "first in man" trial using lipid-polymeric-based particles. Focusing in state-of-the-art technology, this review will provide an insightful vision on nanotechnology-based strategies for cancer treatment, approaching them from a tumor biology-driven perspective, since their early EPR-based dawn to the ones that have truly the potential to address unmet medical needs in the field of oncology, upon targeting key cell subpopulations from the tumor microenvironment.
Collapse
Affiliation(s)
- Nuno A Fonseca
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Ana C Gregório
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Angela Valério-Fernandes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Sérgio Simões
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - João N Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
86
|
van Vught R, Pieters RJ, Breukink E. Site-specific functionalization of proteins and their applications to therapeutic antibodies. Comput Struct Biotechnol J 2014; 9:e201402001. [PMID: 24757499 PMCID: PMC3995230 DOI: 10.5936/csbj.201402001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/28/2014] [Accepted: 02/04/2014] [Indexed: 12/19/2022] Open
Abstract
Protein modifications are often required to study structure and function relationships. Instead of the random labeling of lysine residues, methods have been developed to (sequence) specific label proteins. Next to chemical modifications, tools to integrate new chemical groups for bioorthogonal reactions have been applied. Alternatively, proteins can also be selectively modified by enzymes. Herein we review the methods available for site-specific modification of proteins and their applications for therapeutic antibodies.
Collapse
Affiliation(s)
- Remko van Vught
- Department of Membrane Biochemistry and Biophysics, Institute of Biomembranes, Utrecht University, Padualaan 8, 3584CH Utrecht, The Netherlands
| | - Roland J Pieters
- Department of Medicinal Chemistry and Chemical Biology. Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - Eefjan Breukink
- Department of Membrane Biochemistry and Biophysics, Institute of Biomembranes, Utrecht University, Padualaan 8, 3584CH Utrecht, The Netherlands
| |
Collapse
|
87
|
Wang B, Galliford CV, Low PS. Guiding principles in the design of ligand-targeted nanomedicines. Nanomedicine (Lond) 2014; 9:313-30. [PMID: 24552563 DOI: 10.2217/nnm.13.175] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Medicines for the treatment of most human pathologies are encumbered by unwanted side effects that arise from the deposition of an effective drug into the wrong tissues. The logical remedy for these undesirable properties involves selective targeting of the therapeutic agent to pathologic cells, thereby avoiding collateral toxicity to healthy cells. Since significant advantages can also accrue by incorporating a therapeutic or imaging agent into a nanoparticle, many laboratories are now combining both benefits into a single formulation. This review will focus on the major guiding principles in the design of ligand-targeted nanoparticles, including optimization of their chemical and physical properties, selection of the ideal targeting ligand, engineering of the appropriate surface passivation and linker strategies to achieve selective delivery of the entrapped cargo to the desired diseased cell.
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Chris V Galliford
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
88
|
Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014; 66:2-25. [PMID: 24270007 PMCID: PMC4219254 DOI: 10.1016/j.addr.2013.11.009] [Citation(s) in RCA: 1889] [Impact Index Per Article: 188.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/23/2013] [Accepted: 11/13/2013] [Indexed: 12/17/2022]
Abstract
Cancer nanotherapeutics are progressing at a steady rate; research and development in the field has experienced an exponential growth since early 2000's. The path to the commercialization of oncology drugs is long and carries significant risk; however, there is considerable excitement that nanoparticle technologies may contribute to the success of cancer drug development. The pace at which pharmaceutical companies have formed partnerships to use proprietary nanoparticle technologies has considerably accelerated. It is now recognized that by enhancing the efficacy and/or tolerability of new drug candidates, nanotechnology can meaningfully contribute to create differentiated products and improve clinical outcome. This review describes the lessons learned since the commercialization of the first-generation nanomedicines including DOXIL® and Abraxane®. It explores our current understanding of targeted and non-targeted nanoparticles that are under various stages of development, including BIND-014 and MM-398. It highlights the opportunities and challenges faced by nanomedicines in contemporary oncology, where personalized medicine is increasingly the mainstay of cancer therapy. We revisit the fundamental concepts of enhanced permeability and retention effect (EPR) and explore the mechanisms proposed to enhance preferential "retention" in the tumor, whether using active targeting of nanoparticles, binding of drugs to their tumoral targets or the presence of tumor associated macrophages. The overall objective of this review is to enhance our understanding in the design and development of therapeutic nanoparticles for treatment of cancers.
Collapse
Affiliation(s)
- Nicolas Bertrand
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun Wu
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Xiaoyang Xu
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Omid C Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA.
| |
Collapse
|
89
|
Abstract
Recent advances in nanotechnology and biotechnology have contributed to the development of engineered nanoscale materials as innovative prototypes to be used for biomedical applications and optimized therapy. Due to their unique features, including a large surface area, structural properties, and a long circulation time in blood compared with small molecules, a plethora of nanomaterials has been developed, with the potential to revolutionize the diagnosis and treatment of several diseases, in particular by improving the sensitivity and recognition ability of imaging contrast agents and by selectively directing bioactive agents to biological targets. Focusing on cancer, promising nanoprototypes have been designed to overcome the lack of specificity of conventional chemotherapeutic agents, as well as for early detection of precancerous and malignant lesions. However, several obstacles, including difficulty in achieving the optimal combination of physicochemical parameters for tumor targeting, evading particle clearance mechanisms, and controlling drug release, prevent the translation of nanomedicines into therapy. In spite of this, recent efforts have been focused on developing functionalized nanoparticles for delivery of therapeutic agents to specific molecular targets overexpressed on different cancer cells. In particular, the combination of targeted and controlled-release polymer nanotechnologies has resulted in a new programmable nanotherapeutic formulation of docetaxel, namely BIND-014, which recently entered Phase II clinical testing for patients with solid tumors. BIND-014 has been developed to overcome the limitations facing delivery of nanoparticles to many neoplasms, and represents a validated example of targeted nanosystems with the optimal biophysicochemical properties needed for successful tumor eradication.
Collapse
Affiliation(s)
- Vanna Sanna
- Department of Chemistry and Pharmacy, Laboratory of Nanomedicine, University of Sassari, Sassari, Italy
| | - Nicolino Pala
- Department of Chemistry and Pharmacy, Laboratory of Nanomedicine, University of Sassari, Sassari, Italy
| | - Mario Sechi
- Department of Chemistry and Pharmacy, Laboratory of Nanomedicine, University of Sassari, Sassari, Italy
| |
Collapse
|
90
|
Upponi JR, Torchilin VP. Passive vs. Active Targeting: An Update of the EPR Role in Drug Delivery to Tumors. NANO-ONCOLOGICALS 2014. [DOI: 10.1007/978-3-319-08084-0_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
91
|
Abstract
In recent years, there has been an unprecedented expansion in the field of nanomedicine with the development of new nanoparticles for the diagnosis and treatment of cancer. Nanoparticles have unique biological properties given their small size and large surface area-to-volume ratio, which allows them to bind, absorb, and carry compounds such as small molecule drugs, DNA, RNA, proteins, and probes with high efficiency. Their tunable size, shape, and surface characteristics also enable them to have high stability, high carrier capacity, the ability to incorporate both hydrophilic and hydrophobic substances and compatibility with different administration routes, thereby making them highly attractive in many aspects of oncology. This review article will discuss how nanoparticles are able to function as carriers for chemotherapeutic drugs to increase their therapeutic index; how they can function as therapeutic agents in photodynamic, gene, and thermal therapy; and how nanoparticles can be used as molecular imaging agents to detect and monitor cancer progression.
Collapse
Affiliation(s)
- Avnesh S Thakor
- Visiting Research Scholar, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA; Fellow in Interventional Radiology, University of British Columbia, Vancouver General Hospital, Vancouver, British Columbia, Canada; Academic Fellow, Department of Interventional Radiology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
92
|
Multifunctional nanoparticles for targeted delivery of immune activating and cancer therapeutic agents. J Control Release 2013; 172:1020-34. [PMID: 24140748 DOI: 10.1016/j.jconrel.2013.10.012] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 01/20/2023]
Abstract
Nanoparticles (NPs) have been extensively investigated for applications in both experimental and clinical settings to improve delivery efficiency of therapeutic and diagnostic agents. Most recently, novel multifunctional nanoparticles have attracted much attention because of their ability to carry diverse functionalities to achieve effective synergistic therapeutic treatments. Multifunctional NPs have been designed to co-deliver multiple components, target the delivery of drugs by surface functionalization, and realize therapy and diagnosis simultaneously. In this review, various materials of diverse chemistries for fabricating multifunctional NPs with distinctive architectures are discussed and compared. Recent progress involving multifunctional NPs for immune activation, anticancer drug delivery, and synergistic theranostics is the focus of this review. Overall, this comprehensive review demonstrates that multifunctional NPs have distinctive properties that make them highly suitable for targeted therapeutic delivery in these areas.
Collapse
|
93
|
van der Meel R, Vehmeijer LJC, Kok RJ, Storm G, van Gaal EVB. Ligand-targeted particulate nanomedicines undergoing clinical evaluation: current status. Adv Drug Deliv Rev 2013; 65:1284-98. [PMID: 24018362 DOI: 10.1016/j.addr.2013.08.012] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 08/09/2013] [Accepted: 08/29/2013] [Indexed: 12/25/2022]
Abstract
Since the introduction of Doxil® on the market nearly 20years ago, a number of nanomedicines have become part of treatment regimens in the clinic. With the exception of antibody-drug conjugates, these nanomedicines are all devoid of targeting ligands and rely solely on their physicochemical properties and the (patho)physiological processes in the body for their biodistribution and targeting capability. At the same time, many preclinical studies have reported on nanomedicines exposing targeting ligands, or ligand-targeted nanomedicines, yet none of these have been approved at this moment. In the present review, we provide a concise overview of 13 ligand-targeted particulate nanomedicines (ligand-targeted PNMs) that have progressed into clinical trials. The progress of each ligand-targeted PNM is discussed based on available (pre)clinical data. Main conclusions of these analyses are that (a) ligand-targeted PNMs have proven to be safe and efficacious in preclinical models; (b) the vast majority of ligand-targeted PNMs is generated for the treatment of cancer; (c) contribution of targeting ligands to the PNM efficacy is not unambiguously proven; and (d) targeting ligands do not cause localization of the PNM within the target tissue, but rather provide benefits in terms of target cell internalization and target tissue retention once the PNM has arrived at the target site. Increased understanding of the in vivo fate and interactions of the ligand-targeted PNMs with proteins and cells in the human body is mandatory to rationally advance the clinical translation of ligand-targeted PNMs. Future perspectives for ligand-targeted PNM approaches include the delivery of drugs that are unable or inefficient in passing cellular membranes, treatment of drug resistant tumors, targeting of the tumor blood supply, the generation of targeted vaccines and nanomedicines that are able to cross the blood-brain barrier.
Collapse
Affiliation(s)
- Roy van der Meel
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
94
|
Krishnan V, Rajasekaran AK. Clinical nanomedicine: a solution to the chemotherapy conundrum in pediatric leukemia therapy. Clin Pharmacol Ther 2013; 95:168-78. [PMID: 24013811 DOI: 10.1038/clpt.2013.174] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/27/2013] [Indexed: 01/01/2023]
Abstract
Although chemotherapy-based treatment of leukemia has tremendously improved survival rates in children, induction of treatment-related side effects is a major concern in clinical oncology. The development of nanotechnology-based drug delivery techniques to target clinically approved anticancer agents specifically to leukemic cells should diminish toxic side effects. This review aims to address the rational design of nanotherapeutics in treating hematologic malignancies with a focus on acute lymphoblastic leukemia (ALL)--the most prominent form of pediatric cancer.
Collapse
Affiliation(s)
- V Krishnan
- 1] Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA [2] Nemours Center for Childhood Cancer Research, A.I. duPont Hospital for Children, Wilmington, Delaware, USA [3] Delaware Biotechnology Institute, University of Delaware, Newark, Delaware, USA
| | - A K Rajasekaran
- 1] Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA [2] Nemours Center for Childhood Cancer Research, A.I. duPont Hospital for Children, Wilmington, Delaware, USA [3] Delaware Biotechnology Institute, University of Delaware, Newark, Delaware, USA [4] Department of Biological Sciences, Center for Translational Cancer Research, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
95
|
Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52:83-124. [PMID: 23299465 DOI: 10.1007/s40262-012-0027-4] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of monoclonal antibodies (mAbs) and their functional derivatives represents a growing segment of the development pipeline in the pharmaceutical industry. More than 25 mAbs and derivatives have been approved for a variety of therapeutic applications. In addition, around 500 mAbs and derivatives are currently in different stages of development. mAbs are considered to be large molecule therapeutics (in general, they are 2-3 orders of magnitude larger than small chemical molecule therapeutics), but they are not just big chemicals. These compounds demonstrate much more complex pharmacokinetic and pharmacodynamic behaviour than small molecules. Because of their large size and relatively poor membrane permeability and instability in the conditions of the gastrointestinal tract, parenteral administration is the most usual route of administration. The rate and extent of mAb distribution is very slow and depends on extravasation in tissue, distribution within the particular tissue, and degradation. Elimination primarily happens via catabolism to peptides and amino acids. Although not definitive, work has been published to define the human tissues mainly involved in the elimination of mAbs, and it seems that many cells throughout the body are involved. mAbs can be targeted against many soluble or membrane-bound targets, thus these compounds may act by a variety of mechanisms to achieve their pharmacological effect. mAbs targeting soluble antigen generally exhibit linear elimination, whereas those targeting membrane-bound antigen often exhibit non-linear elimination, mainly due to target-mediated drug disposition (TMDD). The high-affinity interaction of mAbs and their derivatives with the pharmacological target can often result in non-linear pharmacokinetics. Because of species differences (particularly due to differences in target affinity and abundance) in the pharmacokinetics and pharmacodynamics of mAbs, pharmacokinetic/pharmacodynamic modelling of mAbs has been used routinely to expedite the development of mAbs and their derivatives and has been utilized to help in the selection of appropriate dose regimens. Although modelling approaches have helped to explain variability in both pharmacokinetic and pharmacodynamic properties of these drugs, there is a clear need for more complex models to improve understanding of pharmacokinetic processes and pharmacodynamic interactions of mAbs with the immune system. There are different approaches applied to physiologically based pharmacokinetic (PBPK) modelling of mAbs and important differences between the models developed. Some key additional features that need to be accounted for in PBPK models of mAbs are neonatal Fc receptor (FcRn; an important salvage mechanism for antibodies) binding, TMDD and lymph flow. Several models have been described incorporating some or all of these features and the use of PBPK models are expected to expand over the next few years.
Collapse
|
96
|
Langereis S, Geelen T, Grüll H, Strijkers GJ, Nicolay K. Paramagnetic liposomes for molecular MRI and MRI-guided drug delivery. NMR IN BIOMEDICINE 2013; 26:728-44. [PMID: 23703874 DOI: 10.1002/nbm.2971] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 05/07/2023]
Abstract
Liposomes are a versatile class of nanoparticles with tunable properties, and multiple liposomal drug formulations have been clinically approved for cancer treatment. In recent years, an extensive library of gadolinium (Gd)-containing liposomal MRI contrast agents has been developed for molecular and cellular imaging of disease-specific markers and for image-guided drug delivery. This review discusses the advances in the development and novel applications of paramagnetic liposomes in molecular and cellular imaging, and in image-guided drug delivery. A high targeting specificity has been achieved in vitro using ligand-conjugated paramagnetic liposomes. On targeting of internalizing cell receptors, the effective longitudinal relaxivity r1 of paramagnetic liposomes is modulated by compartmentalization effects. This provides unique opportunities to monitor the biological fate of liposomes. In vivo contrast-enhanced MRI studies with nontargeted liposomes have shown the extravasation of liposomes in diseases associated with endothelial dysfunction, such as tumors and myocardial infarction. The in vivo use of targeted paramagnetic liposomes has facilitated the specific imaging of pathophysiological processes, such as angiogenesis and inflammation. Paramagnetic liposomes loaded with drugs have been utilized for therapeutic interventions. MR image-guided drug delivery using such liposomes allows the visualization and quantification of local drug delivery.
Collapse
Affiliation(s)
- Sander Langereis
- Department of Minimally Invasive Healthcare, Philips Research Eindhoven, Eindhoven, the Netherlands
| | | | | | | | | |
Collapse
|
97
|
Sen K, Mandal M. Second generation liposomal cancer therapeutics: transition from laboratory to clinic. Int J Pharm 2013; 448:28-43. [PMID: 23500602 DOI: 10.1016/j.ijpharm.2013.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 03/03/2013] [Accepted: 03/06/2013] [Indexed: 01/10/2023]
Abstract
Recent innovations and developments in nanotechnology have revolutionized cancer therapeutics. Engineered nanomaterials are the current workhorses in the emerging field of cancer nano-therapeutics. Lipid vesicles bearing anti-tumor drugs have turned out to be a clinically feasible and promising nano-therapeutic approach to treat cancer. Efficient entrapment of therapeutics, biocompatibility, biodegradability, low systemic toxicity, low immunogenicity and ability to bypass multidrug resistance mechanisms has made liposomes a versatile drug/gene delivery system in cancer chemotherapy. The present review attempts to explore the recent key advances in liposomal research and the vast arsenal of liposomal formulations currently being utilized in treatment and diagnosis of cancer.
Collapse
Affiliation(s)
- Kacoli Sen
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | | |
Collapse
|
98
|
Baskar S, Muthusamy N. Antibody-based therapeutics for the treatment of human B cell malignancies. Curr Allergy Asthma Rep 2013; 13:33-43. [PMID: 23229130 PMCID: PMC3674564 DOI: 10.1007/s11882-012-0327-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The dynamic expression of various phenotypic markers during B cell development not only defines the particular stage in ontogeny but also provides the necessary growth, differentiation, maturation and survival signals. When a B cell at any given stage becomes cancerous, these cell surface molecules, intracellular signaling molecules, and the over-expressed gene products become favorite targets for potential therapeutic intervention. Various adaptive and adoptive immunotherapeutic approaches induce T cell and antibody responses against cancer cells, and successful remission leading to minimal residual disease has been obtained. Nonetheless, subsequent relapse and development of resistant clones prompted further development and several novel strategies are evolving. Engineered monoclonal antibodies with high affinity and specificity to target antigens have been developed and used either alone or in combination with chemotherapeutic drugs. They are also used as vehicles to deliver cytotoxic drugs, toxins, or radionuclides that are either directly conjugated or encapsulated in liposomal vesicles. Likewise, genetically engineered T cells bearing chimeric antigen receptors are used to redirect cytotoxicity to antigen-positive target cells. This review describes recent advancements in some of these adoptive immunotherapeutic strategies targeting B cell malignancies.
Collapse
Affiliation(s)
- Sivasubramanian Baskar
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10/CRC; Room 3E-3248, Bethesda, MD 20892, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine; Molecular Virology, Immunology and Medical Genetics and Veterinary BioSciences, The OSU Comprehensive Cancer Center, The Ohio State University, 455E, OSUCCC, 410, West 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
99
|
Devadasu VR, Bhardwaj V, Kumar MNVR. Can controversial nanotechnology promise drug delivery? Chem Rev 2012; 113:1686-735. [PMID: 23276295 DOI: 10.1021/cr300047q] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Venkat Ratnam Devadasu
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | | | | |
Collapse
|
100
|
Penel N, Adenis A, Bonneterre J. Role of the investigator in phase 1 trials of anticancer drugs. Lancet Oncol 2012; 13:1177-9. [DOI: 10.1016/s1470-2045(12)70502-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|