Gold nanoparticles in chemotherapy

Last updated
Gold nanoparticles Gold Nanoparticles.jpg
Gold nanoparticles

Gold nanoparticles in chemotherapy and radiotherapy is the use of colloidal gold in therapeutic treatments, often for cancer or arthritis. Gold nanoparticle technology shows promise in the advancement of cancer treatments. Some of the properties that gold nanoparticles possess, such as small size, non-toxicity and non-immunogenicity make these molecules useful candidates for targeted drug delivery systems. With tumor-targeting delivery vectors becoming smaller, the ability to by-pass the natural barriers and obstacles of the body becomes more probable. To increase specificity and likelihood of drug delivery, tumor specific ligands may be grafted onto the particles along with the chemotherapeutic drug molecules, to allow these molecules to circulate throughout the tumor without being redistributed into the body.

Contents

Physical properties

Solutions of gold nanoparticles of various sizes. The size difference causes the difference in colors. Gold255.jpg
Solutions of gold nanoparticles of various sizes. The size difference causes the difference in colors.

Size

Gold nanoparticles range in size depending on which therapy they are being used for. In photothermal cancer therapy, many gold nanoparticle molecules are used in each test and they must all be uniform in size. Including PEG coating, the nanoparticles measured to be ~130 nm in diameter. [1] Gold nanoparticles that act as drug delivery systems in conjugation with chemotherapeutic drugs typically range in size from 10 to 100 nm. [2]

Surface area plays a very important role in drug delivery and per mg of gold, as diameters decrease, the surface areas needed to transport drugs increase to the point where a single 1mL volume of 1.8 nm spherical gold nanoparticles have the same surface area as a cell phone. [3]

Drug vectorization requires greater specificity, and are synthesized within the single digit measurements ranging from 3-7 nm.[ citation needed ]

Antibacterial treatments are testing different sizes for cell type targeting; 10, 20 and 40 nm. [4]

Color

Due to the ability to tune the size and absorption of AuNPs, these molecules can vary in the colors they emit. Colors of AuNP solutions typically range from wine red to pale blue. These colors play a necessary role in the synthesis of AuNPs as indicators of reduction. [5] The color of AuNPs can be modified by the application of pressure which reddish the nanoparticles. [6]

Synthesis

For more on synthesis of AuNPs for medical use, see Colloidal gold

Other synthesis may include cell type targeting. A tumor consists of a multitude of cell types, and thus targeting a single type of cell is ineffective and potentially dangerous. At most, this type of targeting would only have a minor effect on killing the tumor. Tumors are constantly changing and thus phenotype targeting is rendered useless. Two main problems persist: how to get to the target and how to destroy a variety of cells.[ medical citation needed ]

Treatments

Photothermal cancer therapy

A direct method of accessing and destroying tumour cells can be accomplished by photothermal cancer therapy or photodynamic therapy (PDT). This procedure is known to treat small tumours that are difficult to access and avoids the drawbacks (adverse effects) of conventional methods, including the unnecessary destruction of healthy tissues. [7] The cells are destroyed by exposure to light, rupturing membranes causing the release of digestive enzymes. AuNPs have high absorption cross sections requiring only minimal input of irradiation energy. Human breast carcinoma cells infused with metal nanoparticles in vitro have been shown to have an increase in morbidity with exposure to near infrared (NIR). [7] Short term exposure in vivo (4–6 minutes) to NIR had undergone the same effect. Hirsch et al observed that extreme heating in tumours would cause irreversible tissue damage including coagulation, cell shrinkage and loss of nuclear straining. Results of their in vivo nanoshell therapy of mice revealed penetration of the tumor ~5mm.The metal particles were tuned to high absorption and scattering, resulting in effective conversion of light into heat covering a large surface area. [8] The El-Sayed group studied AuNP effects in vitro and in vivo. They determined that the NIR wavelengths were converted into heat on the picosecond timescale, allowing for short exposure of CW to minimize possible exposure to healthy cells. In vitro, photothermal therapy was used in oral epithelial cell lines, (HSC 313 and HOC 3 Clone 8) and one benign epithelial cell line (HaCaT). El-Sayed et al found that the malignant cells that had undergone incubation in AuNPs conjugated with anti-epithelial growth factor receptor (EGFR) required half the energy to destroy a cell than a benign cell. Their material included gold coated silica nanoshells that could selectively absorb NIR waves. The particles were tuned by varying the thickness of the Au shell and changing the size of the silica core. In exposing these particles to NIR, the efficacy of Au was measured through the decrease of EFGR in oral squamous carcinoma cells. [8] There are various biotechnological advances for in vivo delivery of drugs. To effectively target the malignant cells, the AuNPs were conjugated by polyethylene glycol, a process known as PEGylation. This masks the foreign particles from the immune system such that it arrives at its destination and increases circulation time in the system. Antibody conjugation lines the surface of the nanoparticle with cell markers to limit spread only to malignant cells. [8] In vivo testing of mice that developed murine colon carcinoma tumour cells. They were injected with the solution of AuNPs that were allowed to spread after 6 hours. Surrounding cells were swabbed with PEG and exposed to laser treatment for detection of abnormal heating indicating areas where Au nanoshells may have gathered. The injected area was also swabbed with PEG to maximize light penetration. [8]

Despite the unquestionable success of gold nanorods or nanoshells as photothermal agents in preclinical research, they have yet to obtain the approval for clinical use because their size is above the renal excretion threshold. [9] In 2019, the first NIR-absorbing plasmonic ultrasmall-in-nano architecture has been reported, and jointly combine: (i) an efficient photothermal conversion suitable for multiple hyperthermia treatments, and (ii) renal excretion of the building blocks after the therapeutic action. [10]

Radiofrequency therapy

X-ray radiography procedures involves the diagnosis of cancer cells through the process of image acquisition. [11] These techniques rely on the absorption of x-rays on the exposed tissue in order to improve image quality. In certain radiological procedures such as Radiofrequency therapy, a contrast agent is injected into the targeted cancer tissue and result in increased x-ray attenuation.[ medical citation needed ]

Radiofrequency therapy treatment involves the destruction of tumor cancer tissue cells through the differential heating of cancer tissue by radio-frequency diathermy. [12] This differential heating is a result of the blood supply in the body carrying away the heat and cooling the heated tissue.

Gold nanoparticles are excellent absorbers of x-rays, due to its high atomic number of 197Au. This allows for a higher mass of the element, providing for a greater area of x-ray absorption. By acting as a contrast agent and injected into cancerous tumor cells, it would result in a higher dose of the cancerous tissue being exposed during radiotherapy treatment. [13] Additionally gold nanoparticles are more efficiently removed from cells of healthy tissue, in comparison with cancer cells - a feature that makes them a promising radiosensitizers [14]

Angiogenesis therapy

Angiogenesis is a process involving the formation of new blood vessels from pre-existing vessels. It involves the degradation of the extracellular matrix, activation, migration, proliferation, and differentiation of endothelial cells into vessels. It is said to play a large part in the growth and spread of cancer cells. [15]

The process of angiogenesis involves the use of both promoters and inhibitors, balancing the process by only forming new blood vessels when needed. Examples of promoters include Vascular Endothelial Growth Factor (VEGF) and fibroblast growth factor (FGF) Examples of inhibitors include Vascular Endothelial Growth Factor Receptor 1, etc.

Tumor progression occurs as a result of the transition from a tumor in the dormant proliferation stage to the active stage as a result of oxygen and nutrients. This active stage leads to a state of cellular hypoxia, which causes an increased regulation of pro-angiogenesis proteins such as VEGF. This results in the spreading of inflammatory proteins and cancer cells alongside the newly created blood vessels. [16]

AuNPs have the ability to inhibit angiogenesis by directly coordinating to heparin binding growth factors. They inhibit phosphorylation of proteins responsible for angiogenesis in a dose dependent matter. At concentrations 335-670 nM, almost complete inhibition of phosphorylation was observed. [7] As a consequence of angiogenesis, rheumatoid arthritis has been found to develop due to the greater ability to spread inflammatory proteins. Through the inhibition of angiogenesis, the reduction of rheumatoid arthritis is prevalent. [7] In addition, angiogenic inhibitors have a critical limitation due to the instability of biological conditions and high dosage required. To counter this, an emerging strategy for the development of therapies targeting tumor-associated angiogenesis through the use of nanotechnology and anti-angiogenic agents was developed, known as anti-angiogenic therapy. This approach solved the limitation instability by speeding up the delivery of angiogenesis inhibitors. [16]

Gold nanoparticles display anti-angiogenic properties by inhibiting the function of pro-angiogenic heparin-binding growth factors (HG – GFs), with prime examples being the vascular endothelial growth factor 165 (VEGF165) and the basic fibroblast growth factor (bFGF) - both of which are pro-angiogenic promoters. Studies by Rochelle R. Arvizo, et al. have shown that the use of AuNPs of various size and surface charge plays an important role in its inhibitory effects. [17]

In today’s biological fields, the use of nanotechnology has allowed for the indirect use of AuNPs to deliver DNA to mammalian cells; thereby reducing tumor agents and increasing efficiency of electron transfer by modulating the activity of glucose oxidase. Current ongoing research by the Mayo Clinic laboratories includes the examination of AuNPs as messengers to deliver reagents capable of manipulating the angiogenic response in vivo. [18]

Current angiogenic inhibitors used today which are approved by the USFDA to treat cancer is Ayastin, Nexavar, Sutent and Affinitor. [15]

Anti-bacterial therapy

Gold nanoparticles are used as bacteria targeting particles in antibacterial therapy. The therapy targets bacteria with light absorbing gold nanoparticles (10 nm, 20 nm, 40 nm) conjugated with specific antibodies, thus selectively kill bacteria using laser. [4]

Studies has shown the effectiveness of this method on killing Staphylococcus aureus, which is significant human pathogen responsible for a wide range of diseases such as skin and wound infections, toxic shock syndrome, septic arthritis, endocarditis, and osteomyelitis. In this system, the bacteria damage is caused by inducing strong laser which leads to overheating effects accompanied by the bubble-formation phenomena around clustered gold nanoparticles.[ medical citation needed ]

The selective targeting of S. aureus was performed using a monoclonal antibody to one of the major surface-clustered proteins, protein A (spa), which is linked to the peptidoglycan portion of the cell wall. Monoclonal antibodies ensure the targeting of the specific cell, which is essential to this mechanism. Killing efficiency depends on local overheating effects accompanied by the bubble-formation phenomena, the bubble formation would enhance the PT killing effect.Better heating efficiency results from an enhanced ability to confine the nanosecond laser-pulse within the nanocluster’s size. Overlapping of bubbles from different nanoparticles within the nanoclusters decreases the bubble-formation threshold. An increase in the cluster’s average local absorption and its potential redshifting (from 525 nm for a single gold spherical nanoparticle to 700–800 nm for nanoclusters) in response to plasmon-plasmon resonance. [4]

Drug vectorization

Another way in which AuNPs can be used in cancer therapy is as agents for targeted drug delivery. Research shows that AuNPs can be easily functionalized and conjugated with a variety of molecules, including chemotherapeutic drugs such as Doxorubicin. [19] [20] One major complication with the current methods of treating cancer with chemotherapy is that treatment is not optimized to specifically target cancer cells and the widespread distribution of chemotherapeutic drugs throughout the body can cause harmful side effects such as naseua, hair loss, and cardiotoxicity. [20] Since many of the characteristics of AuNPs allow them to target cancer cells specifically and accumulate within tumor cells, these molecules can act as tumor-targeting drug delivery systems. Once within the tumor microenvironment, these complexes dissociate and release the chemotherapeutic, allowing the drug to take effect and eventually cause apoptosis.

Gold nanoparticles have their advantages in drug vectorization. They can pack several different sizes and types of dendrimers and several different types of ligands in order to effectively treat different types of cancers. For example, research shows that 80~90% of breast cancer’s tumor cells have estrogen receptors [21] and 60~70% of prostate cancer’s tumor cells have androgen receptors. [22] These significant amount of hormone receptors play a role in intermolecular actions. This role is now used by targeting and therapeutic ligands on gold nanoparticles to target tissue-selective anti-tumor drug delivery. In order to have multiple targeting and therapeutic ligands bind with gold nanoparticles, the gold nanoparticles must first undergo polymer stabilization. Then, anti-estrogen molecules with thiolated PEG are bound to gold nanoparticles via Au-S bonds, forming thiolate protected gold nanoparticles. [23]

PEGylated gold nanoparticles 3418179 ijn-7-4007f2.png
PEGylated gold nanoparticles

Docetaxel is packed into PEGylated gold nanoparticles [24] Docetaxel is an anti-mitotic chemotherapy medicine which showing great performance in clinical trial. [25] Docetaxel was approved by FDA, to treat several different kinds of cancer. i.e. breast cancer(include locally advanced or metastatic). [25]

Market approval

A Pilot Study of AuroLase™ Therapy (gold nano shells) in refractory and/or recurrent tumors of the head and neck was completed in 2009 [lower-alpha 1] and two trials are currently using AuroLase™ therapy for the treatment of primary/metastatic lung cancer [lower-alpha 2] and for prostate cancer. [lower-alpha 3] Other gold nanoparticles on the market are mostly for synthesis of nanoparticle complexes in research. Nanocomposix specializes in the production of various sizes of nanoparticles, controlled by varying the concentrations of reducing reagent and HAuCl4. [26]

Sigma Aldrich offers six different sizes of spherical gold nanoparticles and have developed gold nanourchins for similar usage. The surface causes a red shift in the surface plasmon peak as compared to spherical gold nanoaprticles. [27]

Nanopartz [28] offers gold nanoparticles and gold nanorods for preclinical in vivo therapeutics that have been used extensively in preclinical therapeutics including photothermal hyperthermia and chemotherapeutic drug delivery. The pilot study using the Ntracker [29] gold nanorods was completed in 2012 and was used on seven canines with varying degrees of solid cancer tumors. [30] [31] The results showed significant loading of the gold nanorods after intravenous injection into the cancer tumors and significant heating of the tumors from an external laser. Images are found at [32]

Adverse effects and limitations

Shape

Depending on the shape of the molecule, the absorbance will vary, i.e. spherical particles will absorb wavelengths in the NIR region with a relatively low absorbance compared to long rods. [33] Chan et al observed that 50 nm spherical nanoparticles were taken up more efficiently than both larger and smaller particles of the same shape. In regards to size, the spheres were taken up more efficiently than the rods. [34] Ability of greater uptake of nanoshells into the cell will localize in the perinuclear membrane and accumulate to deliver toxic effects.

Charge

Electrostatic interactions were also investigated by Rotello et al by conjugating AuNPs with anionic and cationic functional groups. Their results showed that toxicity was more established in AUNPs conjugated with cationic functional groups as a consequence of electrostatic interactions with the anionic cell membrane. [35]

Concentration

The concentrations of gold nanoparticles in biological systems for practical usage range from 1-100 nanoparticles per cell. High concentrations may lead to adverse effects for cell structure and function, which may not appear non-toxic in assays but preparation of the particles have been found to produce abnormal effects in the cell. [36] If large concentrations quickly clear the blood vessels, the nanoshells may accumulate in major organs (mainly the liver and spleen). Residual concentrations of these particles were also found in kidneys, lungs, muscle, brain, and bone of mice after 28 days. The concentration of the solution injected intravenously 2.4*1011 nanoshells/mL. Even without complete clearance from the system, the nanoshells did not cause any physiological complications in the mice. [37] Su et al observed a correlation with the concentration of Au3Cu and cell damage. Cells were incubated in concentrations of 0.001 and 200 mg mL−1 Au3Cu. They concluded a 15% cell viability and dose dependent cell damage. Reduction in cell viability was detected in vivo experiments; also related to dosage. [38] Cytotoxicity is not a major concern in the usage of AuNPs, as they localize in the vesicles and cytoplasm as opposed to the nucleus. Thus, no complications spawned due to their aggregation in these parts of the cell. [39]

Heating

Two key factors to consider when irradiating gold nanoparticles in cancer cells are the lattice cooling rate and lattice heat content. The lattice cooling rate is how fast heat in the particle is distributed to its surroundings. If the cooling rate for a particle is too low, the lattice heat content can be increased with moderate energy radiation (40 µJ/fs with 100-fs laser at 800 nm) to the point where gold nanorods can be melted to create spherical nanoparticles which become photothermally inactive. [40] This decomposition has been shown using gold nanorods coated with phosphatidylcholine ligands in HeLa cells using a pulsed laser and were no longer useful for treatment due to their low NIR radiation absorbance. [41] High energy laser pulses have also been shown to fragment nanorods into smaller particles. [40] While these structural changes induced by laser pulses could be used to deactivate the photothermal effects of these particles after treatment, the resulting spherical particles or other particle fragments could lead to complications during or after treatment when gold nanoparticles are used for clinical treatment and imaging of cancer cells. [40] [41]

A limitation of photothermal chemotherapy using gold nanoparticles involves the choice of laser when conducting treatment. Pulsed lasers offer very selective treatment of cancer cells within a small, localized area, but can lead to potential destruction of particles and have a low heating efficiency due to heat lost during the single pulse excitation. [40] Continuous wave lasers have a higher heating efficiency and work better in heating larger areas with lower risk of destroying the nanoparticles being heated. However, treatment with continuous wave lasers are much longer compared to treatment with a pulsed laser. [40] A limitation of photothermal therapy with respect to the laser used is the depth of the tumor being treated. Most lasers used to induce tumor ablation using gold nanoparticles can only reach several centimeters into soft tissue, making it impossible to reach tumors farther in the body. [42] Finding a way to carry out therapy in cells farther into the body without damaging surrounding cells is essential to making this technique viable as a cancer treatment in the future.

Toxicity

Toxic precursors

Studies in human leukemia cells revealed that prolonged exposure in AuNPs did not harm the cells, even at ~100 μM of Au. Rather they reduced the amount of reactive oxygen species in the cell. However, precursors to AuNP synthesis (CTAB and HAuCl4) were found to be toxic at small concentrations (10 μM); free CTAB especially. Studies in HeLa cells by Niidome et al further support this statement by examining the correlation with the removal of excess CTAB [43] [44] and cell viability rose to 90%. [43]

Toxicity of nanoparticles in vivo and in vitro

After using nanoparticles for photothermal therapy, it has been shown in vitro that high concentrations of reactive oxygen species (ROS) are formed within the treated cancer cells. [45] [46] [47] While these species are not of concern to the dead cancer cells, they can cause oxidative stress in surrounding healthy cells if enough ROS are created leading to healthy cell death. [45] [46] This oxidative stress can be passivated using polymers as reducing agents (after degradation of the nanoparticle) and damage from ROS can be reduced using targeted uptake of the nanoparticles to the cancer cells. The mechanism for the oxidative stress caused by nanoparticles in the body is still the subject of study and provides a possible limitation when using gold nanoparticles with radiation within the body. [45] [46] [47]

While there are many in vitro studies of gold nanoparticles used for chemotherapy, in vivo studies are both rare and often report conflicting results. For example, one in vivo study has shown that 13-nm gold nanoparticles circulated in the bloodstream often “accumulate in the liver and spleen and…have long blood circulation times." [48] Also, nanoparticles from 8 to 37 nanometers have been shown to cause abnormal symptoms leading to death in mice due to medical complications in the spleen, liver, and lungs. Yet, other studies have shown that 20 nm gold nanoparticles can pass into the retina without causing any cytotoxic effects and nanoparticles of 13 nm diameter were not toxic in the body. Many argue that these results differ due to different concentrations on nanoparticles used for these experiments and requires further research. [48]

Biosafety and biokinetics investigations on biodegradable ultrasmall-in-nano architectures have demonstrated that gold nanoparticles are able to avoid metal accumulation in organisms through escaping by the renal pathway. [49] [50]

Part of the issue with these studies is the lack of reliable methods for determining the uptake of gold nanoparticles in vivo without examining the tumor site post-mortem. Gold nanoparticle uptake in cells is often carried out by examining the organs of injected mice post-mortem. This technique cannot be replicated during clinical trials, so new methods need to be developed to determine the uptake of cells to avoid higher concentrations of gold nanoparticles in the body leading to toxic effects. [42] One recently suggested method to counter this limitation is radiolabeling. The uptake of thiolated gold nanoparticles has recently been monitored using 111In-labeled polymer shells that surround the gold nanoparticle and shows a possible way around this problem, but these polymer shells can be removed from the particle making a more stable labeling system required for these kinds of studies. [51]

Other uses

The ligand used to decrease aggregation of gold nanorods. Cetyltrimethyl ammonium bromide.svg
The ligand used to decrease aggregation of gold nanorods.

Gold nanoparticles may be used in an indirectly therapeutic way. The issue of angiogenesis describes the formation of new blood vessels, which not only increased spread of cancerous cells, but may proliferate the spread of proteins responsible for rheumatoid arthritis. As AuNPs reduce angiogenesis, rheumatoid arthritis is reduced as a result. [7] Chamberland et al studied the use of anti-TNF conjugated gold nanorods (AuNRs) ex vivo in rat tail joints to reduce the effect of rheumatoid arthritis. They observed the effects of the drug delivery system via PAT technology. The properties of the AuNRs found to be the most efficient had measurements of 45 x 15 nm with an absorption peak of 660 nm. This tuning allowed for better contrast between the targeted areas and intra-articular tissue. Thus, the etanercept conjugated AuNRs were seen to increase the light sensitivity. The imaging technique provides greater opportunities for sensitive in vivo drug tracking in biothechnology. [52]

HIV

Several valences of AuNPs were found to inhibit HIV fusion. 2-nm AuNP-mercaptobenzoic acid were conjugated to a derivative of a known CCR5 antagonist, which is a small molecule that antagonize CCR5 receptor, and CCR5 is commonly used by HIV to enter the cell. The CCR5 antagonist would bind to CCR5, leaving no spots for HIV to bind. This will ultimately lead to an effect that restrict HIV infection. [7]

Hepatitis B

Prepared AuNPs-Hepatitis B virus (HBV) DNA gene probes could be used to detect HBV DNA directly. The detection-visualized fluorescence-based method is highly sensitive, simple, low cost, which could potentially apply to multi-gene detection chips. [7] The probe used here is essentially a biosensor, to specifically detect a certain material. [53]

Tuberculosis

A successful application of the AuNP-nanoprobe colorimetric method to clinical diagnosis reported by Baptista et al. was the sensitive detection in clinical samples of Mycobacterium tuberculosis , the cause of human tuberculosis. [7]

See also

Related Research Articles

Experimental cancer treatments are mainstream medical therapies intended to treat cancer by improving on, supplementing or replacing conventional methods. However, researchers are still trying to determine whether these treatments are safe and effective treatments. Experimental cancer treatments are normally available only to people who participate in formal research programs, which are called clinical trials. Occasionally, a seriously ill person may be able to access an experimental drug through an expanded access program. Some of the treatments have regulatory approval for treating other conditions. Health insurance and publicly funded health care programs normally refuse to pay for experimental cancer treatments.

Nanomedicine is the medical application of nanotechnology. Nanomedicine ranges from the medical applications of nanomaterials and biological devices, to nanoelectronic biosensors, and even possible future applications of molecular nanotechnology such as biological machines. Current problems for nanomedicine involve understanding the issues related to toxicity and environmental impact of nanoscale materials.

<span class="mw-page-title-main">Angiogenesis</span> Blood vessel formation, when new vessels emerge from existing vessels

Angiogenesis is the physiological process through which new blood vessels form from pre-existing vessels, formed in the earlier stage of vasculogenesis. Angiogenesis continues the growth of the vasculature mainly by processes of sprouting and splitting, but processes such as coalescent angiogenesis, vessel elongation and vessel cooption also play a role. Vasculogenesis is the embryonic formation of endothelial cells from mesoderm cell precursors, and from neovascularization, although discussions are not always precise. The first vessels in the developing embryo form through vasculogenesis, after which angiogenesis is responsible for most, if not all, blood vessel growth during development and in disease.

<span class="mw-page-title-main">Nanoshell</span>

A nanoshell, or rather a nanoshell plasmon, is a type of spherical nanoparticle consisting of a dielectric core which is covered by a thin metallic shell. These nanoshells involve a quasiparticle called a plasmon which is a collective excitation or quantum plasma oscillation where the electrons simultaneously oscillate with respect to all the ions.

<span class="mw-page-title-main">Colloidal gold</span> Suspension of gold nanoparticles in a liquid

Colloidal gold is a sol or colloidal suspension of nanoparticles of gold in a fluid, usually water. The colloid is coloured usually either wine red or blue-purple . Due to their optical, electronic, and molecular-recognition properties, gold nanoparticles are the subject of substantial research, with many potential or promised applications in a wide variety of areas, including electron microscopy, electronics, nanotechnology, materials science, and biomedicine.

An angiogenesis inhibitor is a substance that inhibits the growth of new blood vessels (angiogenesis). Some angiogenesis inhibitors are endogenous and a normal part of the body's control and others are obtained exogenously through pharmaceutical drugs or diet.

<span class="mw-page-title-main">Enhanced permeability and retention effect</span>

The enhanced permeability and retention (EPR) effect is a controversial concept by which molecules of certain sizes tend to accumulate in tumor tissue much more than they do in normal tissues. The general explanation that is given for this phenomenon is that, in order for tumor cells to grow quickly, they must stimulate the production of blood vessels. VEGF and other growth factors are involved in cancer angiogenesis. Tumor cell aggregates as small as 150–200 μm, start to become dependent on blood supply carried out by neovasculature for their nutritional and oxygen supply. These newly formed tumor vessels are usually abnormal in form and architecture. They are poorly aligned defective endothelial cells with wide fenestrations, lacking a smooth muscle layer, or innervation with a wider lumen, and impaired functional receptors for angiotensin II. Furthermore, tumor tissues usually lack effective lymphatic drainage. All of these factors lead to abnormal molecular and fluid transport dynamics, especially for macromolecular drugs. This phenomenon is referred to as the "enhanced permeability and retention (EPR) effect" of macromolecules and lipids in solid tumors. The EPR effect is further enhanced by many pathophysiological factors involved in enhancement of the extravasation of macromolecules in solid tumor tissues. For instance, bradykinin, nitric oxide / peroxynitrite, prostaglandins, vascular permeability factor, tumor necrosis factor and others. One factor that leads to the increased retention is the lack of lymphatics around the tumor region which would filter out such particles under normal conditions.

Targeted drug delivery, sometimes called smart drug delivery, is a method of delivering medication to a patient in a manner that increases the concentration of the medication in some parts of the body relative to others. This means of delivery is largely founded on nanomedicine, which plans to employ nanoparticle-mediated drug delivery in order to combat the downfalls of conventional drug delivery. These nanoparticles would be loaded with drugs and targeted to specific parts of the body where there is solely diseased tissue, thereby avoiding interaction with healthy tissue. The goal of a targeted drug delivery system is to prolong, localize, target and have a protected drug interaction with the diseased tissue. The conventional drug delivery system is the absorption of the drug across a biological membrane, whereas the targeted release system releases the drug in a dosage form. The advantages to the targeted release system is the reduction in the frequency of the dosages taken by the patient, having a more uniform effect of the drug, reduction of drug side-effects, and reduced fluctuation in circulating drug levels. The disadvantage of the system is high cost, which makes productivity more difficult, and the reduced ability to adjust the dosages.

Photothermal therapy (PTT) refers to efforts to use electromagnetic radiation for the treatment of various medical conditions, including cancer. This approach is an extension of photodynamic therapy, in which a photosensitizer is excited with specific band light. This activation brings the sensitizer to an excited state where it then releases vibrational energy (heat), which is what kills the targeted cells.

<span class="mw-page-title-main">Arginylglycylaspartic acid</span> Chemical compound

Arginylglycylaspartic acid (RGD) is the most common peptide motif responsible for cell adhesion to the extracellular matrix (ECM), found in species ranging from Drosophila to humans. Cell adhesion proteins called integrins recognize and bind to this sequence, which is found within many matrix proteins, including fibronectin, fibrinogen, vitronectin, osteopontin, and several other adhesive extracellular matrix proteins. The discovery of RGD and elucidation of how RGD binds to integrins has led to the development of a number of drugs and diagnostics, while the peptide itself is used ubiquitously in bioengineering. Depending on the application and the integrin targeted, RGD can be chemically modified or replaced by a similar peptide which promotes cell adhesion.

Multiple layered plasmonics use electronically responsive media to change and manipulate the plasmonic properties of plasmons. The properties typically being manipulated can include the directed scattering of light and light absorption. The use of these to use “changeable” plasmonics is currently undergoing development in the academic community by allowing them to have multiple sets of functions that are dependent on how they are being manipulated or excited. Under these new manipulations, such as multiple layers that respond to different resonant frequencies, their new functions were designed to accomplish multiple objectives in a single application.

Photoimmunotherapy (PIT) is an oncological treatment that combines photodynamic therapy of tumor with immunotherapy treatment. Combining photodynamic therapy with immunotherapy enhances the immunostimulating response and has synergistic effects for metastatic cancer treatment.

Magnetic-plasmonic (bifunctional) nanoparticles (MP-NPs) consist of both optical (plasmonic) and magnetic components and thus, has the functionality of both of these components. These nanoparticles may take many different forms/shapes including dimer, core-shell, janus, nanorod/wire and nanostar. Typically, the magnetic components consists of iron oxide or nickel while the plasmonic component is oftentimes a metal like gold, silver or another plasmonic nanomaterial. Due to the combination of these two materials into a hybrid nanostructure, the material may be interacted with using either light or magnetic fields and so, are commonly used in biomedical applications that require optical sensing/imaging/heating, magnetic stimulation/manipulation, or both of these functionalities. An example utilizing both of these functionalities is that MP-NPs can attach to biological entities and separate them under an external magnetic field while simultaneously detecting their chemical nature via optical sensing. These dual functionalities are especially useful when studying tissues deep within tissue.

Cancer treatments may vary depending on what type of cancer is being targeted, but one challenge remains in all of them: it is incredibly difficult to target without killing good cells. Cancer drugs and therapies all have very low selective toxicity. However, with the help of nanotechnology and RNA silencing, new and better treatments may be on the horizon for certain forms of cancer.

Hadiyah-Nicole Green is an American medical physicist, known for the development of a method using laser-activated nanoparticles as a potential cancer treatment. She is one of 66 black women to earn a Ph.D. in physics in the United States between 1973 and 2012, and is the second black woman and the fourth black person ever to earn a doctoral degree in physics from The University of Alabama at Birmingham.

Combinatorial ablation and immunotherapy is an oncological treatment that combines various tumor-ablation techniques with immunotherapy treatment. Combining ablation therapy of tumors with immunotherapy enhances the immunostimulating response and has synergistic effects for curative metastatic cancer treatment. Various ablative techniques are utilized including cryoablation, radiofrequency ablation, laser ablation, photodynamic ablation, stereotactic radiation therapy, alpha-emitting radiation therapy, hyperthermia therapy, HIFU. Thus, combinatorial ablation of tumors and immunotherapy is a way of achieving an autologous, in-vivo tumor lysate vaccine and treating metastatic disease.

Nanoparticle drug delivery systems are engineered technologies that use nanoparticles for the targeted delivery and controlled release of therapeutic agents. The modern form of a drug delivery system should minimize side-effects and reduce both dosage and dosage frequency. Recently, nanoparticles have aroused attention due to their potential application for effective drug delivery.

Nanomaterials are materials with a size ranging from 1 to 100 nm in at least one dimension. At the nanoscale, material properties become different. These unique properties can be exploited for a variety of applications, including the use of nanoparticles in skincare and cosmetics products.

Protein nanotechnology is a burgeoning field of research that integrates the diverse physicochemical properties of proteins with nanoscale technology. This field assimilated into pharmaceutical research to give rise to a new classification of nanoparticles termed protein nanoparticles (PNPs). PNPs garnered significant interest due to their favorable pharmacokinetic properties such as high biocompatibility, biodegradability, and low toxicity Together, these characteristics have the potential to overcome the challenges encountered with synthetic NPs drug delivery strategies. These existing challenges including low bioavailability, a slow excretion rate, high toxicity, and a costly manufacturing process, will open the door to considerable therapeutic advancements within oncology, theranostics, and clinical translational research.

Nanomaterials have gained significant attention in the field of cancer research and treatment due to their unique properties and potential applications. These materials, typically on the nanoscale, offer several advantages in the fight against cancer.

References

  1. Patrick O'Neal D.; Hirsch L.R.; Halas N.J.; Payne D.; West J.L. (2004). "Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles". Cancer Letters. 209 (2): 171–176. doi:10.1016/j.canlet.2004.02.004. PMID   15159019.
  2. Dreaden, Erik C; Austin, Lauren A; Mackey, Megan A; El-Sayed, Mostafa A (2016-12-06). "Size matters: gold nanoparticles in targeted cancer drug delivery". Therapeutic Delivery. 3 (4): 457–478. doi:10.4155/tde.12.21. ISSN   2041-5990. PMC   3596176 . PMID   22834077.
  3. Gold Nanoparticle Properties and Applications
  4. 1 2 3 Zharov, V.P.; Mercer, K.E.; Galitovskaya, E.N.; Smeltzer, M.S. (January 2006). "Photothermal Nanotherapeutics and Nanodiagnostics for Selective Killing of Bacteria Targeted with Gold Nanoparticles". Biophysical Journal. 90 (2): 619–627. Bibcode:2006BpJ....90..619Z. doi:10.1529/biophysj.105.061895. PMC   1367066 . PMID   16239330.
  5. Turkevich, J.; Stevenson, P.C.; Hillier, J. (May 18, 1951). "A study of the nucleation and growth processes in the synthesis of colloidal gold". Discussions of the Faraday Society. 11: 55–74. doi:10.1039/DF9511100055. S2CID   97664009.
  6. Medeghini, Fabio (October 9, 2018). "High-Pressure Effect on the Optical Extinction of a Single Gold Nanoparticle". ACS Nano. 12 (10): 10310–10316. doi:10.1021/acsnano.8b05539. PMID   30299926. S2CID   52946816.
  7. 1 2 3 4 5 6 7 8 Boisselier, E.; Didier, A. (April 21, 2009). "Gold nanoparticles in nanomedicine: preparations, imaging, diagnostics, therapies and toxicity". Chemical Society Reviews. 38 (6): 1759–1782. doi:10.1039/b806051g. PMID   19587967.
  8. 1 2 3 4 El-Sayed, I.H.; Huang, X.; El-Sayed, M.A. (29 July 2005). "Selective laser photo-thermal therapy of epithelial carcinoma using anti-EGFR antibody conjugated gold nanoparticles". Cancer Letters. 239 (1): 129–135. doi:10.1016/j.canlet.2005.07.035. PMID   16198049.
  9. Cassano, Domenico; Pocoví-Martínez, Salvador; Voliani, Valerio (2018-01-17). "Ultrasmall-in-Nano Approach: Enabling the Translation of Metal Nanomaterials to Clinics". Bioconjugate Chemistry. 29 (1): 4–16. doi: 10.1021/acs.bioconjchem.7b00664 . ISSN   1043-1802. PMID   29186662.
  10. Cassano, Domenico; Santi, Melissa; D’Autilia, Francesca; Mapanao, Ana Katrina; Luin, Stefano; Voliani, Valerio (2019). "Photothermal effect by NIR-responsive excretable ultrasmall-in-nano architectures". Materials Horizons. 6 (3): 531–537. doi: 10.1039/C9MH00096H . ISSN   2051-6347.
  11. Künzel, R.; Okuno, E.; Levenhagen, R.S.; Umisedo, N.K. (14 February 2013). "Evaluation of the X-Ray Absorption by Gold Nanoparticles Solutions". ISRN Nanotechnology. 2013: 1–5. doi: 10.1155/2013/865283 .
  12. LeVeen, H.H.; Ahmed, N.; Piccone, V.A.; Shugaar, S.; Falk, G. (1980). "Radio-Frequency Therapy:Clinical Experience". Annals of the New York Academy of Sciences. 335 (1): 362–71. Bibcode:1980NYASA.335..362L. doi:10.1111/j.1749-6632.1980.tb50761.x. PMID   6931530. S2CID   29358846.
  13. Hainfeld, James F.; Dilmanian, F. Avraham; Slatkin, Daniel N.; Smilowitz, Henry M. (2008). "Radiotherapy enhancement with gold nanoparticles". Journal of Pharmacy and Pharmacology. 60 (8): 977–985. doi: 10.1211/jpp.60.8.0005 . PMID   18644191. S2CID   32861131.
  14. Ivosev, Vladimir; Sanchez, Gloria Jimenez; Haidar, Darine Abi; Bazzi, Rana; Roux, Stephane; Lacombe, Sandrine (8 December 2016). "Import and Export of Gold Nanoparticles: Exchange Rate in Cancer Cells and Fibroblasts". bioRxiv   10.1101/092601 .
  15. 1 2 Bergers, G.; Benjamin, L.E. (2003). "Tumorigenesis and the angiogenic switch". Nature Reviews. Cancer. 3 (6): 401–410. doi:10.1038/nrc1093. PMID   12778130. S2CID   11096398.
  16. 1 2 Banerjee, Deboshri; Harfouche, Rania; Sengupta, Shiladitya (31 January 2011). "Nanotechnology-mediated targeting of tumor angiogenesis". Vascular Cell. 3 (1): 3. doi: 10.1186/2045-824X-3-3 . PMC   3039831 . PMID   21349160.
  17. Arvizo, Rochelle R.; Rana, Subinoy; Miranda, Oscar R.; Bhattacharya, Resham; Rotello, Vincent M.; Mukherjee, Priyabata (16 January 2011). "Mechanism of anti-angiogenic property of gold nanoparticles: role of nanoparticle size and surface charge". Nanomedicine. 7 (5): 580–587. doi:10.1016/j.nano.2011.01.011. PMC   3138823 . PMID   21333757.
  18. N/A, N/A. "Nanogold in anti-angiogenic therapy". Mayo Clinic. Mayo Foundation for Medical Education and Research. Retrieved 25 March 2015.
  19. Morshed, Ramin A.; Muroski, Megan E.; Dai, Qing; Wegscheid, Michelle L.; Auffinger, Brenda; Yu, Dou; Han, Yu; Zhang, Lingjiao; Wu, Meijing (2016-06-06). "Cell-Penetrating Peptide-Modified Gold Nanoparticles for the Delivery of Doxorubicin to Brain Metastatic Breast Cancer". Molecular Pharmaceutics. 13 (6): 1843–1854. doi:10.1021/acs.molpharmaceut.6b00004. ISSN   1543-8384. PMID   27169484.
  20. 1 2 Fu, Yingjie; Feng, Qishuai; Chen, Yifan; Shen, Yajing; Su, Qihang; Zhang, Yinglei; Zhou, Xiang; Cheng, Yu (2016-09-06). "Comparison of Two Approaches for the Attachment of a Drug to Gold Nanoparticles and Their Anticancer Activities". Molecular Pharmaceutics. 13 (9): 3308–3317. doi:10.1021/acs.molpharmaceut.6b00619. ISSN   1543-8384. PMID   27518201. S2CID   38857901.
  21. Osborne CK (1998). "Tamoxifen in the treatment of breast cancer". N. Engl. J. Med. 339 (22): 1609–1618. doi:10.1056/NEJM199811263392207. PMID   9828250.
  22. Heinlein CA, Chang C (2004). "Androgen receptor in prostate cancer". Endocr. Rev. 25 (2): 276–308. doi: 10.1210/er.2002-0032 . PMID   15082523.
  23. E. Dreaden, L. Austin, M. Mackey, M. El-Sayed,. "Ther Deliv:size matters: gold nanoparticles in targeted cancer drug delivery, 3(4): 457-478; 2012
  24. A. Francois, A. Laroche, N. Pinaud, L. Salmon, J. Ruiz, J. Robert, D. Astruc.; ChemMedChem: Encapsulation of Docetaxel into PEGylated Gold Nanoparticles for Vectorization to Cancer Cells, 2011, 6, 2003 – 2008
  25. 1 2 http://www.cancer.gov/cancertopics/druginfo/fda-docetaxel Archived 2014-09-01 at the Wayback Machine , National Cancer Institute, Last updated 3/28/2014
  26. 1 2 3 4 "Gold Colloid". nanocomposix. Retrieved 24 March 2015.
  27. "Gold Nanoparticles: Properties and Applications". sigmaaldrich. Retrieved 24 March 2015.
  28. "Gold Nanoparticles". Nanopartz.
  29. "Ntracker". nanopartz. Retrieved 13 March 2019.
  30. Schoen, Christian (30 January 2015). Pandia®. In Nanotechnology for Biomedical Imaging and Diagnostics. Wiley Online Library. p. ch 12. doi:10.1002/9781118873151.ch12.
  31. Schuh, Elizabeth (27 July 2016). "Safety and efficacy of targeted hyperthermia treatment utilizing gold nanorod therapy in spontaneous canine neoplasia". BMC Veterinary Research. 2017 (13): 294. doi: 10.1186/s12917-017-1209-y . PMC   5625780 . PMID   28969639.
  32. "Ntracker". Nanopartz. Retrieved 13 March 2019.
  33. Murphy, Catherine J.; Gole, Anand M.; Stone, John W.; Sisco, Patrick N.; Alkilany, Alaadin M.; Goldsmith, Edie C.; Baxter, Sarah C. (December 2008). "Gold Nanoparticles in Biology: Beyond Toxicity to Cellular Imaging". Accounts of Chemical Research. 41 (12): 1721–1730. doi:10.1021/ar800035u. PMID   18712884. S2CID   23664437.
  34. Chithrani, B.D.; Ghazani, A.A.; Chan, W.C.W. (2006). "Determining the Size and Shape Dependence of Gold Nanoparticle Uptake into Mammalian Cells". Nano Lett. 6 (4): 662–668. Bibcode:2006NanoL...6..662C. CiteSeerX   10.1.1.458.2262 . doi:10.1021/nl052396o. PMID   16608261.
  35. Goodman, C.M.; McCusker, C.D.; Yilmaz, T.; Rotello, V.M. (2004). "Toxicity of Gold Nanoparticles Functionalized with Cationic and Anionic Side Chains". Bioconjugate Chemistry. 15 (4): 897–900. doi:10.1021/bc049951i. PMID   15264879.
  36. Pernodet, N.; Fang, X.; Sun, Y.; Bakhtina, A.; Ramakrishnan, A.; Sokolov, J.; Ulman, A.; Rafailovich, M. (2006). "Adverse Effects of Citrate/Gold Nanoparticles on Human Dermal Fibroblasts". Small. 2 (6): 766–773. doi: 10.1002/smll.200500492 . PMID   17193121.
  37. James, W.D.; Hirsch, L.R.; West, P.D.; O'Neal, J.D.; Payne, J (Jun 28, 2011). "Systematic Evaluation of Nanomaterial Toxicity: Utility of Standardized Materials and Rapid Assays". ACS Nano. 5 (6): 4688–4697. doi:10.1021/nn200546k. PMC   3124923 . PMID   21609003.
  38. Su, Chia-Hao; Sheu, Hwo-Shuenn; Lin, Chia-Yun; Huang, Chih-Chia; Lo, Yi-Wei; Pu, Ying-Chih; Weng, Jun-Cheng; Shieh, Dar-Bin; Jyh-Horng, Chen; Chen-Sheng, Yeh (January 31, 2007). "Nanoshell Magnetic Resonance Imaging Contrast Agents". J. Am. Chem. Soc. 129 (7): 2139–2146. doi:10.1021/ja0672066. PMID   17263533. S2CID   9022791.
  39. Selvan, S.T.; Tan, T.T.; Ying, J.Y. (4 May 2005). "Robust, Non-Cytotoxic, Silica-Coated CdSe Quantum Dots with Efficient Photoluminescence". Adv. Mater. 17 (13): 1620–1625. Bibcode:2005AdM....17.1620S. doi:10.1002/adma.200401960. S2CID   96001135.
  40. 1 2 3 4 5 Huang, Xiaohua; El-Sayed, Mostafa A. (2010-01-01). "Gold nanoparticles: Optical properties and implementations in cancer diagnosis and photothermal therapy". Journal of Advanced Research. 1 (1): 13–28. doi: 10.1016/j.jare.2010.02.002 .
  41. 1 2 Takahashi, Hironobu; Niidome, Takuro; Nariai, Ayuko; Niidome, Yasuro; Yamada, Sunao (2006). "Photothermal reshaping of gold nanorods prevents further cell death". Nanotechnology. 17 (17): 4431–4435. Bibcode:2006Nanot..17.4431T. doi:10.1088/0957-4484/17/17/024. S2CID   137276553.
  42. 1 2 Jain, S; Hirst, D G; O'Sullivan, J M (2012-02-01). "Gold nanoparticles as novel agents for cancer therapy". The British Journal of Radiology. 85 (1010): 101–113. doi:10.1259/bjr/59448833. ISSN   0007-1285. PMC   3473940 . PMID   22010024.
  43. 1 2 Takahashi, Hironobu; Niidome, Takuro; Nariai, Ayuko; Niidome, Yasuro; Yamada, Sunao (14 August 2006). "Photothermal reshaping of gold nanorods prevents further cell death". Nanotechnology. 17 (17): 4431–4435. Bibcode:2006Nanot..17.4431T. doi:10.1088/0957-4484/17/17/024. S2CID   137276553.
  44. Niidome, Takuro; Yamagata, Masato; Okamoto, Yuri; Akiyama, Yasuyuki; Takahashi, Hironobu; Kawano, Takahito; Katayama, Yoshiki; Niidome, Yasuro (2 September 2006). "PEG-modified gold nanorods with a stealth character for in vivo applications". Journal of Controlled Release. 114 (3): 343–347. doi:10.1016/j.jconrel.2006.06.017. PMID   16876898.
  45. 1 2 3 Sharma, Harshita; Mishra, Pawan K.; Talegaonkar, Sushama; Vaidya, Bhuvaneshwar (2015-09-01). "Metal nanoparticles: a theranostic nanotool against cancer". Drug Discovery Today. 20 (9): 1143–1151. doi:10.1016/j.drudis.2015.05.009. PMID   26007605.
  46. 1 2 3 Minai, Limor; Yeheskely-Hayon, Daniella; Yelin, Dvir (2013-07-05). "High levels of reactive oxygen species in gold nanoparticle-targeted cancer cells following femtosecond pulse irradiation". Scientific Reports. 3: 2146. Bibcode:2013NatSR...3E2146M. doi:10.1038/srep02146. PMC   3701901 . PMID   23828378.
  47. 1 2 Huang, Yue-Wern; Wu, Chi-heng; Aronstam, Robert S. (2010-10-25). "Toxicity of Transition Metal Oxide Nanoparticles: Recent Insights from in vitro Studies". Materials. 3 (10): 4842–4859. Bibcode:2010Mate....3.4842H. doi: 10.3390/ma3104842 . PMC   5445783 . PMID   28883356.
  48. 1 2 Khlebtsov, Nikolai; Dykman, Lev (2011). "Biodistribution and toxicity of engineered gold nanoparticles: a review of in vitro and in vivo studies". Chem. Soc. Rev. 40 (3): 1647–1671. doi:10.1039/c0cs00018c. PMID   21082078.
  49. Cassano, Domenico; Mapanao, Ana-Katrina; Summa, Maria; Vlamidis, Ylea; Giannone, Giulia; Santi, Melissa; Guzzolino, Elena; Pitto, Letizia; Poliseno, Laura; Bertorelli, Rosalia; Voliani, Valerio (2019-10-21). "Biosafety and Biokinetics of Noble Metals: The Impact of Their Chemical Nature". ACS Applied Bio Materials. 2 (10): 4464–4470. doi:10.1021/acsabm.9b00630. ISSN   2576-6422. PMID   35021406. S2CID   204266885.
  50. Cassano, Domenico; Summa, Maria; Pocoví-Martínez, Salvador; Mapanao, Ana-Katrina; Catelani, Tiziano; Bertorelli, Rosalia; Voliani, Valerio (February 2019). "Biodegradable Ultrasmall-in-Nano Gold Architectures: Mid-Period In Vivo Distribution and Excretion Assessment". Particle & Particle Systems Characterization. 36 (2): 1800464. doi:10.1002/ppsc.201800464. S2CID   104434042.
  51. Kreyling, Wolfgang G.; Abdelmonem, Abuelmagd M.; Ali, Zulqurnain; Alves, Frauke; Geiser, Marianne; Haberl, Nadine; Hartmann, Raimo; Hirn, Stephanie; Aberasturi, Dorleta Jimenez de (2015). "In vivo integrity of polymer-coated gold nanoparticles". Nature Nanotechnology. 10 (7): 619–623. Bibcode:2015NatNa..10..619K. doi:10.1038/nnano.2015.111. PMID   26076469.
  52. Chamberland, David, L.; Agarwal, Ashish; Kotov, Nicholas; Fowlkes, J Brian; Carson, Paul L; Wang, Xueding (11 February 2008). "Photoacoustic tomography of joints aided by an Etanercept-conjugated gold nanoparticle contrast agent—an ex vivo preliminary rat study" (PDF). Nanotechnology. 19 (9): 095101. Bibcode:2008Nanot..19i5101C. doi:10.1088/0957-4484/19/9/095101. hdl: 2027.42/64174 . PMID   21817663. S2CID   30251397.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  53. Alipour, Elias (23 June 2013). "Gold nanoparticle based capacitive immunosensor for detection of hepatitis B surface antigen". Analytical Methods. 5 (17): 4448. doi:10.1039/C3AY26586B. S2CID   55553319.

Reference notes

  1. [26] (NCT00848042)
  2. [26] (NCT01679470)
  3. [26] (NCT02680535)