ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Accelerating the Translation of Nanomaterials in Biomedicine

View Author Information
Center for Bioengineering, Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
§ National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077
Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, California 90095, United States
# Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
Departments of Materials Engineering and Bioengineering, University of Tokyo, Tokyo 113-8654, Japan
Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
Laboratory for Nanoengineering & Drug Delivery, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
Department of Materials, Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
School of Chemical and Biomedical Engineering and School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798
The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
†† Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
‡‡ Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27695, United States
Cite this: ACS Nano 2015, 9, 7, 6644–6654
Publication Date (Web):June 26, 2015
https://doi.org/10.1021/acsnano.5b03569

Copyright © 2015 American Chemical Society. This publication is available under these Terms of Use.

  • Free to Read

Article Views

10116

Altmetric

-

Citations

264
LEARN ABOUT THESE METRICS
PDF (6 MB)

Abstract

Due to their size and tailorable physicochemical properties, nanomaterials are an emerging class of structures utilized in biomedical applications. There are now many prominent examples of nanomaterials being used to improve human health, in areas ranging from imaging and diagnostics to therapeutics and regenerative medicine. An overview of these examples reveals several common areas of synergy and future challenges. This Nano Focus discusses the current status and future potential of promising nanomaterials and their translation from the laboratory to the clinic, by highlighting a handful of successful examples.

This publication is licensed for personal use by The American Chemical Society.

Advances in medicine in the areas of genomics, proteomics, tissue engineering, and regenerative medicine are occurring at a rate that was previously unthinkable. The development of new materials resulting from these breakthroughs, such as those that can be used to replace blood vessels, to promote tissue growth, to monitor blood glucose levels, or to improve the bioavailability of drugs, has been equally rapid and diverse. (1, 2) One of the most exciting frontiers is the development of nanomaterials for biomedical applications. (3, 4) Nanomaterials have size-, shape-, and composition-dependent physical, chemical, optical, and electronic properties, among others, that can be designed and tuned, and they are showing great promise for the diagnosis, treatment, monitoring, and control of disease. (5-8) A recent survey found that more than 247 nanomaterial-based medical products have been approved by the Food and Drug Administration (FDA) and are currently in various stages of clinical study. (9) Their intended uses range from the treatment of clinically unresectable cancers to the preparation of antibacterial hand gels to the regeneration of heart tissue. At the same time, common themes emerge when discussing nanomaterials in medicine. Indeed, one of the biggest issues is how to translate nanomaterials from the laboratory to the clinic effectively.

In this Nano Focus, we highlight these discussions, which fall into three categories: nanotherapeutics, imaging and diagnostics, and tissue regeneration.

At the 2015 Materials Research Society (MRS) Spring Meeting in San Francisco (April 6–10), a special symposium focusing on translational research involving nanomaterials in the biomedical field was held. Researchers from all over the world gathered to exchange ideas and to discuss criteria for the selection, development, synthesis, and utilization of nanomaterials. In this Nano Focus, we highlight these discussions, which fall into three categories: nanotherapeutics, imaging and diagnostics, and tissue regeneration.

Enhancing Efficacy in Nanotherapeutics

Among the numerous nanomaterials explored in therapeutic applications, those often found in clinical trials are gold nanoparticles, polymeric nanoparticles, liposomes, and carbon-based nanomaterials. By sharing expertise across fields, researchers can accelerate the utilization of nanomaterials in addressing the challenges faced by traditional therapeutic agents.
Since the development of gold-nanoparticle-based spherical nucleic acids (SNAs) in 1996, (10) the Mirkin group at Northwestern University has exploited the properties of this class of nanostructures in many areas of biomedicine. Spherical nucleic acids typically consist of a nanoparticle core and a highly oriented and densely packed shell of oligonucleotides. (11) The properties of SNAs emerge from the orientation and arrangement of oligonucleotides on the surfaces of these particles. For example, SNAs are taken up into many different cell types (over 60 tested to date) at high levels and rates without the need for transfection agents, (12) have high affinities for nucleic acid targets (100 times greater than linear DNA of the same sequence), (13) and cross both the blood–brain barrier (14) and the epidermis (15) to reach difficult-to-target tissues in therapeutic applications. Success in the utilization of SNAs for intracellular mRNA detection has led to the commercialization of NanoFlare technology (16, 17) under the trade name SmartFlares (Merck Millipore in partnership with AuraSense, LLC, Skokie, Illinois), and SNAs have also been applied as agents in gene regulation as therapeutics for a host of cancers, including glioblastoma multiforme (an aggressive form of brain cancer), and skin disorders, among others. The Mirkin group has made progress in using SNAs as immunomodulatory agents. (18) Spherical nucleic acids functionalized with oligonucleotides displaying toll-like receptor (TLR)-agonist or TLR-antagonist activity were shown to be capable of either stimulating or modulating the activity of the immune system, respectively. Such structures do so with potencies up to several orders of magnitude higher than the conventional linear nucleic acids from which they are composed. This discovery paves the way for the development of SNAs as therapeutic cancer vaccines. Mirkin’s demonstration of the ability of SNAs to reduce tumor burden and to enhance survival in vivo, in mouse models of lymphoma, proved that immunomodulatory SNAs can be directed to activate the response of the immune system to destroy tumors in an antigen-specific manner (Figure 1).

Figure 1

Figure 1. In vivo activity of immunomodulatory SNAs as cancer vaccines.

A fundamental challenge is the delivery of therapeutic molecules inside target cells in the body. Nanoparticles have shown immense promise as vehicles for intracellular delivery, with proof-of-principle experiments in humans being completed with small interfering RNA (siRNA). (19) However, there are many barriers to achieving safe and effective delivery systems (Figure 2), and potential delivery systems must have multiple functionalities to allow in vivo delivery, making the design criteria difficult to define for nanoparticles capable of accomplishing intracellular delivery. To accelerate the design and discovery process, the Anderson group at the Massachusetts Institute of Technology has pioneered combinatorial methods for nanoparticulate drug delivery. Combinatorial chemical methods have been developed to enable the rapid synthesis and characterization of a range of nanoformulations based on biodegradable polymers, lipid-like materials, and other materials. (20-22) These have generated new formulations with particular promise as delivery vehicles for RNA and other nucleic acids; (23) these formulations have the potential to be used as therapies for many diseases, including cancer. (24)
Furthermore, Shi at Brigham and Women’s Hospital/Harvard Medical School described the rational design and development of lipid–polymer hybrid nanoparticle platforms to address the bottlenecks faced in the delivery of RNA interference (RNAi) therapeutics, such as siRNA. (25-27) Specifically, the clinical applications of RNAi in cancer therapy are currently hindered by the challenge of achieving the effective systemic in vivo delivery of siRNA to tumors. Multiple physiological barriers, such as enzymatic degradation, rapid elimination by renal excretion or the mononuclear phagocyte system, and poor cellular uptake and endosomal escape, must be overcome. By utilizing hybrid nanoparticles, Shi achieved sustained gene silencing, and prolonged circulation of siRNA in the blood for high tumor extravasation and accumulation. The successful application of these RNAi nanoparticles to validate the therapeutic role of Prohibitin1 in non-small cell lung cancer treatment (27) indicates the significant potential of this platform for the validation of many other potential cancer targets and for the clinical development of novel cancer therapies.

Figure 2

Figure 2. Barriers to successful in vivo delivery of nucleic acids using nonviral vectors. Reprinted with permission from ref 19. Copyright 2014 Macmillan Publishers Ltd.

Kataoka at the University of Tokyo has pioneered the synthesis and development of “polymeric micelle drugs”, which have proven useful for targeting a variety of drugs to tissues and organs and to tumors in particular. There has been significant recent progress in the clinical development of polymeric micelles loaded with a variety of cytotoxic reagents. Notably, five different micellar formulations have already been explored in clinical trials in Asia and the United States. (28) A version loaded with paclitaxel is in the final stage of a Phase III clinical trial in Japan for the treatment of recurrent breast cancer, and it is expected to proceed into the application for approval within a year. More recently, Kataoka and colleagues have been active in developing a second generation system of polymeric micelles installed with ligand moieties at their peripheries. Particularly, cRGD-conjugated polymeric micelles were able to cross the blood–brain tumor barrier via a transcytosis mechanism, achieving high efficacy in treating intractable orthotopic glioblastoma in animal experiments. (29) Antibody fragments can also be conjugated to polymeric micelles. In this way, higher drug payloads can be achieved without antibody precipitation or impaired binding compared to when drugs are directly conjugated to antibody molecules using more conventional approaches. (30)
In 1989, the Kabanov group at the University of North Carolina at Chapel Hill investigated the use of polymeric micelles for targeted drug delivery. (31) They discovered that Pluronic block copolymers can be used in the sensitization of multi-drug-resistant (MDR) cancer and cancer stem cells and elucidated the mechanisms responsible for these effects. (32) This research led to first-in-man polymeric micelle drug candidates for the treatment of cancer (SP1049C) that show high efficacy against advanced esophageal cancer in Phase II trials. (33) Recently, they discovered polymeric micelles based on amphiphilic poly(2-oxazoline) blocks with unprecedented high capacities for poorly soluble, uncharged drugs (e.g., taxanes) and drug combinations, enabling increasing therapeutic indices compared to current drug formulations. (34, 35)
Liposomes are another popular nanomaterial used in preclinical and clinical studies, but it is difficult to sustain release from these structures for more than a few days. The Venkatraman group at Nanyang Technological University has developed a subconjunctivally injected nanoliposome drug delivery system for the long-term (3–5 months) delivery of latanoprost that can be used in glaucoma treatment, which went from concept to clinic in less than 5 years (Figure 3). (36, 37) Glaucoma is a chronic progressive optic neuropathy that is characterized by optic nerve changes and visual field loss. Elevated intraocular pressure (IOP) is the main modifiable risk factor. The chronic instillation of daily eye drops to lower IOP is the primary treatment of choice, although this regimen requires patient adherence and correct performance. Hence, a sustained-delivery system would be a big boon to patients with glaucoma. Venkatraman’s group also explored the nanoparticle-mediated sustained delivery of siRNA for preventing fibrosis following surgery. (38) This method has applications in ocular and other types of surgeries/implantations. They have shown sustained efficacy of action with a siRNA-incorporated nanoparticle. As shown in the above two examples, the premise of their work rests on the ability of nanoparticles to sustain drug/protein/siRNA release.

Figure 3

Figure 3. Subconjunctival instillation of nanocarriers incorporating latanoprost lowers eye pressure in glaucoma patients for up to 3 months. Reproduced from ref 37. Copyright 2014 American Chemical Society.

The Chow and Ho groups at the National University of Singapore and the University of California, Los Angeles (UCLA), respectively, study nanodiamonds, which are an emerging class of carbon-based nanomaterials, due to their advantageous surface characteristics. Nanodiamond facets mediate electrostatic properties that have resulted in potent and scalable anthracycline drug binding as well as marked enhancements in magnetic resonance imaging (MRI) efficiency. (39, 40) With regard to drug delivery, nanodiamond–doxorubicin compounds (NDX) were administered to treat drug-resistant breast and liver tumors in mouse models. This study demonstrated the nanodiamond-mediated improvement of drug tolerance; lethal dosages of doxorubicin delivered as NDX resulted in the smallest tumors observed (compared to saline controls and unmodified drug administration), and all of the treated mice survived the full duration of the study. (41) The active targeting of triple-negative breast tumors in vivo using nanodiamond–epirubicin complexes functionalized with the epidermal growth factor receptor (EGFR) antibody resulted in complete tumor regression. (42) More recently, nanodiamond–anthracycline agents have been used to treat hepatic cancer stem cells. (43)
Toward the further enhancement of the potency and safety of cancer therapies, a prevalent challenge in the field of nanomedicine is the ability to move beyond monotherapies toward combinatorial cancer treatments. Using phenotypic instead of genotypic profiling to drive combinatorial optimization, Ho and colleagues developed a powerful mechanism-independent engineering optimization platform, termed Feedback System Control.II (FSC.II), to identify globally optimized nanodiamond–anthracycline drug combinations rapidly. (44) FSC.II does not require the use of feedback, and instead, it utilizes a selected set of experimental validation assays to formulate phenotypic profiles from which drug combinations can rapidly be pin-pointed.
Mitragotri at the University of California, Santa Barbara, also translated nanomaterials to clinical applications (Figure 4). Specifically, he used gold-coated silica nanoparticles for the treatment of acne. These 150 nm, poly(ethylene glycol)-coated, silica–gold nanoparticles are designed to absorb near-infrared light and produce localized heating. The delivery of these nanoparticles into skin is a major hurdle due to the skin’s barrier properties. Mitragotri and colleagues showed that these nanoparticles can be delivered deep into the skin’s sebaceous glands using low-frequency ultrasound. (45) Ultrasound induces cavitation on the surface of the skin, which produces microjets and shock waves that open transport pathways into the glands. (46) Once delivered deep into the glands, the thermal activation of the nanoparticles using near-infrared light caused thermolysis and the inactivation of overactive sebaceous glands, the underlying pathology of acne. This nanoparticle-based technology provides several advantages over standard treatments for acne; for instance, systemic side effects are avoided with this treatment.

Figure 4

Figure 4. Schematic for the nanoparticle-based treatment of acne. Silica–gold nanoparticles are delivered into sebaceous glands using low-frequency ultrasound. Nanoparticles are then activated using near-infrared light to induce thermolysis. Reprinted with permission from ref 45. Copyright 2015 Elsevier.

The Stevens group at the Imperial College of London and collaborators at the Houston Methodist Research Institute have recently reported engineering a platform of mesoporous silicon nanoneedles for the delivery of nanoparticle and other therapeutic payloads to cells and tissues (Figure 5A,B). (47, 48) This technology could prove transformative in the fields of drug delivery, regenerative medicine, and biosensing. The dynamics of the nanoneedle entry to the cell and study of the nanoneedle–cell interface have been elucidated and pave the way for highly controlled delivery of a range of nanoparticle payloads intracellularly. (47) Furthermore, the nanoneedle array can simultaneously deliver both DNA and siRNA with high efficiency (over 90%) and in vivo proved successful in upregulating blood vessel formation in muscle by delivery of the VEGF-165 gene (Figure 5C–E). (48) The Stevens group has also developed several other notable nanomaterials-based technologies, particularly enzyme-response nanoparticle systems that have a wide range of important impacts in the field of biosensing. (8, 49, 50)

Figure 5

Figure 5. Nanoneedle for the delivery of VEGF-165 to upregulate blood vessel formation in muscle. (A) Scanning electron microscope (SEM) micrographs showing the morphology of porous silicon nanoneedle arrays with pitches of 2 μm. Scale bars, 2 μm. (B) High-resolution SEM micrographs of nanoneedle tips showing the nanoneedles’ porous structure and the tunability of tip diameter from less than 100 nm to over 400 nm. Scale bars, 200 nm. (C) Intravital bright-field (top) and confocal (bottom) microscopy images of the vasculature of untreated (left) and hVEGF-165-treated muscles with either direct injection (center) or nanoinjection (right). The fluorescence signal originates from systemically injected FITC–dextran. Scale bars, bright-field 100 μm; confocal 50 μm. (D,E) Quantification of the fraction of fluorescent signal (dextran) (D) and the number of nodes in the vasculature per mm2 (E) within each field of view acquired for untreated control, intramuscular injection (IM) and nanoinjection. *p = 0.05, **p < 0.01, ***p < 0.001. Error bars represent the SD of the averages of five areas taken from three animals. Reprinted with permission of figure and caption text from ref 48. Copyright 2015 Macmillan Publishers Ltd.

From the above discussion of a variety of nanomaterials as nanotherapeutic agents for enhancing treatment efficacy, we conclude that successful translation of nanomaterials relies on the identification of a clinical problem and innovative ideas to solve it through rational design. In addition, more and more versatile nanomaterials are being exploited in emerging research themes, areas such as cancer vaccines (51) and genome editing. (19)

Imaging and Diagnostics with High Sensitivity and Selectivity

Besides their use as therapeutic agents, nanomaterials are also being used for imaging and diagnostics purposes. The most well-known examples include silica nanoparticles, quantum dots, magnetic nanoparticles, and microbubbles. These nanostructures have been used to detect small molecules (like H2O2), cells including circulating tumor cells (CTCs), and tumor tissues.

Successful translation of nanomaterials relies on the identification of a clinical problem and innovative ideas to solve it through rational design

The Sun group at Brown University is interested in monitoring cellular H2O2, which is an important reactive oxygen species generated via oxygen metabolism; it is actively involved in cell signaling and cell growth. (52) Unfortunately, its uncontrolled overproduction can cause the detrimental oxidation of biomolecules and lead to aging, cancer, and other diseases. (53) Sun recently developed dumbbell (Au–Fe3O4 and PdPt–Fe3O4) and core/shell (Au/MnO) magnetic nanoparticles as sensitive probes for H2O2 detection. (54, 55) Dumbbell magnetic nanoparticles were prepared by the controlled nucleation and growth of Fe3O4 on presynthesized noble metal nanoparticles, while the core/shell Au/MnO nanoparticles were made by the controlled oxidation of AuMn alloy nanoparticles. Both dumbbell and core/shell nanoparticles are active for the electrochemical reduction of H2O2 with detection limits reaching as low as 5 nM. Highly sensitive electrochemical sensors have been used to monitor H2O2 concentration levels released from living cells; tumorigenic cells were found to have higher levels of H2O2 than nontumorigenic ones. These composite nanoparticle probes can be used in high-sensitivity cancer detection schemes and may also help to increase the efficacy of cancer therapies.
The Chen group at the National Institute of Biomedical Imaging and Bioengineering of the National Institutes of Health uses nanomaterials as platforms to provide imaging contrast in positron emission tomography (PET). In medical imaging, PET can provide a direct, highly sensitive, and quantitative readout of organ/tissue targeting efficiency and pharmacokinetics. Compared with radiolabeled antibodies, proteins, peptides, and other biologically relevant molecules, radiolabeled nanoparticles represent a new frontier in molecular imaging probe design because they can combine different imaging modalities and targeting ligands in a single vector, synergistically improving the imaging quality. (56) However, the applications of radiolabeled nanoparticles are based on the premise that the radioisotopes are stably attached to the nanomaterials. Chen has developed general rules for selecting appropriate isotopes for given types of nanoparticles as well as adjusting the labeling reaction according to specific applications. The stability (colloidal and radiochemical) of the radiolabeled nanoparticles as well as their biological fate must be assessed; special attention should be paid to labeling strategies as they affect the stability of radiolabeled nanoparticles and might cause discrepancies in the interpretation of PET data (owing to the distribution of nanoparticles).
Wang’s group at the Chinese Academy of Sciences is interested in creating nano–bio interfaces with controllable adhesion properties. The cell-adhesive biointerfaces are based on the cooperative effects of multiscale structural matching and molecular recognition. (57) They explore the relationship between cell-specific adhesion and surface structure (with nanowires, (21) nanofibers, (58) nanofractals, (59) and soft nanotubes (60)). Also, they developed a series of biointerfaces with specific recognition and stimuli-responsive capture and release properties (i.e., temperature, (61) electric, (59) enzymatic, (21) and pH (62)). They have made progress in the isolation of viable rare CTCs from the blood via their designed cell-adhesive biointerfaces and developed adhesion-based CTC isolation approaches as cancer diagnostics with high efficiencies (>97%). (63) In particular, these biointerfaces with controlled cell adhesion are capable of capturing rare viable CTCs for early cancer detection and the monitoring of cancer therapy, single-cell gene analysis, (64) and other purposes.

Biomimetic and Bio-Inspired Scaffolds for Tissue Engineering

Nanomaterials for tissue engineering involve a broad spectrum of nanoscale formulations and structures developed to mimic tissue complexity and to modulate cellular function to yield therapeutic benefits.
The Xia group at the Georgia Institute of Technology and Emory University has been developing practical nanomaterials for medical applications. They use electrospun nanofibers in tissue engineering. (65) Electrospinning has been widely explored to process polymeric materials into nanofibers with tunable and controllable compositions, diameters, porosities, and surface properties. Owing to its small feature size, high porosity, and large surface area, a nonwoven mat of electrospun fibers can serve as a superb scaffold that mimics the extracellular matrix (ECM), which is critical to cell attachment and spreading. The nanofibers themselves can also be functionalized through the encapsulation or attachment of bioactive species, such as ECM proteins, enzymes, and growth factors. In addition, the nanofibers can be readily assembled into a wide variety of arrays or hierarchical structures by manipulating their alignment, stacking, or folding. All these attributes make electrospinning a powerful tool for generating nanostructured materials for a broad range of biomedical applications, including controlled release, drug delivery, and tissue engineering. Xia highlighted the use of aligned nanofibers to control the differentiation of embryonic stem cells into different types of neural lineages and to guide the outgrowth of neurites for peripheral nerve repair. (66) He also pointed out that nanofiber scaffolds could be designed for repairing injuries to the flexor tendon and the tendon-to-bone insertion site; (67) they could also be used as wound dressings for brain surgery. (68)
Karp from Brigham and Women’s Hospital suggests that different approaches are required for solving medical problems versus solving basic science problems. He asserts that one must develop design criteria relevant to solving the problem in animal models, while considering the multiple steps required to bring a technology from the laboratory to the clinic. One must think through scale-up, manufacturing, regulatory issues, and patent strategy and then impose these criteria to advance toward a potential solution. In particular, turning to nature for solutions can aid in the problem-solving process, recognizing that everything living has overcome challenges, and thus we are surrounded by solutions. Through elucidating mechanisms behind these solutions with state-of-the-art tools, he asserts that we can identify ideas to help overcome even the most challenging of problems. Karp has created tissue adhesives using inspiration from slugs and snails, (69) spiny-headed worms, (70) porcupine quills, (71) and spider webs. One of the adhesive technologies led to the formation of a company, Gecko Biomedical (Paris, France), and is on track to be tested for vascular graft applications in humans in late 2015. Of note is that, in these developments, failure should be embraced as part of the problem-solving process and is likely a prerequisite to success. In this work, it was important to build highly functional and multidisciplinary teams that know what resources are available in their environment and how to access them.
Teoh from Nanyang Technological University is interested in processing biomaterials in an environmentally friendly way. (72, 73) To this end, his group has unearthed a solvent-free approach known as cryomilling, where biomaterials are processed at near cryogenic temperatures. Polymer particle size reduction has been achieved due to the high-energy collision process, which occurs below the glass transition temperature. Particle sizes may be reduced to the nanoscale, where surface thermodynamics play critical roles in determining their behavior, particularly the interactions between two chemically distinct phases, as in the case of composite biomaterials. Notably, there is also a significant challenge in obtaining reproducible, well-distributed composites. They have demonstrated that a variety of composite biomaterials that incorporate second phases, such as inorganic elements, trace elements, and even drugs, may be processed efficiently via cryomilling. (73) This technique also addresses second phase distribution issues that plague many other processing techniques, such as solvent-casting, electrospinning, and melt extrusion.

Future Outlook

ARTICLE SECTIONS
Jump To

Tremendously exciting advances in nanomaterials-based medical treatments, from cancer therapies, diabetes administration, and vaccine development to molecular diagnostics, tissue repair, and regeneration, are both underway and yet to come. The confluence of nanomaterials and emerging biomedical science provides vast commercial opportunities. While fundamental science has been fueled with numerous innovations over the past decade, as evidenced by the number of nanomaterial-associated formulations and devices in clinical development, the number of marketed products is still small, compared to traditional medications. Remaining challenges include effectively improving efficacy, while minimizing potential concerns, through the rational design and thorough evaluation of nanomaterials.

The confluence of nanomaterials and emerging biomedical science provides vast commercial opportunities.

In oncology nanomedicine, for example, the acceleration of the translation process relies profoundly on a thorough understanding of how nanocarriers interact with the physiological environment. In addition to general evaluations based on the enhanced permeability and retention effect, biodistribution, and clearance mechanisms, more precise details should be taken into account, such as how the particles pass through the tumor microenvironment and enter cells to reach active sites. (74, 75, 76) Moreover, in designing stimuli-responsive or programmable nanocarriers, (77, 78) a current theme in nanomedicine, closer investigations of the dynamic relationships between the phase transitions of materials and the relevant gradients in the biological environment, such as pH, redox, glucose, ATP, and enzyme activity, should enable more precise targeting and release. (79-82)
Second, a major stumbling block in the translation of nanomaterials for biomedical applications is safety concerns, especially for invasive administration. On one hand, clinical use requires the careful, prolonged evaluation of the local and systemic toxicity of nanomaterials as well as their potential immunogenicity. (83) On the other hand, there is an urgent need to invent and to tailor new materials with excellent biocompatibility. Ideas inspired by nature, mimicking the structures and composites of natural particles, including viruses, vesicles, and cells, have attracted increasing interest and brought promising outcomes.
Third, regarding the rational design of nanomaterials with specific physicochemical properties (84) for clinical applications, it is important to set uniformity in preclinical trials. Variability in particle size, surface properties, and stability as well as differences in cell lines, tumor properties, therapeutic doses, and pharmacokinetics/pharmacodynamics analysis have prevented the systematic comparison of relevant nanomaterials and have been an impediment to creating design rules for optimizing a specific formulation or device.
Last but not least, the design, development, and ultimately commercialization of clinically used nanomaterials require seamless collaboration and commitment between a broad range of research investigators, investors, and regulatory authorities. Key to these activities are platforms for fusing ideas to shepherd emerging technologies further along safe and effective pipelines.
Overall, given the progress that has been made so far, we are optimistic that nanomaterials-based clinical development will continue to be exciting, with growing numbers of innovations as well as those currently garnering FDA approval entering the clinic soon. We hope the ideas and concepts presented in this Nano Focus will be useful in the development of “ideal” nanomaterials features, the expansion of design criteria, and the enlightenment of research opportunities for evolving the next generations of biomedical materials. (85, 86, 87)

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Authors
    • Samir Mitragotri - Center for Bioengineering, Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States Email: [email protected] [email protected] [email protected]
    • Zhen Gu - Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27695, United States Email: [email protected] [email protected] [email protected]
    • Chenjie Xu - Center for Bioengineering, Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United StatesDepartment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United StatesNational Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United StatesCancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, California 90095, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United StatesDepartments of Materials Engineering and Bioengineering, University of Tokyo, Tokyo 113-8654, JapanDepartment of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United StatesLaboratory for Nanoengineering & Drug Delivery, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United StatesDepartment of Materials, Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States▽School of Chemical and Biomedical Engineering and ▲School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United StatesTechnical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, ChinaJoint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27695, United States Email: [email protected] [email protected] [email protected]
  • Authors
    • Daniel G. Anderson - Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
    • Xiaoyuan Chen - National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
    • Edward K. Chow - Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077
    • Dean Ho - Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, California 90095, United States
    • Alexander V. Kabanov - Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
    • Jeffrey M. Karp - Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
    • Kazunori Kataoka - Departments of Materials Engineering and Bioengineering, University of Tokyo, Tokyo 113-8654, Japan
    • Chad A. Mirkin - Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
    • Sarah Hurst Petrosko - Department of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
    • Jinjun Shi - Laboratory for Nanoengineering & Drug Delivery, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
    • Molly M. Stevens - Department of Materials, Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.
    • Shouheng Sun - Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
    • Sweehin Teoh - Center for Bioengineering, Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United StatesDepartment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United StatesNational Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United StatesCancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, California 90095, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United StatesDepartments of Materials Engineering and Bioengineering, University of Tokyo, Tokyo 113-8654, JapanDepartment of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United StatesLaboratory for Nanoengineering & Drug Delivery, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United StatesDepartment of Materials, Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States▽School of Chemical and Biomedical Engineering and ▲School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United StatesTechnical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, ChinaJoint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27695, United States
    • Subbu S. Venkatraman - Center for Bioengineering, Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United StatesDepartment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United StatesNational Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United StatesCancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, California 90095, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United StatesDepartments of Materials Engineering and Bioengineering, University of Tokyo, Tokyo 113-8654, JapanDepartment of Chemistry and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United StatesLaboratory for Nanoengineering & Drug Delivery, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United StatesDepartment of Materials, Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, U.K.Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States▽School of Chemical and Biomedical Engineering and ▲School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United StatesTechnical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, ChinaJoint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United StatesDivision of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United StatesDepartment of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27695, United States
    • Younan Xia - The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
    • Shutao Wang - Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
  • Notes
    The authors declare the following competing financial interest(s): S.M. is a Scientific Advisor to Sebacia Inc. J.M.K. is part owner of and consultant to Gecko Biomedical.

References

ARTICLE SECTIONS
Jump To

This article references 87 other publications.

  1. 1
    Ratner, B. D.; Hoffman, A. S.; Schoen, F. J.; Lemons, J. E. Biomaterials Science: An Introduction to Materials in Medicine; Elsevier Academic Press: Amsterdam, 2004.
  2. 2
    Lee, S.; Henthorn, D. Materials in Biology and Medicine; CRC Press/Taylor & Francis Group: Boca Raton, FL, 2012.
  3. 3
    Langer, R. Drug Delivery and Targeting Nature 1998, 392, 5 10
  4. 4
    Farokhzad, O. C.; Langer, R. Nanomedicine: Developing Smarter Therapeutic and Diagnostic Modalities Adv. Drug Delivery Rev. 2006, 58, 1456 1459
  5. 5
    Doshi, N.; Mitragotri, S. Designer Biomaterials for Nanomedicine Adv. Funct. Mater. 2009, 19, 3843 3854
  6. 6
    Mitragotri, S.; Lahann, J. Physical Approaches to Biomaterial Design Nat. Mater. 2009, 8, 15 23
  7. 7
    Dobrovolskaia, M. A.; McNeil, S. E. Immunological Properties of Engineered Nanomaterials Nat. Nanotechnol. 2007, 2, 469 478
  8. 8
    Howes, P. D.; Chandrawati, R.; Stevens, M. M. Colloidal Nanoparticles as Advanced Biological Sensors Science 2014, 346, 1247390
  9. 9
    Etheridge, M. L.; Campbell, S. A.; Erdman, A. G.; Haynes, C. L.; Wolf, S. M.; McCullough, J. The Big Picture on Nanomedicine: The State of Investigational and Approved Nanomedicine Products Nanomedicine 2013, 9, 1 14
  10. 10
    Mirkin, C. A.; Letsinger, R. L.; Mucic, R. C.; Storhoff, J. J. A DNA-Based Method for Rationally Assembling Nanoparticles into Macroscopic Materials Nature 1996, 382, 607 609
  11. 11
    Cutler, J. I.; Auyeung, E.; Mirkin, C. A. Spherical Nucleic Acids J. Am. Chem. Soc. 2012, 134, 1376 1391
  12. 12
    Rosi, N. L.; Giljohann, D. A.; Thaxton, C. S.; Lytton-Jean, A. K.; Han, M. S.; Mirkin, C. A. Oligonucleotide-Modified Gold Nanoparticles for Intracellular Gene Regulation Science 2006, 312, 1027 1030
  13. 13
    Lytton-Jean, A. K.; Mirkin, C. A. A Thermodynamic Investigation into the Binding Properties of DNA Functionalized Gold Nanoparticle Probes and Molecular Fluorophore Probes J. Am. Chem. Soc. 2005, 127, 12754 12755
  14. 14
    Jensen, S. A.; Day, E. S.; Ko, C. H.; Hurley, L. A.; Luciano, J. P.; Kouri, F. M.; Merkel, T. J.; Luthi, A. J.; Patel, P. C.; Cutler, J. I. Spherical Nucleic Acid Nanoparticle Conjugates as an RNAi-Based Therapy for Glioblastoma Sci. Transl. Med. 2013, 5, 209ra152
  15. 15
    Zheng, D.; Giljohann, D. A.; Chen, D. L.; Massich, M. D.; Wang, X.-Q.; Iordanov, H.; Mirkin, C. A.; Paller, A. S. Topical Delivery of siRNA-Based Spherical Nucleic Acid Nanoparticle Conjugates for Gene Regulation Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 11975 11980
  16. 16
    Seferos, D. S.; Giljohann, D. A.; Hill, H. D.; Prigodich, A. E.; Mirkin, C. A. Nano-Flares: Probes for Transfection and mRNA Detection in Living Cells J. Am. Chem. Soc. 2007, 129, 15477 15479
  17. 17
    Halo, T. L.; McMahon, K. M.; Angeloni, N. L.; Xu, Y.; Wang, W.; Chinen, A. B.; Malin, D.; Strekalova, E.; Cryns, V. L.; Cheng, C. NanoFlares for the Detection, Isolation, and Culture of Live Tumor Cells from Human Blood Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 17104 17109
  18. 18
    Radovic-Moreno, A. F.; Chernyak, N.; Mader, C. C.; Nallagatla, S.; Kang, R. S.; Hao, L.; Walker, D. A.; Halo, T. L.; Merkel, T. J.; Rische, C. H. Immunomodulatory Spherical Nucleic Acids Proc. Natl. Acad. Sci. U.S.A. 2015, 112, 3892 3897
  19. 19
    Yin, H.; Kanasty, R. L.; Eltoukhy, A. A.; Vegas, A. J.; Dorkin, J. R.; Anderson, D. G. Non-viral Vectors for Gene-Based Therapy Nat. Rev. Genet. 2014, 15, 541 555
  20. 20
    Sahay, G.; Querbes, W.; Alabi, C.; Eltoukhy, A.; Sarkar, S.; Zurenko, C.; Karagiannis, E.; Love, K.; Chen, D.; Zoncu, R. Efficiency of siRNA Delivery by Lipid Nanoparticles Is Limited by Endocytic Recycling Nat. Biotechnol. 2013, 31, 653 658
  21. 21
    Lovell, J. F.; Jin, C. S.; Huynh, E.; Jin, H.; Kim, C.; Rubinstein, J. L.; Chan, W. C.; Cao, W.; Wang, L. V.; Zheng, G. Porphysome Nanovesicles Generated by Porphyrin Bilayers for Use as Multimodal Biophotonic Contrast Agents Nat. Mater. 2011, 10, 324 332
  22. 22
    Dahlman, J. E.; Barnes, C.; Khan, O. F.; Thiriot, A.; Jhunjunwala, S.; Shaw, T. E.; Xing, Y.; Sager, H. B.; Sahay, G.; Speciner, L. In Vivo Endothelial siRNA Delivery Using Polymeric Nanoparticles with Low Molecular Weight Nat. Nanotechnol. 2014, 9, 648 655
  23. 23
    Dong, Y.; Love, K. T.; Dorkin, J. R.; Sirirungruang, S.; Zhang, Y.; Chen, D.; Bogorad, R. L.; Yin, H.; Chen, Y.; Vegas, A. J. Lipopeptide Nanoparticles for Potent and Selective siRNA Delivery in Rodents and Nonhuman Primates Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 3955 3960
  24. 24
    Yin, H.; Xue, W.; Chen, S.; Bogorad, R. L.; Benedetti, E.; Grompe, M.; Koteliansky, V.; Sharp, P. A.; Jacks, T.; Anderson, D. G. Genome Editing with Cas9 in Adult Mice Corrects a Disease Mutation and Phenotype Nat. Biotechnol. 2014, 32, 551 553
  25. 25
    Shi, J.; Xiao, Z.; Votruba, A. R.; Vilos, C.; Farokhzad, O. C. Differentially Charged Hollow Core/Shell Lipid–Polymer–Lipid Hybrid Nanoparticles for Small Interfering RNA Delivery Angew. Chem., Int. Ed. 2011, 123, 7165 7169
  26. 26
    Shi, J.; Xu, Y.; Xu, X.; Zhu, X.; Pridgen, E.; Wu, J.; Votruba, A. R.; Swami, A.; Zetter, B. R.; Farokhzad, O. C. Hybrid Lipid–Polymer Nanoparticles for Sustained siRNA Delivery and Gene Silencing Nanomedicine 2014, 10, 897 900
  27. 27
    Zhu, X.; Xu, Y.; Solis, L. M.; Tao, W.; Wang, L.; Behrens, C.; Xu, X.; Zhao, L.; Liu, D.; Wu, J. Long-Circulating siRNA Nanoparticles for Validating Prohibitin1-Targeted Non-Small Cell Lung Cancer Treatment Proc. Natl. Acad. Sci. U.S.A. 2015,  DOI: 10.1073/pnas.1505629112
  28. 28
    Cabral, H.; Kataoka, K. Progress of Drug-Loaded Polymeric Micelles into Clinical Studies J. Controlled Release 2014, 190, 465 476
  29. 29
    Miura, Y.; Takenaka, T.; Toh, K.; Wu, S.; Nishihara, H.; Kano, M. R.; Ino, Y.; Nomoto, T.; Matsumoto, Y.; Koyama, H. Cyclic RGD-Linked Polymeric Micelles for Targeted Delivery of Platinum Anticancer Drugs to Glioblastoma through the Blood–Brain Tumor Barrier ACS Nano 2013, 7, 8583 8592
  30. 30
    Ahn, J.; Miura, Y.; Yamada, N.; Chida, T.; Liu, X.; Kim, A.; Sato, R.; Tsumura, R.; Koga, Y.; Yasunaga, M. Antibody Fragment-Conjugated Polymeric Micelles Incorporating Platinum Drugs for Targeted Therapy of Pancreatic Cancer Biomaterials 2015, 39, 23 30
  31. 31
    Kabanov, A. V.; Chekhonin, V.; Alakhov, V. Y.; Batrakova, E.; Lebedev, A.; Melik-Nubarov, N.; Arzhakov, S.; Levashov, A.; Morozov, G.; Severin, E. The Neuroleptic Activity of Haloperidol Increases after Its Solubilization in Surfactant Micelles: Micelles as Microcontainers for Drug Targeting FEBS Lett. 1989, 258, 343 345
  32. 32
    Alakhova, D. Y.; Kabanov, A. V. Pluronics and MDR Reversal: An Update Mol. Pharmaceutics 2014, 11, 2566 2578
  33. 33
    Valle, J. W.; Armstrong, A.; Newman, C.; Alakhov, V.; Pietrzynski, G.; Brewer, J.; Campbell, S.; Corrie, P.; Rowinsky, E. K.; Ranson, M. A Phase 2 Study of SP1049C, Doxorubicin in P-Glycoprotein-Targeting Pluronics, in Patients with Advanced Adenocarcinoma of the Esophagus and Gastroesophageal Junction Invest. New Drugs 2011, 29, 1029 1037
  34. 34
    Han, Y.; He, Z.; Schulz, A.; Bronich, T. K.; Jordan, R.; Luxenhofer, R.; Kabanov, A. V. Synergistic Combinations of Multiple Chemotherapeutic Agents in High Capacity Poly(2-oxazoline) Micelles Mol. Pharmaceutics 2012, 9, 2302 2313
  35. 35
    He, Z.; Schulz, A.; Wan, X.; Seitz, J.; Bludau, H.; Alakhova, D. Y.; Darr, D. B.; Perou, C. M.; Jordan, R.; Ojima, I. Poly(2-oxazoline) Based Micelles with High Capacity for 3rd Generation Taxoids: Preparation, In Vitro and In Vivo Evaluation J. Controlled Release 2015, 208, 67 75
  36. 36
    Natarajan, J. V.; Ang, M.; Darwitan, A.; Chattopadhyay, S.; Wong, T. T.; Venkatraman, S. S. Nanomedicine for Glaucoma: Liposomes Provide Sustained Release of Latanoprost in the Eye Int. J. Nanomed. 2012, 7, 123 131
  37. 37
    Natarajan, J. V.; Darwitan, A.; Barathi, V. A.; Ang, M.; Htoon, H. M.; Boey, F.; Tam, K. C.; Wong, T. T.; Venkatraman, S. S. Sustained Drug Release in Nanomedicine: A Long-Acting Nanocarrier-Based Formulation for Glaucoma ACS Nano 2014, 8, 419 429
  38. 38
    Tan, Y. F.; Mundargi, R. C.; Chen, M. H. A.; Lessig, J.; Neu, B.; Venkatraman, S. S.; Wong, T. T. Layer-by-Layer Nanoparticles as an Efficient siRNA Delivery Vehicle for SPARC Silencing Small 2014, 10, 1790 1798
  39. 39
    Huang, H.; Pierstorff, E.; Osawa, E.; Ho, D. Active Nanodiamond Hydrogels for Chemotherapeutic Delivery Nano Lett. 2007, 7, 3305 3314
  40. 40
    Manus, L. M.; Mastarone, D. J.; Waters, E. A.; Zhang, X.-Q.; Schultz-Sikma, E. A.; MacRenaris, K. W.; Ho, D.; Meade, T. J. Gd(III)-Nanodiamond Conjugates for MRI Contrast Enhancement Nano Lett. 2010, 10, 484 489
  41. 41
    Chow, E. K.; Zhang, X.-Q.; Chen, M.; Lam, R.; Robinson, E.; Huang, H.; Schaffer, D.; Osawa, E.; Goga, A.; Ho, D. Nanodiamond Therapeutic Delivery Agents Mediate Enhanced Chemoresistant Tumor Treatment Sci. Transl. Med. 2011, 3, 73ra21
  42. 42
    Moore, L.; Chow, E. K.-H.; Osawa, E.; Bishop, J. M.; Ho, D. Diamond-Lipid Hybrids Enhance Chemotherapeutic Tolerance and Mediate Tumor Regression Adv. Mater. 2013, 25, 3532 3541
  43. 43
    Wang, X.; Low, X. C.; Hou, W.; Abdullah, L. N.; Toh, T. B.; Mohd Abdul Rashid, M.; Ho, D.; Chow, E. K.-H. Epirubicin-Adsorbed Nanodiamonds Kill Chemoresistant Hepatic Cancer Stem Cells ACS Nano 2014, 8, 12151 12166
  44. 44
    Wang, H.; Lee, D.-K.; Chen, K.-Y.; Chen, J.-Y.; Zhang, K.; Silva, A.; Ho, C.-M.; Ho, D. Mechanism-Independent Optimization of Combinatorial Nanodiamond and Unmodified Drug Delivery Using a Phenotypically Driven Platform Technology ACS Nano 2015, 9, 3332 3344
  45. 45
    Paithankar, D.; Hwang, B. H.; Munavalli, G.; Kauvar, A.; Lloyd, J.; Blomgren, R.; Faupel, L.; Meyer, T.; Mitragotri, S. Ultrasonic Delivery of Silica–Gold Nanoshells for Photothermolysis of Sebaceous Glands in Humans: Nanotechnology from the Bench to Clinic J. Controlled Release 2015, 206, 30 36
  46. 46
    Tezel, A.; Mitragotri, S. Interactions of Inertial Cavitation Bubbles with Stratum Corneum Lipid Bilayers during Low-Frequency Sonophoresis Biophys. J. 2003, 85, 3502 3512
  47. 47
    Chiappini, C.; Martinez, J. O.; De Rosa, E.; Almeida, C. S.; Tasciotti, E.; Stevens, M. M. Biodegradable Nanoneedles for Localized Delivery of Nanoparticles in Vivo: Exploring the Biointerface ACS Nano 2015, 9, 5500 5509
  48. 48
    Chiappini, C.; De Rosa, E.; Martinez, J.; Liu, X.; Steele, J.; Stevens, M.; Tasciotti, E. Biodegradable Silicon Nanoneedles Delivering Nucleic Acids Intracellularly Induce Localized In Vivo Neovascularization Nat. Mater. 2015, 14, 532 539
  49. 49
    De La Rica, R.; Stevens, M. M. Plasmonic ELISA for the Ultrasensitive Detection of Disease Biomarkers with the Naked Eye Nat. Nanotechnol. 2012, 7, 821 824
  50. 50
    Chapman, R.; Lin, Y.; Burnapp, M.; Bentham, A.; Hillier, D.; Zabron, A.; Khan, S.; Tyreman, M.; Stevens, M. M. Multivalent Nanoparticle Networks Enable Point-of-Care Detection of Human Phospholipase-A2 in Serum ACS Nano 2015, 9, 2565 2573
  51. 51
    Dewitte, H.; Verbeke, R.; Breckpot, K.; De Smedt, S. C.; Lentacker, I. Nanoparticle Design To Induce Tumor Immunity and Challenge the Suppressive Tumor Microenvironment Nano Today 2014, 9, 743 758
  52. 52
    Gough, D. R.; Cotter, T. G. Hydrogen Peroxide: A Jekyll and Hyde Signalling Molecule Cell Death Dis. 2011, 2, e213
  53. 53
    Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free Radicals, Antioxidants and Functional Foods: Impact on Human Health Pharmacogn. Rev. 2010, 4, 118 126
  54. 54
    Sun, X.; Guo, S.; Liu, Y.; Sun, S. Dumbbell-like PtPd–Fe3O4 Nanoparticles for Enhanced Electrochemical Detection of H2O2 Nano Lett. 2012, 12, 4859 4863
  55. 55
    Zhu, H.; Sigdel, A.; Zhang, S.; Su, D.; Xi, Z.; Li, Q.; Sun, S. Core/Shell Au/MnO Nanoparticles Prepared through Controlled Oxidation of AuMn as an Electrocatalyst for Sensitive H2O2 Detection Angew. Chem., Int. Ed. 2014, 53, 12508 12512
  56. 56
    Sun, X.; Cai, W.; Chen, X. Positron Emission Tomography Imaging Using Radiolabeled Inorganic Nanomaterials Acc. Chem. Res. 2015, 48, 286 294
  57. 57
    Liu, X.; Wang, S. Three-Dimensional Nano-Biointerface as a New Platform for Guiding Cell Fate Chem. Soc. Rev. 2014, 43, 2385 2401
  58. 58
    Ma, L.; Yang, G.; Wang, N.; Zhang, P. C.; Guo, F. Y.; Meng, J. X.; Zhang, F. L.; Hu, Z. J.; Wang, S. T.; Zhao, Y. Trap Effect of Three-Dimensional Fibers Network for High Efficient Cancer-Cell Capture Adv. Healthcare Mater. 2015, 4, 838 843
  59. 59
    Zhang, P.; Chen, L.; Xu, T.; Liu, H.; Liu, X.; Meng, J.; Yang, G.; Jiang, L.; Wang, S. Programmable Fractal Nanostructured Interfaces for Specific Recognition and Electrochemical Release of Cancer Cells Adv. Mater. 2013, 25, 3566 3570
  60. 60
    Liu, X.; Chen, L.; Liu, H.; Yang, G.; Zhang, P.; Han, D.; Wang, S.; Jiang, L. Bio-Inspired Soft Polystyrene Nanotube Substrate for Rapid and Highly Efficient Breast Cancer Cell Capture NPG Asia Mater. 2013, 5, e63
  61. 61
    Liu, H.; Liu, X.; Meng, J.; Zhang, P.; Yang, G.; Su, B.; Sun, K.; Chen, L.; Han, D.; Wang, S.; Jiang, L. Hydrophobic Interaction-Mediated Capture and Release of Cancer Cells on Thermoresponsive Nanostructured Surfaces Adv. Mater. 2013, 25, 922 927
  62. 62
    Liu, H. L.; Li, Y. Y.; Sun, K.; Fan, J. B.; Zhang, P. C.; Meng, J. X.; Wang, S. T.; Jiang, L. Dual-Responsive Surfaces Modified with Phenylboronic Acid-Containing Polymer Brush To Reversibly Capture and Release Cancer Cells J. Am. Chem. Soc. 2013, 135, 7603 7609
  63. 63
    Wang, S.; Liu, K.; Liu, J.; Yu, Z. T. F.; Xu, X.; Zhao, L.; Lee, T.; Lee, E. K.; Reiss, J.; Lee, Y.-K. Highly Efficient Capture of Circulating Tumor Cells by Using Nanostructured Silicon Substrates with Integrated Chaotic Micromixers Angew. Chem., Int. Ed. 2011, 50, 3084 3088
  64. 64
    Duan, R.; Zuo, X.; Wang, S.; Quan, X.; Chen, D.; Chen, Z.; Jiang, L.; Fan, C.; Xia, F. Lab in a Tube: Ultrasensitive Detection of MicroRNAs at the Single-Cell Level and in Breast Cancer Patients Using Quadratic Isothermal Amplification J. Am. Chem. Soc. 2013, 135, 4604 4607
  65. 65
    Liu, W.; Thomopoulos, S.; Xia, Y. Electrospun Nanofibers for Regenerative Medicine Adv. Healthcare Mater. 2012, 1, 10 25
  66. 66
    Xie, J.; Liu, W.; MacEwan, M. R.; Bridgman, P. C.; Xia, Y. Neurite Outgrowth on Electrospun Nanofibers with Uniaxial Alignment: The Effects of Fiber Density, Surface Coating, and Supporting Substrate ACS Nano 2014, 8, 1878 1885
  67. 67
    Smith, L.; Xia, Y.; Galatz, L. M.; Genin, G. M.; Thomopoulos, S. Tissue-Engineering Strategies for the Tendon/Ligament-to-Bone Insertion Connect. Tissue Res. 2012, 53, 95 105
  68. 68
    Xie, J.; MacEwan, M. R.; Ray, W. Z.; Liu, W.; Siewe, D. Y.; Xia, Y. Radially Aligned, Electrospun Nanofibers as Dural Substitutes for Wound Closure and Tissue Regeneration Applications ACS Nano 2010, 4, 5027 5036
  69. 69
    Lang, N.; Pereira, M. J.; Lee, Y.; Friehs, I.; Vasilyev, N. V.; Feins, E. N.; Ablasser, K.; O’Cearbhaill, E. D.; Xu, C.; Fabozzo, A. A Blood-Resistant Surgical Glue for Minimally Invasive Repair of Vessels and Heart Defects Sci. Transl. Med. 2014, 6, 218ra6
  70. 70
    Yang, S. Y.; O’Cearbhaill, E. D.; Sisk, G. C.; Park, K. M.; Cho, W. K.; Villiger, M.; Bouma, B. E.; Pomahac, B.; Karp, J. M. A Bio-Inspired Swellable Microneedle Adhesive for Mechanical Interlocking with Tissue Nat. Commun. 2013, 4, 1702
  71. 71
    Cho, W. K.; Ankrum, J. A.; Guo, D.; Chester, S. A.; Yang, S. Y.; Kashyap, A.; Campbell, G. A.; Wood, R. J.; Rijal, R. K.; Karnik, R. Microstructured Barbs on the North American Porcupine Quill Enable Easy Tissue Penetration and Difficult Removal Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 21289 21294
  72. 72
    Liu, Y.; Lim, J.; Teoh, S.-H. Review: Development of Clinically Relevant Scaffolds for Vascularised Bone Tissue Engineering Biotechnol. Adv. 2013, 31, 688 705
  73. 73
    Lim, J.; Chong, M. S. K.; Chan, J. K. Y.; Teoh, S. H. Polymer Powder Processing of Cryomilled Polycaprolactone for Solvent-Free Generation of Homogeneous Bioactive Tissue Engineering Scaffolds Small 2014, 10, 2495 2502
  74. 74
    Peer, D.; Karp, J. M.; Hong, S.; Farokhzad, O. C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for Cancer Therapy Nat. Nanotechnol. 2007, 2, 751 760
  75. 75
    Chow, E. K.-H.; Ho, D. Cancer Nanomedicine: From Drug Delivery to Imaging Sci. Transl. Med. 2013, 5, 216rv4
  76. 76
    Godwin, H.; Nameth, C.; Avery, D.; Bergeson, L. L.; Bernard, D.; Beryt, E.; Boyes, W.; Brown, S.; Clippinger, A. J.; Cohen, Y. Nanomaterial Categorization for Assessing Risk Potential to Facilitate Regulatory Decision-Making ACS Nano 2015, 9, 3409 3417
  77. 77
    Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-Responsive Nanocarriers for Drug Delivery Nat. Mater. 2013, 12, 991 1003
  78. 78
    Pacardo, D. B.; Ligler, F. S.; Gu, Z. Programmable Nanomedicine: Synergistic and Sequential Drug Delivery Systems Nanoscale 2015, 7, 3381 3391
  79. 79
    Lu, Y.; Sun, W.; Gu, Z. Stimuli-Responsive Nanomaterials for Therapeutic Protein Delivery J. Controlled Release 2014, 194, 1 19
  80. 80
    Gu, Z.; Aimetti, A. A.; Wang, Q.; Dang, T. T.; Zhang, Y.; Veiseh, O.; Cheng, H.; Langer, R. S.; Anderson, D. G. Injectable Nano-network for Glucose-Mediated Insulin Delivery ACS Nano 2013, 7, 4194 4201
  81. 81
    Mo, R.; Jiang, T.; DiSanto, R.; Tai, W.; Gu, Z. ATP-Triggered Anticancer Drug Delivery Nat. Commun. 2014, 5, 3364
  82. 82
    Gu, Z.; Dang, T. T.; Ma, M.; Tang, B. C.; Cheng, H.; Jiang, S.; Dong, Y.; Zhang, Y.; Anderson, D. G. Glucose-Responsive Microgels Integrated with Enzyme Nanocapsules for Closed-Loop Insulin Delivery ACS Nano 2013, 7, 6758 6766
  83. 83
    Godwin, H.; Nameth, C.; Avery, D.; Bergeson, L. L.; Bernard, D.; Beryt, E.; Boyes, W.; Brown, S.; Clippinger, A. J.; Cohen, Y. Nanomaterial Categorization for Assessing Risk Potential to Facilitate Regulatory Decision-Making ACS Nano 2015, 9, 3409 3417
  84. 84
    Mitragotri, S.; Burke, P. A.; Langer, R. Overcoming the Challenges in Administering Biopharmaceuticals: Formulation and Delivery Strategies Nat. Rev. Drug Discovery 2014, 13, 655 672
  85. 85
    Chertok, B.; Webber, M. J.; Succi, M. D.; Langer, R. Drug Delivery Interfaces in the 21st Century: From Science Fiction Ideas to Viable Technologies Mol. Pharmaceutics 2013, 10, 3531 3543
  86. 86
    Hammond, P. T. A Growing Place for Nano in Medicine ACS Nano 2014, 8, 7551 7552
  87. 87
    Khademhosseini, A.; Peppas, N. A. Micro- and Nanoengineering of Biomaterials for Healthcare Applications Adv Healthc Mater 2013, 2, 10 12

Cited By

ARTICLE SECTIONS
Jump To

This article is cited by 264 publications.

  1. Menghuan Zhao, Peixin Guan, Shuxin Xu, Haifeng Lu, Zhen Liu. Molecularly Imprinted Nanomedicine for Anti-angiogenic Cancer Therapy via Blocking Vascular Endothelial Growth Factor Signaling. Nano Letters 2023, 23 (18) , 8674-8682. https://doi.org/10.1021/acs.nanolett.3c02514
  2. Wenhui Yi, Asif Khalid, Naila Arshad, M. Sohail Asghar, Muhammad Sultan Irshad, Xianbao Wang, Yueyang Yi, Jinhai Si, Xun Hou, Hong Rong Li. Recent Progress and Perspective of an Evolving Carbon Family From 0D to 3D: Synthesis, Biomedical Applications, and Potential Challenges. ACS Applied Bio Materials 2023, 6 (6) , 2043-2088. https://doi.org/10.1021/acsabm.3c00076
  3. Junyu Chen, Liucheng Mao, Yuming Jiang, Huihui Liu, Xiao Wang, Lingwei Meng, Qiuyao Du, Jing Han, Liuying He, Hongye Huang, Yawei Wang, Caiqiao Xiong, Yen Wei, Zongxiu Nie. Revealing the In Situ Behavior of Aggregation-Induced Emission Nanoparticles and Their Biometabolic Effects via Mass Spectrometry Imaging. ACS Nano 2023, 17 (5) , 4463-4473. https://doi.org/10.1021/acsnano.2c10058
  4. Larissa A. Maiorova, Nagao Kobayashi, Denis S. Salnikov, Sergey M. Kuzmin, Tamara V. Basova, Oscar I. Koifman, Vladimir I. Parfenyuk, Victor A. Bykov, Yurii A. Bobrov, Peng Yang. Supermolecular Nanoentities of Vitamin B12 Derivative as a Link in the Evolution of the Parent Molecules During Self-Assembly at the Air–Water Interface. Langmuir 2023, 39 (9) , 3246-3254. https://doi.org/10.1021/acs.langmuir.2c02964
  5. Yaron Marciano, Virginia del Solar, Nazia Nayeem, Dhwanit Dave, Jiye Son, María Contel, Rein V. Ulijn. Encapsulation of Gold-Based Anticancer Agents in Protease-Degradable Peptide Nanofilaments Enhances Their Potency. Journal of the American Chemical Society 2023, 145 (1) , 234-246. https://doi.org/10.1021/jacs.2c09820
  6. Gregory Jameson, Xinyao Xiang, Rafael Brüschweiler. Quantitative Multistate Binding Model of Silica Nanoparticle–Protein Interactions Obtained from Multinuclear Spin Relaxation. The Journal of Physical Chemistry B 2022, 126 (44) , 9089-9094. https://doi.org/10.1021/acs.jpcb.2c05967
  7. Shih-Ting Wang, Honghu Zhang, Sunting Xuan, Dmytro Nykypanchuk, Yugang Zhang, Guillaume Freychet, Benjamin M. Ocko, Ronald N. Zuckermann, Nevena Todorova, Oleg Gang. Compact Peptoid Molecular Brushes for Nanoparticle Stabilization. Journal of the American Chemical Society 2022, 144 (18) , 8138-8152. https://doi.org/10.1021/jacs.2c00743
  8. Elham Hatami, Prashanth K. B. Nagesh, Neeraj Chauhan, Meena Jaggi, Subhash C. Chauhan, Murali M. Yallapu. In Situ Nanoparticle Self-Assembly for Combination Delivery of Therapeutics to Non-Small Cell Lung Cancer. ACS Applied Bio Materials 2022, 5 (3) , 1104-1119. https://doi.org/10.1021/acsabm.1c01158
  9. Glenn A. O. Cremers, Bas J. H. M. Rosier, Ab Meijs, Nicholas B. Tito, Sander M. J. van Duijnhoven, Hans van Eenennaam, Lorenzo Albertazzi, Tom F. A. de Greef. Determinants of Ligand-Functionalized DNA Nanostructure–Cell Interactions. Journal of the American Chemical Society 2021, 143 (27) , 10131-10142. https://doi.org/10.1021/jacs.1c02298
  10. Yuliang Xie, Joseph Rufo, Ruoyu Zhong, Joseph Rich, Peng Li, Kam W. Leong, Tony Jun Huang. Microfluidic Isolation and Enrichment of Nanoparticles. ACS Nano 2020, 14 (12) , 16220-16240. https://doi.org/10.1021/acsnano.0c06336
  11. Dong-Jun Koo, Jinahn Choi, Minchul Ahn, Benjamin H. Ahn, Dal-Hee Min, Sung-Yon Kim. Large-Scale 3D Optical Mapping and Quantitative Analysis of Nanoparticle Distribution in Tumor Vascular Microenvironment. Bioconjugate Chemistry 2020, 31 (7) , 1784-1794. https://doi.org/10.1021/acs.bioconjchem.0c00263
  12. Chan Ho Chung, Wonsik Jung, Hyeongseop Keum, Tae Woo Kim, Sangyong Jon. Nanoparticles Derived from the Natural Antioxidant Rosmarinic Acid Ameliorate Acute Inflammatory Bowel Disease. ACS Nano 2020, 14 (6) , 6887-6896. https://doi.org/10.1021/acsnano.0c01018
  13. Zhiyuan Han, Suresh Sarkar, Andrew M. Smith. Zwitterion and Oligo(ethylene glycol) Synergy Minimizes Nonspecific Binding of Compact Quantum Dots. ACS Nano 2020, 14 (3) , 3227-3241. https://doi.org/10.1021/acsnano.9b08658
  14. Tianxing Man, Xiongfeng Zhu, Yu Ting Chow, Emma R. Dawson, Ximiao Wen, Alexander N. Patananan, Tingyi Leo Liu, Chuanzhen Zhao, Cong Wu, Jason S. Hong, Pei-Shan Chung, Daniel L. Clemens, Bai-Yu Lee, Paul S. Weiss, Michael A. Teitell, Pei-Yu Chiou. Intracellular Photothermal Delivery for Suspension Cells Using Sharp Nanoscale Tips in Microwells. ACS Nano 2019, 13 (9) , 10835-10844. https://doi.org/10.1021/acsnano.9b06025
  15. Andreja Majerle, Dominik T. Schmieden, Roman Jerala, Anne S. Meyer. Synthetic Biology for Multiscale Designed Biomimetic Assemblies: From Designed Self-Assembling Biopolymers to Bacterial Bioprinting. Biochemistry 2019, 58 (16) , 2095-2104. https://doi.org/10.1021/acs.biochem.8b00922
  16. David H. Howe, James L. Hart, Riki M. McDaniel, Mitra L. Taheri, Andrew J. D. Magenau. Functionalization-Induced Self-Assembly of Block Copolymers for Nanoparticle Synthesis. ACS Macro Letters 2018, 7 (12) , 1503-1508. https://doi.org/10.1021/acsmacrolett.8b00815
  17. Min Qiu, Xiuxiu Wang, Huanli Sun, Jian Zhang, Chao Deng, Zhiyuan Zhong. Cyclic RGD-Peptide-Functionalized Polylipopeptide Micelles for Enhanced Loading and Targeted Delivery of Monomethyl Auristatin E. Molecular Pharmaceutics 2018, 15 (11) , 4854-4861. https://doi.org/10.1021/acs.molpharmaceut.8b00498
  18. Theodora U. J. Bruun, Anne-Marie C. Andersson, Simon J. Draper, Mark Howarth. Engineering a Rugged Nanoscaffold To Enhance Plug-and-Display Vaccination. ACS Nano 2018, 12 (9) , 8855-8866. https://doi.org/10.1021/acsnano.8b02805
  19. Thomas Bonnard, Anand Jayapadman, Jasmine A. Putri, Jiwei Cui, Yi Ju, Catherine Carmichael, Thomas A. Angelovich, Stephen H. Cody, Shauna French, Karline Pascaud, Hannah A. Pearce, Shweta Jagdale, Frank Caruso, Christoph E. Hagemeyer. Low-Fouling and Biodegradable Protein-Based Particles for Thrombus Imaging. ACS Nano 2018, 12 (7) , 6988-6996. https://doi.org/10.1021/acsnano.8b02588
  20. Chao Wang, Di Wen, Zhen Gu. Cellular Bioparticulates with Therapeutics for Cancer Immunotherapy. Bioconjugate Chemistry 2018, 29 (3) , 702-708. https://doi.org/10.1021/acs.bioconjchem.7b00619
  21. Peijian Feng, Yulei Chen, Lei Zhang, Cheng-Gen Qian, Xuanzhong Xiao, Xu Han, and Qun-Dong Shen . Near-Infrared Fluorescent Nanoprobes for Revealing the Role of Dopamine in Drug Addiction. ACS Applied Materials & Interfaces 2018, 10 (5) , 4359-4368. https://doi.org/10.1021/acsami.7b12005
  22. Aaron Y. Mochizuki, Isaura M. Frost, Melina B. Mastrodimos, Ashley S. Plant, Anthony C. Wang, Theodore B. Moore, Robert M. Prins, Paul S. Weiss, and Steven J. Jonas . Precision Medicine in Pediatric Neurooncology: A Review. ACS Chemical Neuroscience 2018, 9 (1) , 11-28. https://doi.org/10.1021/acschemneuro.7b00388
  23. Benhui Hu, Wan Ru Leow, Pingqiang Cai, Yong-Qiang Li, Yun-Long Wu, and Xiaodong Chen . Nanomechanical Force Mapping of Restricted Cell-To-Cell Collisions Oscillating between Contraction and Relaxation. ACS Nano 2017, 11 (12) , 12302-12310. https://doi.org/10.1021/acsnano.7b06063
  24. Bin Zheng, Hanjie Wang, Huizhuo Pan, Chao Liang, Wanying Ji, Li Zhao, Hongbin Chen, Xiaoqun Gong, Xiaoli Wu, and Jin Chang . Near-Infrared Light Triggered Upconversion Optogenetic Nanosystem for Cancer Therapy. ACS Nano 2017, 11 (12) , 11898-11907. https://doi.org/10.1021/acsnano.7b06395
  25. Chieu D. Tran, James Makuvaza, Erik Munson, and Brian Bennett . Biocompatible Copper Oxide Nanoparticle Composites from Cellulose and Chitosan: Facile Synthesis, Unique Structure, and Antimicrobial Activity. ACS Applied Materials & Interfaces 2017, 9 (49) , 42503-42515. https://doi.org/10.1021/acsami.7b11969
  26. Christopher Poon, Sampreeti Chowdhuri, Cheng-Hsiang Kuo, Yun Fang, Francis J. Alenghat, Danielle Hyatt, Kian Kani, Mitchell E. Gross, and Eun Ji Chung . Protein Mimetic and Anticancer Properties of Monocyte-Targeting Peptide Amphiphile Micelles. ACS Biomaterials Science & Engineering 2017, 3 (12) , 3273-3282. https://doi.org/10.1021/acsbiomaterials.7b00600
  27. Heemin Kang, Sungkyu Kim, Dexter Siu Hong Wong, Hee Joon Jung, Sien Lin, Kaijie Zou, Rui Li, Gang Li, Vinayak P. Dravid, and Liming Bian . Remote Manipulation of Ligand Nano-Oscillations Regulates Adhesion and Polarization of Macrophages in Vivo. Nano Letters 2017, 17 (10) , 6415-6427. https://doi.org/10.1021/acs.nanolett.7b03405
  28. Min Qiu, Jia Ouyang, Huanli Sun, Fenghua Meng, Ru Cheng, Jian Zhang, Liang Cheng, Qing Lan, Chao Deng, and Zhiyuan Zhong . Biodegradable Micelles Based on Poly(ethylene glycol)-b-polylipopeptide Copolymer: A Robust and Versatile Nanoplatform for Anticancer Drug Delivery. ACS Applied Materials & Interfaces 2017, 9 (33) , 27587-27595. https://doi.org/10.1021/acsami.7b10533
  29. Paul S. Weiss (Editor-in-Chief). Upcoming Tenth Anniversary at ACS Nano. ACS Nano 2017, 11 (6) , 5189-5190. https://doi.org/10.1021/acsnano.7b04198
  30. Tatiana N. Pashirova, Irina V. Zueva, Konstantin A. Petrov, Vasily M. Babaev, Svetlana S. Lukashenko, Ildar Kh. Rizvanov, Eliana B. Souto, Evgeny E. Nikolsky, Lucia Ya. Zakharova, Patrick Masson, and Oleg G. Sinyashin . Nanoparticle-Delivered 2-PAM for Rat Brain Protection against Paraoxon Central Toxicity. ACS Applied Materials & Interfaces 2017, 9 (20) , 16922-16932. https://doi.org/10.1021/acsami.7b04163
  31. Ulrich Tritschler, Sam Pearce, Jessica Gwyther, George R. Whittell, and Ian Manners . 50th Anniversary Perspective: Functional Nanoparticles from the Solution Self-Assembly of Block Copolymers. Macromolecules 2017, 50 (9) , 3439-3463. https://doi.org/10.1021/acs.macromol.6b02767
  32. Lang Rao, Bo Cai, Lin-Lin Bu, Qing-Quan Liao, Shi-Shang Guo, Xing-Zhong Zhao, Wen-Fei Dong, and Wei Liu . Microfluidic Electroporation-Facilitated Synthesis of Erythrocyte Membrane-Coated Magnetic Nanoparticles for Enhanced Imaging-Guided Cancer Therapy. ACS Nano 2017, 11 (4) , 3496-3505. https://doi.org/10.1021/acsnano.7b00133
  33. Tomoya Suma, Jiwei Cui, Markus Müllner, Shiwei Fu, Jenny Tran, Ka Fung Noi, Yi Ju, and Frank Caruso . Modulated Fragmentation of Proapoptotic Peptide Nanoparticles Regulates Cytotoxicity. Journal of the American Chemical Society 2017, 139 (11) , 4009-4018. https://doi.org/10.1021/jacs.6b11302
  34. Xiuli Hu, Yuqi Zhang, Zhigang Xie, Xiabin Jing, Adriano Bellotti, and Zhen Gu . Stimuli-Responsive Polymersomes for Biomedical Applications. Biomacromolecules 2017, 18 (3) , 649-673. https://doi.org/10.1021/acs.biomac.6b01704
  35. Keting Xin, Man Li, Di Lu, Xuan Meng, Jun Deng, Deling Kong, Dan Ding, Zheng Wang, and Yanjun Zhao . Bioinspired Coordination Micelles Integrating High Stability, Triggered Cargo Release, and Magnetic Resonance Imaging. ACS Applied Materials & Interfaces 2017, 9 (1) , 80-91. https://doi.org/10.1021/acsami.6b09425
  36. Mattias Björnmalm, Jiwei Cui, Nadja Bertleff-Zieschang, Danzi Song, Matthew Faria, Md. Arifur Rahim, and Frank Caruso . Nanoengineering Particles through Template Assembly. Chemistry of Materials 2017, 29 (1) , 289-306. https://doi.org/10.1021/acs.chemmater.6b02848
  37. Jiwei Cui, Matthew Faria, Mattias Björnmalm, Yi Ju, Tomoya Suma, Sylvia T. Gunawan, Joseph J. Richardson, Hamed Heidari, Sara Bals, Edmund J. Crampin, and Frank Caruso . A Framework to Account for Sedimentation and Diffusion in Particle–Cell Interactions. Langmuir 2016, 32 (47) , 12394-12402. https://doi.org/10.1021/acs.langmuir.6b01634
  38. Mouzhe Xie, Alexandar L. Hansen, Jiaqi Yuan, and Rafael Brüschweiler . Residue-Specific Interactions of an Intrinsically Disordered Protein with Silica Nanoparticles and Their Quantitative Prediction. The Journal of Physical Chemistry C 2016, 120 (42) , 24463-24468. https://doi.org/10.1021/acs.jpcc.6b08213
  39. Mattias Björnmalm, Matthew Faria, Xi Chen, Jiwei Cui, and Frank Caruso . Dynamic Flow Impacts Cell–Particle Interactions: Sedimentation and Particle Shape Effects. Langmuir 2016, 32 (42) , 10995-11001. https://doi.org/10.1021/acs.langmuir.6b03216
  40. Mattias Björnmalm, Matthew Faria, and Frank Caruso . Increasing the Impact of Materials in and beyond Bio-Nano Science. Journal of the American Chemical Society 2016, 138 (41) , 13449-13456. https://doi.org/10.1021/jacs.6b08673
  41. Anton A. A. Smith, Kaja Zuwala, Oliver Pilgram, Karen Singers Johansen, Martin Tolstrup, Frederik Dagnæs-Hansen, and Alexander N. Zelikin . Albumin–Polymer–Drug Conjugates: Long Circulating, High Payload Drug Delivery Vehicles. ACS Macro Letters 2016, 5 (10) , 1089-1094. https://doi.org/10.1021/acsmacrolett.6b00544
  42. Lingyan Lv, Yan Jiang, Xin Liu, Baoyan Wang, Wei Lv, Yue Zhao, Huihui Shi, Quanyin Hu, Hongliang Xin, Qunwei Xu, and Zhen Gu . Enhanced Antiglioblastoma Efficacy of Neovasculature and Glioma Cells Dual Targeted Nanoparticles. Molecular Pharmaceutics 2016, 13 (10) , 3506-3517. https://doi.org/10.1021/acs.molpharmaceut.6b00523
  43. Yanqi Ye, Jinqiang Wang, Quanyin Hu, Gabrielle M. Hochu, Hongliang Xin, Chao Wang, and Zhen Gu . Synergistic Transcutaneous Immunotherapy Enhances Antitumor Immune Responses through Delivery of Checkpoint Inhibitors. ACS Nano 2016, 10 (9) , 8956-8963. https://doi.org/10.1021/acsnano.6b04989
  44. Karel Ulbrich, Kateřina Holá, Vladimir Šubr, Aristides Bakandritsos, Jiří Tuček, and Radek Zbořil . Targeted Drug Delivery with Polymers and Magnetic Nanoparticles: Covalent and Noncovalent Approaches, Release Control, and Clinical Studies. Chemical Reviews 2016, 116 (9) , 5338-5431. https://doi.org/10.1021/acs.chemrev.5b00589
  45. Sourabh Shukla, R. Dixon Dorand, Jay T. Myers, Sarah E. Woods, Neetu M. Gulati, Phoebe L. Stewart, Ulrich Commandeur, Alex Y. Huang, and Nicole F. Steinmetz . Multiple Administrations of Viral Nanoparticles Alter in Vivo Behavior—Insights from Intravital Microscopy. ACS Biomaterials Science & Engineering 2016, 2 (5) , 829-837. https://doi.org/10.1021/acsbiomaterials.6b00060
  46. Jisung Kim, Mia J. Biondi, Jordan J. Feld, and Warren C. W. Chan . Clinical Validation of Quantum Dot Barcode Diagnostic Technology. ACS Nano 2016, 10 (4) , 4742-4753. https://doi.org/10.1021/acsnano.6b01254
  47. Jiwei Cui, Benjamin Hibbs, Sylvia T. Gunawan, Julia A. Braunger, Xi Chen, Joseph J. Richardson, Eric Hanssen, and Frank Caruso . Immobilized Particle Imaging for Quantification of Nano- and Microparticles. Langmuir 2016, 32 (14) , 3532-3540. https://doi.org/10.1021/acs.langmuir.6b00229
  48. Quanyin Hu, Wujin Sun, Yue Lu, Hunter N. Bomba, Yanqi Ye, Tianyue Jiang, Ari J. Isaacson, and Zhen Gu . Tumor Microenvironment-Mediated Construction and Deconstruction of Extracellular Drug-Delivery Depots. Nano Letters 2016, 16 (2) , 1118-1126. https://doi.org/10.1021/acs.nanolett.5b04343
  49. Weinan Xu, Alexander A. Steinschulte, Felix A. Plamper, Volodymyr F. Korolovych, and Vladimir V. Tsukruk . Hierarchical Assembly of Star Polymer Polymersomes into Responsive Multicompartmental Microcapsules. Chemistry of Materials 2016, 28 (3) , 975-985. https://doi.org/10.1021/acs.chemmater.5b04934
  50. Dongquan Shi, Xingquan Xu, Yanqi Ye, Kai Song, Yixiang Cheng, Jin Di, Quanyin Hu, Jianxin Li, Huangxian Ju, Qing Jiang, and Zhen Gu . Photo-Cross-Linked Scaffold with Kartogenin-Encapsulated Nanoparticles for Cartilage Regeneration. ACS Nano 2016, 10 (1) , 1292-1299. https://doi.org/10.1021/acsnano.5b06663
  51. Quanyin Hu Zhen Gu . Cell Membrane-Mediated Anticancer Drug Delivery. 2016, 197-211. https://doi.org/10.1021/bk-2016-1224.ch010
  52. Warren W. C. Chan (Associate Editor), Sharon Glotzer (Associate Editor), Yury Gogotsi (Associate Editor), Jason H. Hafner (Associate Editor), Paula T. Hammond (Associate Editor), Mark C. Hersam (Associate Editor), Ali Javey (Associate Editor), Cherie R. Kagan (Associate Editor), Ali Khademhosseini (Associate Editor), Nicholas A. Kotov (Associate Editor), Shuit-Tong Lee (Associate Editor), Helmuth Möhwald (Associate Editor), Paul A. Mulvaney (Associate Editor), Andre E. Nel (Associate Editor), Peter J. Nordlander (Associate Editor), Wolfgang J. Parak (Associate Editor), Reginald M. Penner (Associate Editor), Andrey L. Rogach (Associate Editor), Raymond E. Schaak (Associate Editor), Molly M. Stevens (Associate Editor), Andrew T. S. Wee (Associate Editor), C. Grant Willson (Associate Editor), Heather L. Tierney (Managing Editor), and Paul S. Weiss (Editor-in-Chief). Grand Plans for Nano. ACS Nano 2015, 9 (12) , 11503-11505. https://doi.org/10.1021/acsnano.5b07280
  53. Jin Di, Shanshan Yao, Yanqi Ye, Zheng Cui, Jicheng Yu, Tushar K. Ghosh, Yong Zhu, and Zhen Gu . Stretch-Triggered Drug Delivery from Wearable Elastomer Films Containing Therapeutic Depots. ACS Nano 2015, 9 (9) , 9407-9415. https://doi.org/10.1021/acsnano.5b03975
  54. Wolfgang J. Parak (Associate Editor), Andre E. Nel (Associate Editor), Paul S. Weiss (Editor-in-Chief). Grand Challenges for Nanoscience and Nanotechnology. ACS Nano 2015, 9 (7) , 6637-6640. https://doi.org/10.1021/acsnano.5b04386
  55. Kajal P. Baviskar, Brijesh M. Shah, Anjali P. Bedse, Shilpa S. Raut, Suchita P. Dhamane, Dhara J. Dave. Nanoparticle Properties. 2024, 153-191. https://doi.org/10.1002/9781394175482.ch5
  56. Sonia yadav, Sruthy Varghese, Reena Devi, Davinder Kumar, Naveen Khatri, Gajendra Singh, Virender Kumar. Biomaterials in Medical Applications. Current Materials Science 2024, 17 (3) , 212-239. https://doi.org/10.2174/2666145416666230420094148
  57. İlhan Özen, Aslı Demir, Muhammed İbrahim Bahtiyari, Xungai Wang, Azadeh Nilghaz, Peng Wu, Kamyar Shirvanimoghaddam, Minoo Naebe. Multifaceted applications of thymol/carvacrol-containing polymeric fibrous structures. Advanced Industrial and Engineering Polymer Research 2024, 7 (2) , 182-200. https://doi.org/10.1016/j.aiepr.2023.09.001
  58. Rosangela Mastrangelo, David Chelazzi, Piero Baglioni. New horizons on advanced nanoscale materials for Cultural Heritage conservation. Nanoscale Horizons 2024, 9 (4) , 566-579. https://doi.org/10.1039/D3NH00383C
  59. Gherardo Baudo, Hannah Flinn, Morgan Holcomb, Anjana Tiwari, Sirena Soriano, Francesca Taraballi, Biana Godin, Assaf Zinger, Sonia Villapol. Sex‐dependent improvement in traumatic brain injury outcomes after liposomal delivery of dexamethasone in mice. Bioengineering & Translational Medicine 2024, 21 https://doi.org/10.1002/btm2.10647
  60. Ashraf Abedin, Jhonattan David Manosalvas Mora, Adiba Azad, Srikar Bhattar, S. M. Rezwanul Islam, Mohammad Hasibul Hasan. Recent Advancement of Nanotechnology in Bio Applications. 2024, 59-104. https://doi.org/10.1007/978-3-031-39404-1_2
  61. Alexander Shao‐Rong Pang, Vishalli Dinesh, Natalie Yan‐Lin Pang, Tarini Dinesh, Kimberley Yun‐Lin Pang, George W. Yip, Boon Huat Bay, Dinesh Kumar Srinivasan. Precision medicine in myocardial infarction: Nanotheranostic strategies. Nano Select 2023, 69 https://doi.org/10.1002/nano.202300127
  62. H.M. Abelaira, A.B. de Moura, M.M. Cardoso, E. de Pieri, J.S. Abel, G.P. Luiz, E.M. Sombrio, L.A. Borghezan, R.S. Anastácio, L.A. Cruz, T.G. de Souza, Corrêa MEAB, I.R. Lima, C. da Costa, A.G. Dal Bó, Silveira PCL, R.A. Machado-de-Ávila. Sertraline associated with gold nanoparticles reduce cellular toxicity and induce sex-specific responses in behavior and neuroinflammation biomarkers in a mouse model of anxiety. Pharmacology Biochemistry and Behavior 2023, 233 , 173661. https://doi.org/10.1016/j.pbb.2023.173661
  63. Xin Li, Yue Gao, Helin Li, Jean-Pierre Majoral, Xiangyang Shi, Andrij Pich. Smart and bioinspired systems for overcoming biological barriers and enhancing disease theranostics. Progress in Materials Science 2023, 140 , 101170. https://doi.org/10.1016/j.pmatsci.2023.101170
  64. Kamal Tripathi, Hitesh Garg, R. Rajesh, Satyavani Vemparala. The conformational phase diagram of charged polymers in the presence of attractive bridging crowders. The Journal of Chemical Physics 2023, 159 (20) https://doi.org/10.1063/5.0172696
  65. Yinfeng Yang, Xiaomei Wang, Fangfang Yang, Bin Mu, Aiqin Wang. Progress and future prospects of hemostatic materials based on nanostructured clay minerals. Biomaterials Science 2023, 11 (23) , 7469-7488. https://doi.org/10.1039/D3BM01326J
  66. Asmaa Ibrahim, Hend A. Ezzat, Mervat Abd El Aal. Electronic properties and molecular electrostatic potential mapping of edge functionalized GQDs with ZnO, CuO, and TiO2. Optical and Quantum Electronics 2023, 55 (12) https://doi.org/10.1007/s11082-023-05427-y
  67. Vladimir F. Mironov, Mudaris N. Dimukhametov, Andrey V. Nemtarev, Tatiana N. Pashirova, Olga V. Tsepaeva, Alexandra D. Voloshina, Alexandra B. Vyshtakalyuk, Igor A. Litvinov, Anna P. Lyubina, Anastasiia S. Sapunova, Dinara F. Abramova, Vladimir V. Zobov. Novel Mitochondria-Targeted Amphiphilic Aminophosphonium Salts and Lipids Nanoparticles: Synthesis, Antitumor Activity and Toxicity. Nanomaterials 2023, 13 (21) , 2840. https://doi.org/10.3390/nano13212840
  68. Priyanka, Mai Abdel Haleem Abusalah, Hitesh Chopra, Abhilasha Sharma, Suhad Asad Mustafa, Om Prakash Choudhary, Manish Sharma, Manish Dhawan, Rajiv Khosla, Aanchal Loshali, Ankush Sundriyal, Jyoti Saini. Nanovaccines: A game changing approach in the fight against infectious diseases. Biomedicine & Pharmacotherapy 2023, 167 , 115597. https://doi.org/10.1016/j.biopha.2023.115597
  69. Mei Wu, Ruochen Shi, Ruishi Qi, Yuehui Li, Jinlong Du, Peng Gao. Four-dimensional electron energy-loss spectroscopy. Ultramicroscopy 2023, 253 , 113818. https://doi.org/10.1016/j.ultramic.2023.113818
  70. Honglei Zhang, Jiali Sun, Yu Zhang, Zhenqian Zhang, Xiaoshuang Wang, Zhilin Liu, Xuefei Zhang, Zhaohui Tang, Xuesi Chen. Preparation of an Ultrahigh-DAR PDL1 monoclonal antibody-polymeric-SN38 conjugate for precise colon cancer therapy. Biomaterials 2023, 301 , 122285. https://doi.org/10.1016/j.biomaterials.2023.122285
  71. K. Vignesh, D. Sivaganesh, V. Kavitha, M. Prema Rani. Unveiling the luminescent brilliance of europium-sourced cerium oxide: a comprehensive exploration for biomedical advancements through in vitro and in vivo studies. Applied Physics A 2023, 129 (10) https://doi.org/10.1007/s00339-023-06957-8
  72. Weihua Zhao, Xinyi Zhang, Ruiting Tian, Hongbo Li, Shengliang Zhong, Ruqin Yu. The sensor platform combined with dual signal amplification and based on UCNPs and CRISPR/Cas12a for MiRNA-21 detection. Sensors and Actuators B: Chemical 2023, 393 , 134238. https://doi.org/10.1016/j.snb.2023.134238
  73. Ganhua Xie, Shipei Zhu, Paul Y. Kim, Shubao Jiang, Qinpiao Yi, Pei Li, Zonglin Chu, Brett A. Helms, Thomas P. Russell. Relaxing Wrinkles in Jammed Interfacial Assemblies. Angewandte Chemie 2023, 135 (36) https://doi.org/10.1002/ange.202307713
  74. Ganhua Xie, Shipei Zhu, Paul Y. Kim, Shubao Jiang, Qinpiao Yi, Pei Li, Zonglin Chu, Brett A. Helms, Thomas P. Russell. Relaxing Wrinkles in Jammed Interfacial Assemblies. Angewandte Chemie International Edition 2023, 62 (36) https://doi.org/10.1002/anie.202307713
  75. Rekha Thiruvengadam, Jin Hee Kim. Therapeutic strategy for oncovirus-mediated oral cancer: A comprehensive review. Biomedicine & Pharmacotherapy 2023, 165 , 115035. https://doi.org/10.1016/j.biopha.2023.115035
  76. Giovanna Viola, Fulvio Floriani, Carlo Giorgio Barracchia, Francesca Munari, Mariapina D'Onofrio, Michael Assfalg. Ultrasmall Gold Nanoparticles as Clients of Biomolecular Condensates. Chemistry – A European Journal 2023, 29 (46) https://doi.org/10.1002/chem.202301274
  77. Maurice Retout, Bryan Gosselin, Amina Adrović, Pascale Blond, Ivan Jabin, Gilles Bruylants. Ultra-stable silver nanoplates: efficient and versatile colorimetric reporters for dipstick assays. Nanoscale 2023, 15 (28) , 11981-11989. https://doi.org/10.1039/D3NR02378H
  78. Feng Han, Qingchen Meng, En Xie, Kexin Li, Jie Hu, Qianglong Chen, Jiaying Li, Fengxuan Han. Engineered biomimetic micro/nano-materials for tissue regeneration. Frontiers in Bioengineering and Biotechnology 2023, 11 https://doi.org/10.3389/fbioe.2023.1205792
  79. Vidhya Rekha Umapathy, Prabhu Manickam Natarajan, Bhuminathan Swamikannu. Review of the Role of Nanotechnology in Overcoming the Challenges Faced in Oral Cancer Diagnosis and Treatment. Molecules 2023, 28 (14) , 5395. https://doi.org/10.3390/molecules28145395
  80. Karuvelan Murugan, Rajakannu Subashini, Udayadasan Sathiskumar, Greeshma Odukkathil. Calotropis procera flower extract for the synthesis of double edged octahedral α-Fe 2 O 3 nanoparticles via a greener approach: an insight into its structure property relationship for Escherichia coli. New Journal of Chemistry 2023, 47 (24) , 11584-11593. https://doi.org/10.1039/D3NJ01044A
  81. Darshan Tagadur Govindaraju, Chih-Hao Chen, K. T. Shalumon, Hao-Hsi Kao, Jyh-Ping Chen. Bioactive Nanostructured Scaffold-Based Approach for Tendon and Ligament Tissue Engineering. Nanomaterials 2023, 13 (12) , 1847. https://doi.org/10.3390/nano13121847
  82. Ranjeet Singh, Prateek Srivastava, Partha Pratim Manna. Chemokine-targeted nanoparticles: stimulation of the immune system in cancer immunotherapy. Exploration of Immunology 2023, , 123-147. https://doi.org/10.37349/ei.2023.00093
  83. Zahra Tariq, Muhammad Imran Qadeer, Iram Anjum, Christophe Hano, Sumaira Anjum. Thalassemia and Nanotheragnostics: Advanced Approaches for Diagnosis and Treatment. Biosensors 2023, 13 (4) , 450. https://doi.org/10.3390/bios13040450
  84. Hoi Man Leung, Hoi Ching Chu, Zheng-Wei Mao, Pik Kwan Lo. Versatile nanodiamond-based tools for therapeutics and bioimaging. Chemical Communications 2023, 59 (15) , 2039-2055. https://doi.org/10.1039/D2CC06495B
  85. İlhan Özen, Xungai Wang. Biomedicine: electrospun nanofibrous hormonal therapies through skin/tissue—a review. International Journal of Polymeric Materials and Polymeric Biomaterials 2023, 72 (1) , 21-39. https://doi.org/10.1080/00914037.2021.1985493
  86. Shuo Wang, Shuhao Ma, Rongxin Li, Xiaojing Qi, Keqin Han, Ling Guo, Xuejin Li. Probing the Interaction Between Supercarrier RBC Membrane and Nanoparticles for Optimal Drug Delivery. Journal of Molecular Biology 2023, 435 (1) , 167539. https://doi.org/10.1016/j.jmb.2022.167539
  87. Xinghua Guo, Pan Song, Feng Li, Qihao Yan, Yan Bai, Jincan He, Qishi Che, Hua Cao, Jiao Guo, Zhengquan Su. Research Progress of Design Drugs and Composite Biomaterials in Bone Tissue Engineering. International Journal of Nanomedicine 2023, Volume 18 , 3595-3622. https://doi.org/10.2147/IJN.S415666
  88. Sonali Naik, Arun Torris, S. Kiran. Construction and application of bionanomaterials. 2023, 567-594. https://doi.org/10.1016/B978-0-12-820557-0.00009-6
  89. Yi Yang, Dexu Luo, Muhammad Inam, Jialin Hu, You Zhou, Chuanshan Xu, Wenjie Chen. A scientometrics study of the nanomedicines assisted in respiratory diseases. Frontiers in Bioengineering and Biotechnology 2022, 10 https://doi.org/10.3389/fbioe.2022.1053653
  90. Mehrdad Afarid, Shirin Mahmoodi, Roghayyeh Baghban. Recent achievements in nano-based technologies for ocular disease diagnosis and treatment, review and update. Journal of Nanobiotechnology 2022, 20 (1) https://doi.org/10.1186/s12951-022-01567-7
  91. Xiaoyu Liu, Yunru Yu, Dechen Liu, Jingbo Li, Ji Sun, Qiong Wei, Yuanjin Zhao, S. J. Pandol, Ling Li. Porous microcapsules encapsulating β cells generated by microfluidic electrospray technology for diabetes treatment. NPG Asia Materials 2022, 14 (1) https://doi.org/10.1038/s41427-022-00385-5
  92. Manav Raj Kar, Urjjarani Patel, Saikat Bhaumik. Highly stable and water dispersible polymer-coated CsPbBr 3 nanocrystals for Cu-ion detection in water. Materials Advances 2022, 3 (23) , 8629-8638. https://doi.org/10.1039/D2MA00719C
  93. Jamelah S. Al-Otaibi, Y. Sheena Mary, Y. Shyma Mary, Savaş Kaya, Goncagül Serdaroglu. DFT computational study of trihalogenated aniline derivative’s adsorption onto graphene/fullerene/fullerene-like nanocages, X 12 Y 12 (X = Al, B, and Y = N, P). Journal of Biomolecular Structure and Dynamics 2022, 40 (19) , 8630-8643. https://doi.org/10.1080/07391102.2021.1914172
  94. Darshan T.G., Chih-Hao Chen, Chang-Yi Kuo, K.T. Shalumon, Yen-Miao Chien, Hao-Hsi Kao, Jyh-Ping Chen. Development of high resilience spiral wound suture-embedded gelatin/PCL/heparin nanofiber membrane scaffolds for tendon tissue engineering. International Journal of Biological Macromolecules 2022, 221 , 314-333. https://doi.org/10.1016/j.ijbiomac.2022.09.001
  95. Y. Shyma Mary, Y. Sheena Mary, Zakir Ullah. Computational Study of Sorbic Acid Drug Adsorption onto Coronene/Fullerene/Fullerene-Like X12Y12 (X = Al, B and Y = N, P) Nanocages: DFT and Molecular Docking Investigations. Journal of Cluster Science 2022, 33 (4) , 1809-1819. https://doi.org/10.1007/s10876-021-02106-4
  96. Wenbin Diao, Ben Yang, Sipeng Sun, Anping Wang, Rongguan Kou, Qianyun Ge, Mengqi Shi, Bo Lian, Tongyi Sun, Jingliang Wu, Jingkun Bai, Meihua Qu, Yubing Wang, Wenjing Yu, Zhiqin Gao. PNA-Modified Liposomes Improve the Delivery Efficacy of CAPIRI for the Synergistic Treatment of Colorectal Cancer. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.893151
  97. Muhammed Onur Avci, Nedim Muzoglu, Aysel Ersoy Yilmaz, Binboga Siddik Yarman. Antibacterial, cytotoxicity and biodegradability studies of polycaprolactone nanofibers holding green synthesized Ag nanoparticles using atropa belladonna extract. Journal of Biomaterials Science, Polymer Edition 2022, 33 (9) , 1157-1180. https://doi.org/10.1080/09205063.2022.2045665
  98. Zechang Chen, Junbo Zhuang, Jiadong Pang, Zehao Liu, Penghao Zhang, Haijun Deng, Liming Zhang, Baoxiong Zhuang. Application of a cationic amylose derivative loaded with single‐walled carbon nanotubes for gene delivery therapy and photothermal therapy of colorectal cancer. Journal of Biomedical Materials Research Part A 2022, 110 (5) , 1052-1061. https://doi.org/10.1002/jbm.a.37351
  99. Hong Liang, Fangming Zhang, Yannv Hong, Yue Wu, Huanzhang Xie, Chen Zhang, Zonghua Wang, Zhonglei Lu, Huanghao Yang. Synergistic Silencing of Skp2 by siRNA Self‐Assembled Nanoparticles as a Therapeutic Strategy for Advanced Prostate Cancer. Small 2022, 18 (14) https://doi.org/10.1002/smll.202106046
  100. Woohyun Park, Van Phuc Nguyen, Yale Jeon, Bongjoong Kim, Yanxiu Li, Jonghun Yi, Hyungjun Kim, Jung Woo Leem, Young L. Kim, Dong Rip Kim, Yannis M. Paulus, Chi Hwan Lee. Biodegradable silicon nanoneedles for ocular drug delivery. Science Advances 2022, 8 (13) https://doi.org/10.1126/sciadv.abn1772
Load more citations
  • Abstract

    Figure 1

    Figure 1. In vivo activity of immunomodulatory SNAs as cancer vaccines.

    Figure 2

    Figure 2. Barriers to successful in vivo delivery of nucleic acids using nonviral vectors. Reprinted with permission from ref 19. Copyright 2014 Macmillan Publishers Ltd.

    Figure 3

    Figure 3. Subconjunctival instillation of nanocarriers incorporating latanoprost lowers eye pressure in glaucoma patients for up to 3 months. Reproduced from ref 37. Copyright 2014 American Chemical Society.

    Figure 4

    Figure 4. Schematic for the nanoparticle-based treatment of acne. Silica–gold nanoparticles are delivered into sebaceous glands using low-frequency ultrasound. Nanoparticles are then activated using near-infrared light to induce thermolysis. Reprinted with permission from ref 45. Copyright 2015 Elsevier.

    Figure 5

    Figure 5. Nanoneedle for the delivery of VEGF-165 to upregulate blood vessel formation in muscle. (A) Scanning electron microscope (SEM) micrographs showing the morphology of porous silicon nanoneedle arrays with pitches of 2 μm. Scale bars, 2 μm. (B) High-resolution SEM micrographs of nanoneedle tips showing the nanoneedles’ porous structure and the tunability of tip diameter from less than 100 nm to over 400 nm. Scale bars, 200 nm. (C) Intravital bright-field (top) and confocal (bottom) microscopy images of the vasculature of untreated (left) and hVEGF-165-treated muscles with either direct injection (center) or nanoinjection (right). The fluorescence signal originates from systemically injected FITC–dextran. Scale bars, bright-field 100 μm; confocal 50 μm. (D,E) Quantification of the fraction of fluorescent signal (dextran) (D) and the number of nodes in the vasculature per mm2 (E) within each field of view acquired for untreated control, intramuscular injection (IM) and nanoinjection. *p = 0.05, **p < 0.01, ***p < 0.001. Error bars represent the SD of the averages of five areas taken from three animals. Reprinted with permission of figure and caption text from ref 48. Copyright 2015 Macmillan Publishers Ltd.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 87 other publications.

    1. 1
      Ratner, B. D.; Hoffman, A. S.; Schoen, F. J.; Lemons, J. E. Biomaterials Science: An Introduction to Materials in Medicine; Elsevier Academic Press: Amsterdam, 2004.
    2. 2
      Lee, S.; Henthorn, D. Materials in Biology and Medicine; CRC Press/Taylor & Francis Group: Boca Raton, FL, 2012.
    3. 3
      Langer, R. Drug Delivery and Targeting Nature 1998, 392, 5 10
    4. 4
      Farokhzad, O. C.; Langer, R. Nanomedicine: Developing Smarter Therapeutic and Diagnostic Modalities Adv. Drug Delivery Rev. 2006, 58, 1456 1459
    5. 5
      Doshi, N.; Mitragotri, S. Designer Biomaterials for Nanomedicine Adv. Funct. Mater. 2009, 19, 3843 3854
    6. 6
      Mitragotri, S.; Lahann, J. Physical Approaches to Biomaterial Design Nat. Mater. 2009, 8, 15 23
    7. 7
      Dobrovolskaia, M. A.; McNeil, S. E. Immunological Properties of Engineered Nanomaterials Nat. Nanotechnol. 2007, 2, 469 478
    8. 8
      Howes, P. D.; Chandrawati, R.; Stevens, M. M. Colloidal Nanoparticles as Advanced Biological Sensors Science 2014, 346, 1247390
    9. 9
      Etheridge, M. L.; Campbell, S. A.; Erdman, A. G.; Haynes, C. L.; Wolf, S. M.; McCullough, J. The Big Picture on Nanomedicine: The State of Investigational and Approved Nanomedicine Products Nanomedicine 2013, 9, 1 14
    10. 10
      Mirkin, C. A.; Letsinger, R. L.; Mucic, R. C.; Storhoff, J. J. A DNA-Based Method for Rationally Assembling Nanoparticles into Macroscopic Materials Nature 1996, 382, 607 609
    11. 11
      Cutler, J. I.; Auyeung, E.; Mirkin, C. A. Spherical Nucleic Acids J. Am. Chem. Soc. 2012, 134, 1376 1391
    12. 12
      Rosi, N. L.; Giljohann, D. A.; Thaxton, C. S.; Lytton-Jean, A. K.; Han, M. S.; Mirkin, C. A. Oligonucleotide-Modified Gold Nanoparticles for Intracellular Gene Regulation Science 2006, 312, 1027 1030
    13. 13
      Lytton-Jean, A. K.; Mirkin, C. A. A Thermodynamic Investigation into the Binding Properties of DNA Functionalized Gold Nanoparticle Probes and Molecular Fluorophore Probes J. Am. Chem. Soc. 2005, 127, 12754 12755
    14. 14
      Jensen, S. A.; Day, E. S.; Ko, C. H.; Hurley, L. A.; Luciano, J. P.; Kouri, F. M.; Merkel, T. J.; Luthi, A. J.; Patel, P. C.; Cutler, J. I. Spherical Nucleic Acid Nanoparticle Conjugates as an RNAi-Based Therapy for Glioblastoma Sci. Transl. Med. 2013, 5, 209ra152
    15. 15
      Zheng, D.; Giljohann, D. A.; Chen, D. L.; Massich, M. D.; Wang, X.-Q.; Iordanov, H.; Mirkin, C. A.; Paller, A. S. Topical Delivery of siRNA-Based Spherical Nucleic Acid Nanoparticle Conjugates for Gene Regulation Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 11975 11980
    16. 16
      Seferos, D. S.; Giljohann, D. A.; Hill, H. D.; Prigodich, A. E.; Mirkin, C. A. Nano-Flares: Probes for Transfection and mRNA Detection in Living Cells J. Am. Chem. Soc. 2007, 129, 15477 15479
    17. 17
      Halo, T. L.; McMahon, K. M.; Angeloni, N. L.; Xu, Y.; Wang, W.; Chinen, A. B.; Malin, D.; Strekalova, E.; Cryns, V. L.; Cheng, C. NanoFlares for the Detection, Isolation, and Culture of Live Tumor Cells from Human Blood Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 17104 17109
    18. 18
      Radovic-Moreno, A. F.; Chernyak, N.; Mader, C. C.; Nallagatla, S.; Kang, R. S.; Hao, L.; Walker, D. A.; Halo, T. L.; Merkel, T. J.; Rische, C. H. Immunomodulatory Spherical Nucleic Acids Proc. Natl. Acad. Sci. U.S.A. 2015, 112, 3892 3897
    19. 19
      Yin, H.; Kanasty, R. L.; Eltoukhy, A. A.; Vegas, A. J.; Dorkin, J. R.; Anderson, D. G. Non-viral Vectors for Gene-Based Therapy Nat. Rev. Genet. 2014, 15, 541 555
    20. 20
      Sahay, G.; Querbes, W.; Alabi, C.; Eltoukhy, A.; Sarkar, S.; Zurenko, C.; Karagiannis, E.; Love, K.; Chen, D.; Zoncu, R. Efficiency of siRNA Delivery by Lipid Nanoparticles Is Limited by Endocytic Recycling Nat. Biotechnol. 2013, 31, 653 658
    21. 21
      Lovell, J. F.; Jin, C. S.; Huynh, E.; Jin, H.; Kim, C.; Rubinstein, J. L.; Chan, W. C.; Cao, W.; Wang, L. V.; Zheng, G. Porphysome Nanovesicles Generated by Porphyrin Bilayers for Use as Multimodal Biophotonic Contrast Agents Nat. Mater. 2011, 10, 324 332
    22. 22
      Dahlman, J. E.; Barnes, C.; Khan, O. F.; Thiriot, A.; Jhunjunwala, S.; Shaw, T. E.; Xing, Y.; Sager, H. B.; Sahay, G.; Speciner, L. In Vivo Endothelial siRNA Delivery Using Polymeric Nanoparticles with Low Molecular Weight Nat. Nanotechnol. 2014, 9, 648 655
    23. 23
      Dong, Y.; Love, K. T.; Dorkin, J. R.; Sirirungruang, S.; Zhang, Y.; Chen, D.; Bogorad, R. L.; Yin, H.; Chen, Y.; Vegas, A. J. Lipopeptide Nanoparticles for Potent and Selective siRNA Delivery in Rodents and Nonhuman Primates Proc. Natl. Acad. Sci. U.S.A. 2014, 111, 3955 3960
    24. 24
      Yin, H.; Xue, W.; Chen, S.; Bogorad, R. L.; Benedetti, E.; Grompe, M.; Koteliansky, V.; Sharp, P. A.; Jacks, T.; Anderson, D. G. Genome Editing with Cas9 in Adult Mice Corrects a Disease Mutation and Phenotype Nat. Biotechnol. 2014, 32, 551 553
    25. 25
      Shi, J.; Xiao, Z.; Votruba, A. R.; Vilos, C.; Farokhzad, O. C. Differentially Charged Hollow Core/Shell Lipid–Polymer–Lipid Hybrid Nanoparticles for Small Interfering RNA Delivery Angew. Chem., Int. Ed. 2011, 123, 7165 7169
    26. 26
      Shi, J.; Xu, Y.; Xu, X.; Zhu, X.; Pridgen, E.; Wu, J.; Votruba, A. R.; Swami, A.; Zetter, B. R.; Farokhzad, O. C. Hybrid Lipid–Polymer Nanoparticles for Sustained siRNA Delivery and Gene Silencing Nanomedicine 2014, 10, 897 900
    27. 27
      Zhu, X.; Xu, Y.; Solis, L. M.; Tao, W.; Wang, L.; Behrens, C.; Xu, X.; Zhao, L.; Liu, D.; Wu, J. Long-Circulating siRNA Nanoparticles for Validating Prohibitin1-Targeted Non-Small Cell Lung Cancer Treatment Proc. Natl. Acad. Sci. U.S.A. 2015,  DOI: 10.1073/pnas.1505629112
    28. 28
      Cabral, H.; Kataoka, K. Progress of Drug-Loaded Polymeric Micelles into Clinical Studies J. Controlled Release 2014, 190, 465 476
    29. 29
      Miura, Y.; Takenaka, T.; Toh, K.; Wu, S.; Nishihara, H.; Kano, M. R.; Ino, Y.; Nomoto, T.; Matsumoto, Y.; Koyama, H. Cyclic RGD-Linked Polymeric Micelles for Targeted Delivery of Platinum Anticancer Drugs to Glioblastoma through the Blood–Brain Tumor Barrier ACS Nano 2013, 7, 8583 8592
    30. 30
      Ahn, J.; Miura, Y.; Yamada, N.; Chida, T.; Liu, X.; Kim, A.; Sato, R.; Tsumura, R.; Koga, Y.; Yasunaga, M. Antibody Fragment-Conjugated Polymeric Micelles Incorporating Platinum Drugs for Targeted Therapy of Pancreatic Cancer Biomaterials 2015, 39, 23 30
    31. 31
      Kabanov, A. V.; Chekhonin, V.; Alakhov, V. Y.; Batrakova, E.; Lebedev, A.; Melik-Nubarov, N.; Arzhakov, S.; Levashov, A.; Morozov, G.; Severin, E. The Neuroleptic Activity of Haloperidol Increases after Its Solubilization in Surfactant Micelles: Micelles as Microcontainers for Drug Targeting FEBS Lett. 1989, 258, 343 345
    32. 32
      Alakhova, D. Y.; Kabanov, A. V. Pluronics and MDR Reversal: An Update Mol. Pharmaceutics 2014, 11, 2566 2578
    33. 33
      Valle, J. W.; Armstrong, A.; Newman, C.; Alakhov, V.; Pietrzynski, G.; Brewer, J.; Campbell, S.; Corrie, P.; Rowinsky, E. K.; Ranson, M. A Phase 2 Study of SP1049C, Doxorubicin in P-Glycoprotein-Targeting Pluronics, in Patients with Advanced Adenocarcinoma of the Esophagus and Gastroesophageal Junction Invest. New Drugs 2011, 29, 1029 1037
    34. 34
      Han, Y.; He, Z.; Schulz, A.; Bronich, T. K.; Jordan, R.; Luxenhofer, R.; Kabanov, A. V. Synergistic Combinations of Multiple Chemotherapeutic Agents in High Capacity Poly(2-oxazoline) Micelles Mol. Pharmaceutics 2012, 9, 2302 2313
    35. 35
      He, Z.; Schulz, A.; Wan, X.; Seitz, J.; Bludau, H.; Alakhova, D. Y.; Darr, D. B.; Perou, C. M.; Jordan, R.; Ojima, I. Poly(2-oxazoline) Based Micelles with High Capacity for 3rd Generation Taxoids: Preparation, In Vitro and In Vivo Evaluation J. Controlled Release 2015, 208, 67 75
    36. 36
      Natarajan, J. V.; Ang, M.; Darwitan, A.; Chattopadhyay, S.; Wong, T. T.; Venkatraman, S. S. Nanomedicine for Glaucoma: Liposomes Provide Sustained Release of Latanoprost in the Eye Int. J. Nanomed. 2012, 7, 123 131
    37. 37
      Natarajan, J. V.; Darwitan, A.; Barathi, V. A.; Ang, M.; Htoon, H. M.; Boey, F.; Tam, K. C.; Wong, T. T.; Venkatraman, S. S. Sustained Drug Release in Nanomedicine: A Long-Acting Nanocarrier-Based Formulation for Glaucoma ACS Nano 2014, 8, 419 429
    38. 38
      Tan, Y. F.; Mundargi, R. C.; Chen, M. H. A.; Lessig, J.; Neu, B.; Venkatraman, S. S.; Wong, T. T. Layer-by-Layer Nanoparticles as an Efficient siRNA Delivery Vehicle for SPARC Silencing Small 2014, 10, 1790 1798
    39. 39
      Huang, H.; Pierstorff, E.; Osawa, E.; Ho, D. Active Nanodiamond Hydrogels for Chemotherapeutic Delivery Nano Lett. 2007, 7, 3305 3314
    40. 40
      Manus, L. M.; Mastarone, D. J.; Waters, E. A.; Zhang, X.-Q.; Schultz-Sikma, E. A.; MacRenaris, K. W.; Ho, D.; Meade, T. J. Gd(III)-Nanodiamond Conjugates for MRI Contrast Enhancement Nano Lett. 2010, 10, 484 489
    41. 41
      Chow, E. K.; Zhang, X.-Q.; Chen, M.; Lam, R.; Robinson, E.; Huang, H.; Schaffer, D.; Osawa, E.; Goga, A.; Ho, D. Nanodiamond Therapeutic Delivery Agents Mediate Enhanced Chemoresistant Tumor Treatment Sci. Transl. Med. 2011, 3, 73ra21
    42. 42
      Moore, L.; Chow, E. K.-H.; Osawa, E.; Bishop, J. M.; Ho, D. Diamond-Lipid Hybrids Enhance Chemotherapeutic Tolerance and Mediate Tumor Regression Adv. Mater. 2013, 25, 3532 3541
    43. 43
      Wang, X.; Low, X. C.; Hou, W.; Abdullah, L. N.; Toh, T. B.; Mohd Abdul Rashid, M.; Ho, D.; Chow, E. K.-H. Epirubicin-Adsorbed Nanodiamonds Kill Chemoresistant Hepatic Cancer Stem Cells ACS Nano 2014, 8, 12151 12166
    44. 44
      Wang, H.; Lee, D.-K.; Chen, K.-Y.; Chen, J.-Y.; Zhang, K.; Silva, A.; Ho, C.-M.; Ho, D. Mechanism-Independent Optimization of Combinatorial Nanodiamond and Unmodified Drug Delivery Using a Phenotypically Driven Platform Technology ACS Nano 2015, 9, 3332 3344
    45. 45
      Paithankar, D.; Hwang, B. H.; Munavalli, G.; Kauvar, A.; Lloyd, J.; Blomgren, R.; Faupel, L.; Meyer, T.; Mitragotri, S. Ultrasonic Delivery of Silica–Gold Nanoshells for Photothermolysis of Sebaceous Glands in Humans: Nanotechnology from the Bench to Clinic J. Controlled Release 2015, 206, 30 36
    46. 46
      Tezel, A.; Mitragotri, S. Interactions of Inertial Cavitation Bubbles with Stratum Corneum Lipid Bilayers during Low-Frequency Sonophoresis Biophys. J. 2003, 85, 3502 3512
    47. 47
      Chiappini, C.; Martinez, J. O.; De Rosa, E.; Almeida, C. S.; Tasciotti, E.; Stevens, M. M. Biodegradable Nanoneedles for Localized Delivery of Nanoparticles in Vivo: Exploring the Biointerface ACS Nano 2015, 9, 5500 5509
    48. 48
      Chiappini, C.; De Rosa, E.; Martinez, J.; Liu, X.; Steele, J.; Stevens, M.; Tasciotti, E. Biodegradable Silicon Nanoneedles Delivering Nucleic Acids Intracellularly Induce Localized In Vivo Neovascularization Nat. Mater. 2015, 14, 532 539
    49. 49
      De La Rica, R.; Stevens, M. M. Plasmonic ELISA for the Ultrasensitive Detection of Disease Biomarkers with the Naked Eye Nat. Nanotechnol. 2012, 7, 821 824
    50. 50
      Chapman, R.; Lin, Y.; Burnapp, M.; Bentham, A.; Hillier, D.; Zabron, A.; Khan, S.; Tyreman, M.; Stevens, M. M. Multivalent Nanoparticle Networks Enable Point-of-Care Detection of Human Phospholipase-A2 in Serum ACS Nano 2015, 9, 2565 2573
    51. 51
      Dewitte, H.; Verbeke, R.; Breckpot, K.; De Smedt, S. C.; Lentacker, I. Nanoparticle Design To Induce Tumor Immunity and Challenge the Suppressive Tumor Microenvironment Nano Today 2014, 9, 743 758
    52. 52
      Gough, D. R.; Cotter, T. G. Hydrogen Peroxide: A Jekyll and Hyde Signalling Molecule Cell Death Dis. 2011, 2, e213
    53. 53
      Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free Radicals, Antioxidants and Functional Foods: Impact on Human Health Pharmacogn. Rev. 2010, 4, 118 126
    54. 54
      Sun, X.; Guo, S.; Liu, Y.; Sun, S. Dumbbell-like PtPd–Fe3O4 Nanoparticles for Enhanced Electrochemical Detection of H2O2 Nano Lett. 2012, 12, 4859 4863
    55. 55
      Zhu, H.; Sigdel, A.; Zhang, S.; Su, D.; Xi, Z.; Li, Q.; Sun, S. Core/Shell Au/MnO Nanoparticles Prepared through Controlled Oxidation of AuMn as an Electrocatalyst for Sensitive H2O2 Detection Angew. Chem., Int. Ed. 2014, 53, 12508 12512
    56. 56
      Sun, X.; Cai, W.; Chen, X. Positron Emission Tomography Imaging Using Radiolabeled Inorganic Nanomaterials Acc. Chem. Res. 2015, 48, 286 294
    57. 57
      Liu, X.; Wang, S. Three-Dimensional Nano-Biointerface as a New Platform for Guiding Cell Fate Chem. Soc. Rev. 2014, 43, 2385 2401
    58. 58
      Ma, L.; Yang, G.; Wang, N.; Zhang, P. C.; Guo, F. Y.; Meng, J. X.; Zhang, F. L.; Hu, Z. J.; Wang, S. T.; Zhao, Y. Trap Effect of Three-Dimensional Fibers Network for High Efficient Cancer-Cell Capture Adv. Healthcare Mater. 2015, 4, 838 843
    59. 59
      Zhang, P.; Chen, L.; Xu, T.; Liu, H.; Liu, X.; Meng, J.; Yang, G.; Jiang, L.; Wang, S. Programmable Fractal Nanostructured Interfaces for Specific Recognition and Electrochemical Release of Cancer Cells Adv. Mater. 2013, 25, 3566 3570
    60. 60
      Liu, X.; Chen, L.; Liu, H.; Yang, G.; Zhang, P.; Han, D.; Wang, S.; Jiang, L. Bio-Inspired Soft Polystyrene Nanotube Substrate for Rapid and Highly Efficient Breast Cancer Cell Capture NPG Asia Mater. 2013, 5, e63
    61. 61
      Liu, H.; Liu, X.; Meng, J.; Zhang, P.; Yang, G.; Su, B.; Sun, K.; Chen, L.; Han, D.; Wang, S.; Jiang, L. Hydrophobic Interaction-Mediated Capture and Release of Cancer Cells on Thermoresponsive Nanostructured Surfaces Adv. Mater. 2013, 25, 922 927
    62. 62
      Liu, H. L.; Li, Y. Y.; Sun, K.; Fan, J. B.; Zhang, P. C.; Meng, J. X.; Wang, S. T.; Jiang, L. Dual-Responsive Surfaces Modified with Phenylboronic Acid-Containing Polymer Brush To Reversibly Capture and Release Cancer Cells J. Am. Chem. Soc. 2013, 135, 7603 7609
    63. 63
      Wang, S.; Liu, K.; Liu, J.; Yu, Z. T. F.; Xu, X.; Zhao, L.; Lee, T.; Lee, E. K.; Reiss, J.; Lee, Y.-K. Highly Efficient Capture of Circulating Tumor Cells by Using Nanostructured Silicon Substrates with Integrated Chaotic Micromixers Angew. Chem., Int. Ed. 2011, 50, 3084 3088
    64. 64
      Duan, R.; Zuo, X.; Wang, S.; Quan, X.; Chen, D.; Chen, Z.; Jiang, L.; Fan, C.; Xia, F. Lab in a Tube: Ultrasensitive Detection of MicroRNAs at the Single-Cell Level and in Breast Cancer Patients Using Quadratic Isothermal Amplification J. Am. Chem. Soc. 2013, 135, 4604 4607
    65. 65
      Liu, W.; Thomopoulos, S.; Xia, Y. Electrospun Nanofibers for Regenerative Medicine Adv. Healthcare Mater. 2012, 1, 10 25
    66. 66
      Xie, J.; Liu, W.; MacEwan, M. R.; Bridgman, P. C.; Xia, Y. Neurite Outgrowth on Electrospun Nanofibers with Uniaxial Alignment: The Effects of Fiber Density, Surface Coating, and Supporting Substrate ACS Nano 2014, 8, 1878 1885
    67. 67
      Smith, L.; Xia, Y.; Galatz, L. M.; Genin, G. M.; Thomopoulos, S. Tissue-Engineering Strategies for the Tendon/Ligament-to-Bone Insertion Connect. Tissue Res. 2012, 53, 95 105
    68. 68
      Xie, J.; MacEwan, M. R.; Ray, W. Z.; Liu, W.; Siewe, D. Y.; Xia, Y. Radially Aligned, Electrospun Nanofibers as Dural Substitutes for Wound Closure and Tissue Regeneration Applications ACS Nano 2010, 4, 5027 5036
    69. 69
      Lang, N.; Pereira, M. J.; Lee, Y.; Friehs, I.; Vasilyev, N. V.; Feins, E. N.; Ablasser, K.; O’Cearbhaill, E. D.; Xu, C.; Fabozzo, A. A Blood-Resistant Surgical Glue for Minimally Invasive Repair of Vessels and Heart Defects Sci. Transl. Med. 2014, 6, 218ra6
    70. 70
      Yang, S. Y.; O’Cearbhaill, E. D.; Sisk, G. C.; Park, K. M.; Cho, W. K.; Villiger, M.; Bouma, B. E.; Pomahac, B.; Karp, J. M. A Bio-Inspired Swellable Microneedle Adhesive for Mechanical Interlocking with Tissue Nat. Commun. 2013, 4, 1702
    71. 71
      Cho, W. K.; Ankrum, J. A.; Guo, D.; Chester, S. A.; Yang, S. Y.; Kashyap, A.; Campbell, G. A.; Wood, R. J.; Rijal, R. K.; Karnik, R. Microstructured Barbs on the North American Porcupine Quill Enable Easy Tissue Penetration and Difficult Removal Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 21289 21294
    72. 72
      Liu, Y.; Lim, J.; Teoh, S.-H. Review: Development of Clinically Relevant Scaffolds for Vascularised Bone Tissue Engineering Biotechnol. Adv. 2013, 31, 688 705
    73. 73
      Lim, J.; Chong, M. S. K.; Chan, J. K. Y.; Teoh, S. H. Polymer Powder Processing of Cryomilled Polycaprolactone for Solvent-Free Generation of Homogeneous Bioactive Tissue Engineering Scaffolds Small 2014, 10, 2495 2502
    74. 74
      Peer, D.; Karp, J. M.; Hong, S.; Farokhzad, O. C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for Cancer Therapy Nat. Nanotechnol. 2007, 2, 751 760
    75. 75
      Chow, E. K.-H.; Ho, D. Cancer Nanomedicine: From Drug Delivery to Imaging Sci. Transl. Med. 2013, 5, 216rv4
    76. 76
      Godwin, H.; Nameth, C.; Avery, D.; Bergeson, L. L.; Bernard, D.; Beryt, E.; Boyes, W.; Brown, S.; Clippinger, A. J.; Cohen, Y. Nanomaterial Categorization for Assessing Risk Potential to Facilitate Regulatory Decision-Making ACS Nano 2015, 9, 3409 3417
    77. 77
      Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-Responsive Nanocarriers for Drug Delivery Nat. Mater. 2013, 12, 991 1003
    78. 78
      Pacardo, D. B.; Ligler, F. S.; Gu, Z. Programmable Nanomedicine: Synergistic and Sequential Drug Delivery Systems Nanoscale 2015, 7, 3381 3391
    79. 79
      Lu, Y.; Sun, W.; Gu, Z. Stimuli-Responsive Nanomaterials for Therapeutic Protein Delivery J. Controlled Release 2014, 194, 1 19
    80. 80
      Gu, Z.; Aimetti, A. A.; Wang, Q.; Dang, T. T.; Zhang, Y.; Veiseh, O.; Cheng, H.; Langer, R. S.; Anderson, D. G. Injectable Nano-network for Glucose-Mediated Insulin Delivery ACS Nano 2013, 7, 4194 4201
    81. 81
      Mo, R.; Jiang, T.; DiSanto, R.; Tai, W.; Gu, Z. ATP-Triggered Anticancer Drug Delivery Nat. Commun. 2014, 5, 3364
    82. 82
      Gu, Z.; Dang, T. T.; Ma, M.; Tang, B. C.; Cheng, H.; Jiang, S.; Dong, Y.; Zhang, Y.; Anderson, D. G. Glucose-Responsive Microgels Integrated with Enzyme Nanocapsules for Closed-Loop Insulin Delivery ACS Nano 2013, 7, 6758 6766
    83. 83
      Godwin, H.; Nameth, C.; Avery, D.; Bergeson, L. L.; Bernard, D.; Beryt, E.; Boyes, W.; Brown, S.; Clippinger, A. J.; Cohen, Y. Nanomaterial Categorization for Assessing Risk Potential to Facilitate Regulatory Decision-Making ACS Nano 2015, 9, 3409 3417
    84. 84
      Mitragotri, S.; Burke, P. A.; Langer, R. Overcoming the Challenges in Administering Biopharmaceuticals: Formulation and Delivery Strategies Nat. Rev. Drug Discovery 2014, 13, 655 672
    85. 85
      Chertok, B.; Webber, M. J.; Succi, M. D.; Langer, R. Drug Delivery Interfaces in the 21st Century: From Science Fiction Ideas to Viable Technologies Mol. Pharmaceutics 2013, 10, 3531 3543
    86. 86
      Hammond, P. T. A Growing Place for Nano in Medicine ACS Nano 2014, 8, 7551 7552
    87. 87
      Khademhosseini, A.; Peppas, N. A. Micro- and Nanoengineering of Biomaterials for Healthcare Applications Adv Healthc Mater 2013, 2, 10 12

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect