[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (52)

Search Parameters:
Keywords = dendrimer size effect

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 2695 KiB  
Review
Lipidic and Inorganic Nanoparticles for Targeted Glioblastoma Multiforme Therapy: Advances and Strategies
by Ewelina Musielak and Violetta Krajka-Kuźniak
Micro 2025, 5(1), 2; https://doi.org/10.3390/micro5010002 - 3 Jan 2025
Viewed by 475
Abstract
Due to their biocompatibility, nontoxicity, and surface conjugation properties, nanomaterials are effective nanocarriers capable of encapsulating chemotherapeutic drugs and facilitating targeted delivery across the blood–brain barrier (BBB). Although research on nanoparticles for brain cancer treatment is still in its early stages, these systems [...] Read more.
Due to their biocompatibility, nontoxicity, and surface conjugation properties, nanomaterials are effective nanocarriers capable of encapsulating chemotherapeutic drugs and facilitating targeted delivery across the blood–brain barrier (BBB). Although research on nanoparticles for brain cancer treatment is still in its early stages, these systems hold great potential to revolutionize drug delivery. Glioblastoma multiforme (GBM) is one of the most common and lethal brain tumors, and its heterogeneous and aggressive nature complicates current treatments, which primarily rely on surgery. One of the significant obstacles to effective treatment is the poor penetration of drugs across the BBB. Moreover, GBM is often referred to as a “cold” tumor, characterized by an immunosuppressive tumor microenvironment (TME) and minimal immune cell infiltration, which limits the effectiveness of immunotherapies. Therefore, developing novel, more effective treatments is critical to improving the survival rate of GBM patients. Current strategies for enhancing treatment outcomes focus on the controlled, targeted delivery of chemotherapeutic agents to GBM cells across the BBB using nanoparticles. These therapies must be designed to engage specialized transport systems, allowing for efficient BBB penetration, improved therapeutic efficacy, and reduced systemic toxicity and drug degradation. Lipid and inorganic nanoparticles can enhance brain delivery while minimizing side effects. These formulations may include epitopes—small antigen fragments that bind directly to free antibodies, B cell receptors, or T cell receptors—that interact with transport systems and enable BBB crossing, thereby boosting therapeutic efficacy. Lipid-based nanoparticles (LNPs), such as liposomes, niosomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), are among the most promising delivery systems due to their unique properties, including their size, surface modification capabilities, and proven biosafety. Additionally, inorganic nanoparticles such as gold nanoparticles, mesoporous silica, superparamagnetic iron oxide nanoparticles, and dendrimers offer promising alternatives. Inorganic nanoparticles (INPs) can be easily engineered, and their surfaces can be modified with various elements or biological ligands to enhance BBB penetration, targeted delivery, and biocompatibility. Strategies such as surface engineering and functionalization have been employed to ensure biocompatibility and reduce cytotoxicity, making these nanoparticles safer for clinical applications. The use of INPs in GBM treatment has shown promise in improving the efficacy of traditional therapies like chemotherapy, radiotherapy, and gene therapy, as well as advancing newer treatment strategies, including immunotherapy, photothermal and photodynamic therapies, and magnetic hyperthermia. This article reviews the latest research on lipid and inorganic nanoparticles in treating GBM, focusing on active and passive targeting approaches. Full article
(This article belongs to the Section Microscale Biology and Medicines)
Show Figures

Figure 1

Figure 1
<p>Diagram of the structure of the human brain [<a href="#B9-micro-05-00002" class="html-bibr">9</a>].</p>
Full article ">
17 pages, 2461 KiB  
Article
New Approaches for Basophil Activation Tests Employing Dendrimeric Antigen–Silica Nanoparticle Composites
by Silvia Calvo-Serrano, Esther Matamoros, Jose Antonio Céspedes, Rubén Fernández-Santamaría, Violeta Gil-Ocaña, Ezequiel Perez-Inestrosa, Cecilia Frecha, Maria I. Montañez, Yolanda Vida, Cristobalina Mayorga and Maria J. Torres
Pharmaceutics 2024, 16(8), 1039; https://doi.org/10.3390/pharmaceutics16081039 - 3 Aug 2024
Cited by 2 | Viewed by 1156
Abstract
In vitro cell activation through specific IgE bound to high-affinity receptors on the basophil surface is a widely used strategy for the evaluation of IgE-mediated immediate hypersensitivity reactions to betalactams. Cellular activation requires drug conjugation to a protein to form a large enough [...] Read more.
In vitro cell activation through specific IgE bound to high-affinity receptors on the basophil surface is a widely used strategy for the evaluation of IgE-mediated immediate hypersensitivity reactions to betalactams. Cellular activation requires drug conjugation to a protein to form a large enough structure displaying a certain distance between haptens to allow the cross-linking of two IgE antibodies bound to the basophil’s surface, triggering their degranulation. However, no information about the size and composition of these conjugates is available. Routine in vitro diagnosis using the basophil activation test uses free amoxicillin, which is assumed to conjugate to a carrier present in blood. To standardize the methodology, we propose the use of well-controlled and defined nanomaterials functionalized with amoxicilloyl. Silica nanoparticles decorated with PAMAM–dendrimer–amoxicilloyl conjugates (NpDeAXO) of different sizes and amoxicilloyl densities (50–300 µmol amoxicilloyl/gram nanoparticle) have been prepared and chemically characterized. Two methods of synthesis were performed to ensure reproducibility and stability. Their functional effect on basophils was measured using an in-house basophil activation test (BAT) that determines CD63+ or CD203chigh activation markers. It was observed that NpDeAXO nanocomposites are not only able to specifically activate basophils but also do so in a more effective way than free amoxicillin, pointing to a translational potential diagnosis. Full article
Show Figures

Figure 1

Figure 1
<p>General procedure for the chemical modification of ϕ<b>dNp</b> dispersions and <b>50Nps</b> with different DeAXO densities in their surface.</p>
Full article ">Figure 2
<p>NMR spectra of (<b>a</b>) AX in basic D<sub>2</sub>O and (<b>b</b>) <b>50NpDeAXO</b> in D<sub>2</sub>O suspensions.</p>
Full article ">Figure 3
<p>Basophil activation test (BAT) dose–response curves of <b>NpDeAXO</b> of different sizes: 20 nm (<b>A</b>,<b>B</b>), 30 nm (<b>C</b>,<b>D</b>), and 50 nm (<b>E</b>,<b>F</b>) and (<b>G</b>,<b>H</b>). <span class="html-italic">Np dispersions</span> or <span class="html-italic">solid-state Nps</span> labels at the top of the figure only indicate the synthetic methodology used for Np preparation. Black lines represent healthy controls (HCs), and blue and green lines represent allergic patients (APs). Size sample included HCs (N = 10) and APs (N = 10) for Nps synthesized as dispersions (<b>A</b>–<b>F</b>), while HCs (N = 45) and APs (N = 54) were included in the study for the Nps synthesized as a solid state. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>BAT dose–response curves of Nps with different DeAXO surface densities: 300 µmol AXO/gNp (<b>A</b>,<b>B</b>); 130 µmolAXO/gNp (<b>C</b>,<b>D</b>); 100 µmol AXO/gNp (<b>E</b>,<b>F</b>); 80 µmol AXO/gNp (<b>G</b>,<b>H</b>); 50 µmol AXO/gNp (<b>I</b>,<b>J</b>). Black lines represent healthy controls (HCs), and blue and green lines represent allergic patients (APs). Size sample included HCs (N = 6) and APs (N = 4) (<b>A</b>–<b>J</b>). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>BAT dose–response curves using <b>50NpDeAXO</b> and free AX at three different concentrations. APs (N = 54) are depicted by blue or green lines and HCs (N = 45) are depicted by black lines. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Percentage of positive cases in allergic patients (N = 54) in BAT using CD63 (<b>A</b>) and CD203c<sup>high</sup> (<b>B</b>) as basophil activation markers. Positive cases were obtained after using the cut-offs described in <a href="#app1-pharmaceutics-16-01039" class="html-app">Table S4</a> for AX and <b>50NpDeAXO</b> at the different concentrations with both CD63 and CD203c.</p>
Full article ">
24 pages, 3937 KiB  
Article
Exploring the Influence of Spacers in EDTA–β-Cyclodextrin Dendrimers: Physicochemical Properties and In Vitro Biological Behavior
by Israel González-Méndez, Kendra Sorroza-Martínez, Ignacio González-Sánchez, Jesús Gracia-Mora, María Josefa Bernad-Bernad, Marco Cerbón, Ernesto Rivera and Anatoly K. Yatsimirsky
Int. J. Mol. Sci. 2023, 24(19), 14422; https://doi.org/10.3390/ijms241914422 - 22 Sep 2023
Cited by 4 | Viewed by 1503
Abstract
The synthesis of a new family of ethylenediaminetetraacetic acid (EDTA) core dimers and G0 dendrimers end-capped with two and four β-cyclodextrin (βCD) moieties was performed by click-chemistry conjugation, varying the spacers attached to the core. The structure analyses were achieved in DMSO-d [...] Read more.
The synthesis of a new family of ethylenediaminetetraacetic acid (EDTA) core dimers and G0 dendrimers end-capped with two and four β-cyclodextrin (βCD) moieties was performed by click-chemistry conjugation, varying the spacers attached to the core. The structure analyses were achieved in DMSO-d6 and the self-inclusion process was studied in D2O by 1H-NMR spectroscopy for all platforms. It was demonstrated that the interaction with adamantane carboxylic acid (AdCOOH) results in a guest-induced shift of the self-inclusion effect, demonstrating the full host ability of the βCD units in these new platforms without any influence of the spacer. The results of the quantitative size and water solubility measurements demonstrated the equivalence between the novel EDTA-βCD platforms and the classical PAMAM-βCD dendrimer. Finally, we determined the toxicity for all EDTA-βCD platforms in four different cell lines: two human breast cancer cells (MCF-7 and MDA-MB-231), human cervical adenocarcinoma cancer cells (HeLa), and human lung adenocarcinoma cells (SK-LU-1). The new EDTA-βCD carriers did not present any cytotoxicity in the tested cell lines, which showed that these new classes of platforms are promising candidates for drug delivery. Full article
(This article belongs to the Special Issue Current Uses and Applications of Cyclodextrins)
Show Figures

Figure 1

Figure 1
<p>Comparison of <sup>1</sup>H-NMR spectra of EDTA4BenCD dendrimer (<b>E</b>), (<b>A</b>) acquired in DMSO-<span class="html-italic">d</span><sub>6</sub> and (<b>B</b>) acquired in D<sub>2</sub>O.</p>
Full article ">Figure 2
<p>Possible conformations adopted for EDTA4BenCD dendrimer <b>E</b>, with 0 to 4 reversed cavities.</p>
Full article ">Figure 3
<p><sup>1</sup>H-NMR titration experiment of EDTA4BenCD dendrimer (<b>E</b>). The 5.0–8.2 ppm and 1.5–3.0 ppm regions are shown for the mixtures of <b>E</b> and AdCOOH in D<sub>2</sub>O with decreasing molar fraction from top to bottom, expressed with respect to the concentration in βCD cavities for EDTA4BenCD dendrimer (<b>E</b>) (XCD = [βCD]/([βCD] + [AdCOOH]). Total concentration [βCD] + [AdCOOH] = 7.3 mM.</p>
Full article ">Figure 4
<p>(<b>A</b>) <sup>1</sup>H-NMR spectra of IC EDTA4BenCD dendrimer (<b>E</b>) with AdCOOH in D<sub>2</sub>O. (<b>B</b>) Amplification of aliphatic zone in the 2D NMR NOESY spectrum of IC EDTA4BenCD dendrimer (<b>E</b>) with AdCOOH in D<sub>2</sub>O.</p>
Full article ">Figure 5
<p>(<b>A</b>) Determination of particle size by DLS. (<b>B</b>) Comparison of the determined sizes for dendrimers EDTA-βCD (<b>D</b>–<b>F</b>) and dimers EDTA-βCD (<b>A</b>–<b>C</b>).</p>
Full article ">Figure 6
<p>Cell viability in different cell lines for EDTA βCD molecules (<b>A</b>–<b>F</b>). The treated cells were cultured in six wells by concentration, and the experiments were performed independently three times. Data are means ± SD.</p>
Full article ">Scheme 1
<p>Synthetic route for the obtention of <span class="html-italic">m</span>N<sub>3</sub>βCD.</p>
Full article ">Scheme 2
<p>Synthetic route of dimers EDTA-βCD.</p>
Full article ">Scheme 3
<p>Synthetic route for dendrimers EDTA-βCD.</p>
Full article ">
12 pages, 3452 KiB  
Article
Polyamidoamine Dendrimers Functionalized Water-Stable Metal–Organic Frameworks for Sensitive Fluorescent Detection of Heavy Metal Ions in Aqueous Solution
by Dandan Guo, Nadeem Muhammad, Shuxin Yu, Jinhui Wang, Shaohua Huang and Yan Zhu
Polymers 2023, 15(16), 3444; https://doi.org/10.3390/polym15163444 - 18 Aug 2023
Cited by 3 | Viewed by 1435
Abstract
In this work, polyamidoamine (PAMAM)-functionalized water-stable Al-based metal–organic frameworks (MIL-53(Al)-NH2) were proposed with enhanced fluorescence intensity, and used for the sensitive detection of heavy metal ions in aqueous solution. The size and morphology of MIL-53(Al)-NH2 were effectively optimized by regulating [...] Read more.
In this work, polyamidoamine (PAMAM)-functionalized water-stable Al-based metal–organic frameworks (MIL-53(Al)-NH2) were proposed with enhanced fluorescence intensity, and used for the sensitive detection of heavy metal ions in aqueous solution. The size and morphology of MIL-53(Al)-NH2 were effectively optimized by regulating the component of the reaction solvents. PAMAM dendrimers were subsequently grafted onto the surface with glutaraldehyde as a cross-linking agent. It was found that the size and morphology of MIL-53(Al)-NH2 have great influence on their fluorescence properties, and PAMAM grafting could distinctly further improve their fluorescence intensity. With higher fluorescence intensity, the PAMAM-grafted MIL-53(Al)-NH2 showed good linearity (R2 = 0.9925–0.9990) and satisfactory sensitivity (LOD = 1.1–8.6 μmol) in heavy metal ions determination. Fluorescence enhancement and heavy metal ions detection mechanisms were discussed following the experimental results. Furthermore, analogous water-stable Materials of Institute Lavoisier (MIL) metal–organic frameworks such as MIL-53(Fe)-NH2 were also proved to have similar fluorescence enhancement performance after PAMAM modification, which demonstrates the universality of the method and the great application prospects in the design of PAMAM-functionalized high-sensitivity fluorescence sensors. Full article
Show Figures

Figure 1

Figure 1
<p>Preparation of PAMAM-grafted MOFs materials.</p>
Full article ">Figure 2
<p>SEM images of MIL-53(Al)-NH<sub>2</sub> obtained with a water proportion of 0% (<b>A</b>,<b>E</b>), 25% (<b>B</b>,<b>F</b>), 50% (<b>C</b>,<b>G</b>) and 75% (<b>D</b>,<b>H</b>).</p>
Full article ">Figure 3
<p>(<b>A</b>) XRD patterns of as-synthesized MIL-53(Al)-NH<sub>2</sub> with different proportions of water; (<b>B</b>) FTIR spectra of MIL-53(Al)-NH<sub>2</sub> materials synthesized with different proportions of water.</p>
Full article ">Figure 4
<p>XPS spectra of nitrogen in (<b>A</b>) MIL-53(Al)-NH<sub>2</sub> and (<b>B</b>) PAMAM-grafted MIL-53(Al)-NH<sub>2</sub>. The yellow line and black lines in the figure are the baseline and the total result of all peak fits, respectively.</p>
Full article ">Figure 5
<p>(<b>A</b>) Fluorescence emission spectra of MIL-53(Al)-NH<sub>2</sub> synthesized with different proportions of water (excitation slit: 2.5 nm; emission slit: 5.0 nm); (<b>B</b>) Fluorescence emission spectra of MIL-53(Al)-NH<sub>2</sub> grafted with PAMAM dendrimers (excitation slit: 2.5 nm; emission slit: 2.5 nm); Effect of pH (<b>C</b>) and concentrations (<b>D</b>) on fluorescence intensity of MIL-53-1.0G PAMAM (excitation slit: 1.0 nm; emission slit: 10 nm).</p>
Full article ">Figure 6
<p>Linear relationships between heavy metal ions concentrations and fluorescence quenching efficiency.</p>
Full article ">Figure 7
<p>(<b>A</b>) Full XPS spectrum of Pb<sup>2+</sup> quenched MIL-53(Al)-2.0G PAMAM; (<b>B</b>) Pb 4f spectra after Pb<sup>2+</sup> treatment (The red and blue line is the orbital peak fitting result of Pb 4f, and two black lines are the baseline and the total result of all peak fits, respectively); (<b>C</b>–<b>E</b>) EDS-mapping of Pb<sup>2+</sup> quenched MIL-53(Al)-2.0G PAMAM.</p>
Full article ">
30 pages, 3281 KiB  
Review
Recent Advances in Nanomaterials-Based Targeted Drug Delivery for Preclinical Cancer Diagnosis and Therapeutics
by Harshita Tiwari, Nilesh Rai, Swati Singh, Priyamvada Gupta, Ashish Verma, Akhilesh Kumar Singh, Kajal, Prafull Salvi, Santosh Kumar Singh and Vibhav Gautam
Bioengineering 2023, 10(7), 760; https://doi.org/10.3390/bioengineering10070760 - 25 Jun 2023
Cited by 46 | Viewed by 11201
Abstract
Nano-oncology is a branch of biomedical research and engineering that focuses on using nanotechnology in cancer diagnosis and treatment. Nanomaterials are extensively employed in the field of oncology because of their minute size and ultra-specificity. A wide range of nanocarriers, such as dendrimers, [...] Read more.
Nano-oncology is a branch of biomedical research and engineering that focuses on using nanotechnology in cancer diagnosis and treatment. Nanomaterials are extensively employed in the field of oncology because of their minute size and ultra-specificity. A wide range of nanocarriers, such as dendrimers, micelles, PEGylated liposomes, and polymeric nanoparticles are used to facilitate the efficient transport of anti-cancer drugs at the target tumor site. Real-time labeling and monitoring of cancer cells using quantum dots is essential for determining the level of therapy needed for treatment. The drug is targeted to the tumor site either by passive or active means. Passive targeting makes use of the tumor microenvironment and enhanced permeability and retention effect, while active targeting involves the use of ligand-coated nanoparticles. Nanotechnology is being used to diagnose the early stage of cancer by detecting cancer-specific biomarkers using tumor imaging. The implication of nanotechnology in cancer therapy employs photoinduced nanosensitizers, reverse multidrug resistance, and enabling efficient delivery of CRISPR/Cas9 and RNA molecules for therapeutic applications. However, despite recent advancements in nano-oncology, there is a need to delve deeper into the domain of designing and applying nanoparticles for improved cancer diagnostics. Full article
(This article belongs to the Collection Nanoparticles for Therapeutic and Diagnostic Applications)
Show Figures

Figure 1

Figure 1
<p>Nanotechnology tools-based cancer targeting diagnosis and treatment. Various types of nanotechnological tools such as liposome, polymers, micelles, and dendrimers are exploited for targeted delivery of drug nanoparticles. The targeting of these nanoparticles may be active or passive. Nanotechnology-based cancer diagnosis is possible by the application of microfluidic chip, identification of biomarkers, nanoscale probe and quantum dots. Cancer treatment is made possible by nanotechnology-based photoinduced gene silencing, RNA mediated cancer nanotherapy, overcoming multidrug resistance and nanotechnology-based CRISPR/Cas9 delivery.</p>
Full article ">Figure 2
<p>Mechanism of passive and active targeting of NP-drug conjugates. During passive targeting, the drug-loaded NPs are transported via leaky blood vasculature through EPR effect due to poor lymphatic drainage and release of drug from the nanoparticles takes place due to acidic microenvironment of tumor. On the other hand, active targeting involves binding of ligand-conjugated drug-loaded nanoparticles to the antigenic receptor on cancer cells followed by receptor-mediated endocytic uptake.</p>
Full article ">Figure 3
<p>Lactosomal nanophotosensitizer mediated photodynamic treatment. Lactosomal nanophotosensitizer is designed by the incorporation of a polypeptide and photosensitizer conjugated with siRNAs. The lactosomal nanophotosensitizer upon administration is subjected to irradiation due to which the bound siRNAs are released and are responsible for radiation resistance gene silencing. The nanophotosensitizer is also responsible for the generation of ROS which promotes apoptosis. Additionally, phototreatment also induces the overexpression of ALA peptide resulting in ALA-mediated cell cytotoxicity.</p>
Full article ">Figure 4
<p>Mechanism of multidrug resistance and its reversal on treatment with NP-drug conjugates. MDR may occur as a result of reduced binding affinity of receptor with the drug. Hyper-activation of efflux transporters also leads to MDR causing rapid efflux of drug which results in reduced drug uptake and hence less intracellular drug accumulation takes place. MDR may also be caused due to defective apoptotic machinery or due to hypoxia which is responsible for overexpression of <span class="html-italic">Hif-1</span> gene and <span class="html-italic">Hsp90</span> gene. Other than this, the expression of tumor suppressor gene (<span class="html-italic">p53</span>) and pro-apoptotic genes (<span class="html-italic">Bax</span>, <span class="html-italic">Bak</span> and <span class="html-italic">Bid</span>) is reduced while the expression of anti-apoptotic genes (<span class="html-italic">Bcl-2</span>, <span class="html-italic">Bcl-xL</span>, <span class="html-italic">Bcl-W</span>) is increased. For MDR reversal, drug-loaded NPs are used that have higher binding affinity with receptors on a cancer cell which facilitates rapid uptake of drugs and its increased intracellular accumulation. The drug is afterwards released in the tumor cell due to low pH at the perinuclear surface which prevents the normal cells from system cytotoxicity, thereby resisting MDR. NP-loaded drug targets the genetic machinery and results in inhibition of <span class="html-italic">Hsp90</span> gene, which further causes suppression of <span class="html-italic">Hif-1</span> gene. Furthermore, the released drug also enhances the expression of <span class="html-italic">p53</span>, <span class="html-italic">Bax</span>, <span class="html-italic">Bak</span>, <span class="html-italic">Bid</span> and inhibits the action of <span class="html-italic">Bcl-2</span>, <span class="html-italic">Bcl-xL</span> and <span class="html-italic">Bcl-W</span> genes, which induce apoptotic machinery. The drug also targets P13K/Akt/mTOR pathway and mitochondrial DNA repairing machinery, resulting in mitochondrial DNA damage and release of Cyt-c, thereby inducing apoptosis.</p>
Full article ">
14 pages, 2290 KiB  
Article
Erlotinib-Loaded Dendrimer Nanocomposites as a Targeted Lung Cancer Chemotherapy
by Wafa K. Fatani, Fadilah S. Aleanizy, Fulwah Y. Alqahtani, Mohammed M. Alanazi, Abdullah A. Aldossari, Faiyaz Shakeel, Nazrul Haq, Hosam Abdelhady, Hamad M. Alkahtani and Ibrahim A. Alsarra
Molecules 2023, 28(9), 3974; https://doi.org/10.3390/molecules28093974 - 8 May 2023
Cited by 20 | Viewed by 2810
Abstract
Lung cancer is the main cause of cancer-related mortality globally. Erlotinib is a tyrosine kinase inhibitor, affecting both cancerous cell proliferation and survival. The emergence of oncological nanotechnology has provided a novel drug delivery system for erlotinib. The aims of this current investigation [...] Read more.
Lung cancer is the main cause of cancer-related mortality globally. Erlotinib is a tyrosine kinase inhibitor, affecting both cancerous cell proliferation and survival. The emergence of oncological nanotechnology has provided a novel drug delivery system for erlotinib. The aims of this current investigation were to formulate two different polyamidoamine (PAMAM) dendrimer generations—generation 4 (G4) and generation 5 (G5) PAMAM dendrimer—to study the impact of two different PAMAM dendrimer formulations on entrapment by drug loading and encapsulation efficiency tests; to assess various characterizations, including particle size distribution, polydispersity index, and zeta potential; and to evaluate in vitro drug release along with assessing in situ human lung adenocarcinoma cell culture. The results showed that the average particle size of G4 and G5 nanocomposites were 200 nm and 224.8 nm, with polydispersity index values of 0.05 and 0.300, zeta potential values of 11.54 and 4.26 mV of G4 and G5 PAMAM dendrimer, respectively. Comparative in situ study showed that cationic G4 erlotinib-loaded dendrimer was more selective and had higher antiproliferation activity against A549 lung cells compared to neutral G5 erlotinib-loaded dendrimers and erlotinib alone. These conclusions highlight the potential effect of cationic G4 dendrimer as a targeting-sustained-release carrier for erlotinib. Full article
Show Figures

Figure 1

Figure 1
<p>TEM images at higher magnifications to inspect the morphology of (<b>a</b>) blank Generation 4-FITC (G4-FITC) polyamidoamine (PAMAM) dendrimers; (<b>b</b>) erlotinib conjugated G4-FITC PAMAM dendrimers proving erlotinib placement within the dendrimer; (<b>c</b>) blank generation (G5-FITC) PAMAM dendrimers; and (<b>d</b>) erlotinib conjugated G5-FITC PAMAM dendrimers proving encapsulation of erlotinib within the dendrimer cavity.</p>
Full article ">Figure 2
<p>In vitro release of erlotinib, assessed as a percentage of cumulative release from dendrimers G4-FITC and G5-FITC, showing comparable prolonged release patterns. (<b>a</b>) Comparative in vitro release study at pH 5.4 for PAMAM dendrimer G4-FITC and G5-FITC; (<b>b</b>) comparative in vitro release study at pH 7.4 for PAMAM dendrimer G4-FITC and G5-FITC; and (<b>c</b>) comparison in vitro release study for ERL tablet between two media: 0.01 N HCl and 0.02% Tween 80 in 0.01 N HCl.</p>
Full article ">Figure 3
<p>(<b>a</b>) MTT results showing Erlotinib decreased the A549 cells’ viability significantly with all the doses (10, 20, 40, 80 µg/mL) (<span class="html-italic">p</span> &lt; 0.0001); (<b>b</b>) effect of PAMAM dendrimers G4-FITC and ERL-G4-FITC complex after 72 h. Exposure on the cell viability of A549 cells under controlled conditions; (<b>c</b>) effect of PAMAM G5-FITC and ERL-G5-FITC complex after 72 h. Exposure on the cell viability of A549 cells under controlled conditions; (<b>d</b>) a dose of 80 µg/mL was selected to compare the cell viability between PAMAM dendrimers G4-FITC, ERL-G4-FITC complex, PAMAM dendrimers G5-FITC, and ERL-G5-FITC complex. *—Represents a comparison between each group of treatment to the control (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001). #—Represents a comparison between each dose of G4 or G5 PAMAM dendrimer and the matched dose of ERL G4 or G5 complexes (## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, #### <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>Flow cytometry analysis after treatment for 72 h (<span class="html-italic">n</span> = 3/group); **** <span class="html-italic">p</span> &lt; 0.0001, ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Stability of erlotinib from formula obtained from four different temperatures (4 °C, 25 °C, 37 °C, 50 °C) for (<b>a</b>) PAMAM dendrimer G4-FITC complex; (<b>b</b>) PAMAM dendrimer G5-FITC complex for a period of 6 months.</p>
Full article ">
32 pages, 5406 KiB  
Review
Advancement in Solubilization Approaches: A Step towards Bioavailability Enhancement of Poorly Soluble Drugs
by Lakshmi Kumari, Yash Choudhari, Preeti Patel, Ghanshyam Das Gupta, Dilpreet Singh, Jessica M. Rosenholm, Kuldeep Kumar Bansal and Balak Das Kurmi
Life 2023, 13(5), 1099; https://doi.org/10.3390/life13051099 - 27 Apr 2023
Cited by 56 | Viewed by 14344
Abstract
A drug’s aqueous solubility is defined as the ability to dissolve in a particular solvent, and it is currently a major hurdle in bringing new drug molecules to the market. According to some estimates, up to 40% of commercialized products and 70–90% of [...] Read more.
A drug’s aqueous solubility is defined as the ability to dissolve in a particular solvent, and it is currently a major hurdle in bringing new drug molecules to the market. According to some estimates, up to 40% of commercialized products and 70–90% of drug candidates in the development stage are poorly soluble, which results in low bioavailability, diminished therapeutic effects, and dosage escalation. Because of this, solubility must be taken into consideration when developing and fabricating pharmaceutical products. To date, a number of approaches have been investigated to address the problem of poor solubility. This review article attempts to summarize several conventional methods utilized to increase the solubility of poorly soluble drugs. These methods include the principles of physical and chemical approaches such as particle size reduction, solid dispersion, supercritical fluid technology, cryogenic technology, inclusion complex formation techniques, and floating granules. It includes structural modification (i.e., prodrug, salt formation, co-crystallization, use of co-solvents, hydrotrophy, polymorphs, amorphous solid dispersions, and pH variation). Various nanotechnological approaches such as liposomes, nanoparticles, dendrimers, micelles, metal organic frameworks, nanogels, nanoemulsions, nanosuspension, carbon nanotubes, and so forth have also been widely investigated for solubility enhancement. All these approaches have brought forward the enhancement of the bioavailability of orally administered drugs by improving the solubility of poorly water-soluble drugs. However, the solubility issues have not been completely resolved, owing to several challenges associated with current approaches, such as reproducibility in large scale production. Considering that there is no universal approach for solving solubility issues, more research is needed to simplify the existing technologies, which could increase the number of commercially available products employing these techniques. Full article
(This article belongs to the Special Issue New Trends in Pharmaceutical Science: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Fate of drug molecule after administering via oral route.</p>
Full article ">Figure 2
<p>Conventional methods for solubility enhancement.</p>
Full article ">Figure 3
<p>Nanocarrier-mediated solubility enhancement techniques.</p>
Full article ">Figure 4
<p>Mechanism of prodrug solubility enhancement.</p>
Full article ">Figure 5
<p>Mechanism of drug’s solubilization with the aid of liposomes.</p>
Full article ">Figure 6
<p>A schematic illustration of supercritical antisolvent (SAS) apparatus. S1: CO<sub>2</sub> supply; S2: Liquid solution supply; P1 &amp; P2; Pumps; RB: Refrigerating bath; LS: Liquid Separator; PC: Precipitation Chamber; MV: Micrometric Valve; BPV: Block-pressure Valve; R: Rotameter.</p>
Full article ">
20 pages, 10665 KiB  
Article
Combination of Copper Metallodendrimers with Conventional Antitumor Drugs to Combat Cancer in In Vitro Models
by Marcin Hołota, Sylwia Michlewska, Sandra Garcia-Gallego, Natalia Sanz del Olmo, Paula Ortega, Maria Bryszewska, Francisco Javier de la Mata and Maksim Ionov
Int. J. Mol. Sci. 2023, 24(4), 4076; https://doi.org/10.3390/ijms24044076 - 17 Feb 2023
Cited by 7 | Viewed by 2558
Abstract
Copper carbosilane metallodendrimers containing chloride ligands and nitrate ligands were mixed with commercially available conventional anticancer drugs, doxorubicin, methotrexate and 5-fluorouracil, for a possible therapeutic system. To verify the hypothesis that copper metallodendrimers can form conjugates with anticancer drugs, their complexes were biophysically [...] Read more.
Copper carbosilane metallodendrimers containing chloride ligands and nitrate ligands were mixed with commercially available conventional anticancer drugs, doxorubicin, methotrexate and 5-fluorouracil, for a possible therapeutic system. To verify the hypothesis that copper metallodendrimers can form conjugates with anticancer drugs, their complexes were biophysically characterized using zeta potential and zeta size methods. Next, to confirm the existence of a synergetic effect of dendrimers and drugs, in vitro studies were performed. The combination therapy has been applied in two cancer cell lines: MCF-7 (human breast cancer cell line) and HepG2 (human liver carcinoma cell line). The doxorubicin (DOX), methotrexate (MTX) and 5-fluorouracil (5-FU) were more effective against cancer cells when conjugated with copper metallodendrimers. Such combination significantly decreased cancer cell viability when compared to noncomplexed drugs or dendrimers. The incubation of cells with drug/dendrimer complexes resulted in the increase of the reactive oxygen species (ROS) levels and the depolarization of mitochondrial membranes. Copper ions present in the dendrimer structures enhanced the anticancer properties of the whole nanosystem and improved drug effects, inducing both the apoptosis and necrosis of MCF-7 (human breast cancer cell line) and HepG2 (human liver carcinoma cell line) cancer cells. Full article
(This article belongs to the Special Issue Biopolymers as Nanoparticles Carriers)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Zeta potential (<b>A</b>) and zeta average size (<b>B</b>) of DOX—top panels; 5-FU—middle panels; and MTX—bottom panels, at the presence of increasing concentrations of copper dendrimers. Drugs concentration 10 µmol/L. The measurements were performed using sodium phosphate buffer 10 mmol/L, pH 7.4. Results are mean ± standard deviation (SD), n = 3.</p>
Full article ">Figure 2
<p>Cytotoxicity profiles of anticancer drugs, copper dendrimers and their (dendrimer/drug) complexes towards MCF7 cells. MTT assay, incubation time 72 h in phosphate saline buffer 10 mmol/L, pH 7.4. Results are means ± SD, from a min. 3 independent experiments. Statistically significant differences vs. control: * <span class="html-italic">p</span> &lt;0.05, *** <span class="html-italic">p</span> &lt; 0.001, vs. free drug <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001, vs. free dendrimer ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Cytotoxicity profiles of anticancer drugs, copper dendrimers and their (dendrimer/drug) complexes towards HEP G2 cells. MTT assay, incubation time 72 h in phosphate saline buffer 10 mmol/L, pH 7.4. Results are means ± SD, from a min. 3 independent experiments. Statistically significant differences vs. control: * <span class="html-italic">p</span> &lt;0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, vs. free drug <span>$</span> <span class="html-italic">p</span> &lt;0.05, <span>$</span><span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.001, vs. free dendrimer ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Time-dependent ROS production in MCF7 (<b>A</b>) or HEP G2 (<b>B</b>) cells induced by copper dendrimers or their complexes with anticancer drugs DOX, 5-Fu and MTX. Fluorescent probe H<sub>2</sub>DCFDA 2 µmol/L, incubation time 0.5 h, 3 h, 24 h, 48 h, phosphate-buffered saline 10 mmol/L, pH 7.4. Results are means ± SD, from a min. 3 independent experiments. Statistically significant differences vs. control: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. Control (untreated cells) is 100% ROS production.</p>
Full article ">Figure 5
<p>Changes in the mitochondrial membrane potential (ΔΨ<sub>m</sub>) in MCF7 (<b>A</b>) and HEP G2 (<b>B</b>) cells after exposure to copper dendrimers or their complexes with anticancer drugs DOX, 5-Fu and MTX, measured by JC-1 fluorescent probe in PBS10 mmol/L, pH 7.4. Results are means ± SD, from a min. 3 independent experiments. Statistically significant differences vs. control: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Images of MCF7 (<b>A</b>) and HEP G2 (<b>B</b>) cells after 48 h exposure to dendrimers, anticancer drugs or dendrimer drug complexes. Incubation time 48 h. The concentrations of drugs were as follows: DOX, 0.1 µmol/L (DOX/dendrimer molar ratio 1:7); 5-FU, 1 µmol/L (5-FU/dendrimer molar ratio 1:1); MTX, 2 nmol/L (MTX/dendrimer molar ratio: 1:32). Cells were stained with orange acridine (OA) and ethidium bromide (EB) and visualized by confocal microscopy. Living cells: morphologically normal (green nucleus); early apoptotic cells: condensed or fragmented chromatin (green nucleus); late apoptotic cells: fragmented and condensed (red chromatin); necrotic: red morphologically normal cells. Scale bar = 25 µm.</p>
Full article ">Figure 7
<p>The percentages of cells in different phases and apoptosis profile evaluated by flow cytometry and measured using annexin V/propidium iodide staining in PBS 10 mmol/L, pH 7.4. MCF7 (<b>A</b>) and HEP G2 (<b>B</b>) cells interacted with dendrimers, anticancer drugs or dendrimer drug complexes. Incubation time 48 h. The concentrations of drugs were as follows: DOX, 0.1 µmol/L (DOX/dendrimer molar ratio 1:7); 5-FU, 1 µmol/L (5-FU/dendrimer molar ratio 1:1); MTX, 2 nmol/L (MTX/dendrimer molar ratio: 1:32). Results are means ± SD, from a min. 3 independent experiments. Statistically significant differences vs. control: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Dendritic structures of Gn-[NCPh(<span class="html-italic">o</span>-N)CuCl<sub>2</sub>·H<sub>2</sub>O]m (CCD-Cl) and Gn-[NCPh(o-N)Cu(ONO<sub>2</sub>)<sub>2</sub>·H<sub>2</sub>O]m (CCD-NO) systems.</p>
Full article ">
16 pages, 3742 KiB  
Article
Complexes of Cationic Pyridylphenylene Dendrimers with Anionic Liposomes: The Role of Dendrimer Composition in Membrane Structural Changes
by Anna A. Efimova, Svetlana A. Sorokina, Kseniya S. Trosheva, Alexander A. Yaroslavov and Zinaida B. Shifrina
Int. J. Mol. Sci. 2023, 24(3), 2225; https://doi.org/10.3390/ijms24032225 - 22 Jan 2023
Cited by 6 | Viewed by 1958
Abstract
In the last decades, dendrimers have received attention in biomedicine that requires detailed study on the mechanism of their interaction with cell membranes. In this article, we report on the role of dendrimer structure in their interaction with liposomes. Here, the interactions between [...] Read more.
In the last decades, dendrimers have received attention in biomedicine that requires detailed study on the mechanism of their interaction with cell membranes. In this article, we report on the role of dendrimer structure in their interaction with liposomes. Here, the interactions between cationic pyridylphenylene dendrimers of the first, second, and third generations with mixed or completely charged pyridyl periphery (D16+, D215+, D229+, and D350+) with cholesterol-containing (CL/Chol/DOPC) anionic liposomes were investigated by microelectrophoresis, dynamic light scattering, fluorescence spectroscopy, and conductometry. It was found that the architecture of the dendrimer, namely the generation, the amount of charged pyridynium groups, the hydrophobic phenylene units, and the rigidity of the spatial structure, determined the special features of the dendrimer–liposome interactions. The binding of D350+ and D229+ with almost fully charged peripheries to liposomes was due to electrostatic forces: the dendrimer molecules could be removed from the liposomal surfaces by NaCl addition. D350+ and D229+ did not display a disruptive effect toward membranes, did not penetrate into the hydrophobic lipid bilayer, and were able to migrate between liposomes. For D215+, a dendrimer with a mixed periphery, hydrophobic interactions of phenylene units with the hydrocarbon tails of lipids were observed, along with electrostatic complexation with liposomes. As a result, defects were formed in the bilayer, which led to irreversible interactions with lipid membranes wherein there was no migration of D215+ between liposomes. A first-generation dendrimer, D16+, which was characterized by small size, a high degree of hydrophobicity, and a rigid structure, when interacting with liposomes caused significant destruction of liposomal membranes. Evidently, this interaction was irreversible: the addition of salt did not lead to the dissociation of the complex. Full article
(This article belongs to the Special Issue Polymer Nanomaterials in Biomedicine)
Show Figures

Figure 1

Figure 1
<p>Structures of cationic pyridylphenylene dendrimers. Dendrimers have different hydrophobicity depending on the amount of charged pyridine groups. D<sub>3</sub><sup>50+</sup> is a third-generation dendrimer with a completely charged periphery, D<sub>2</sub><sup>29+</sup> and D<sub>2</sub><sup>15+</sup> are second-generation dendrimers characterized by completely charged (for D<sub>2</sub><sup>29+</sup>) and mixed (for D<sub>2</sub><sup>15+</sup>) periphery. A first-generation dendrimer with a mixed periphery, D<sub>1</sub><sup>6+</sup>, is characterized by small size and a high degree of hydrophobicity.</p>
Full article ">Figure 2
<p>EPMs of the dendrimer–liposome complexes versus ratios of ionic groups [+]/[−] for D<sub>3</sub><sup>50+</sup> (1), D<sub>2</sub><sup>29+</sup> (2), D<sub>2</sub><sup>15+</sup> (3), and D<sub>1</sub><sup>6+</sup> (4), where [+] is the molar concentration of the pyridinium groups of dendrimers, and [−] is the molar concentration of negatively charged cardiolipin headgroups. CL/Chol/DOPC liposomes, [CL] = 1.5 × 10<sup>−4</sup> mol/L, total lipid concentration of 1 mg/mL. 10<sup>−3</sup> M phosphate buffer, pH 7.2.</p>
Full article ">Figure 3
<p>Hydrodynamic diameters of dendrimer–liposome complexes versus ratios of ionic groups [+]/[−] for D<sub>3</sub><sup>50+</sup> (1), D<sub>2</sub><sup>29+</sup> (2), D<sub>2</sub><sup>15+</sup> (3), and D<sub>1</sub><sup>6+</sup> (4), where [+] is the molar concentration of the pyridinium groups of dendrimers, and [−] is the molar concentration of negatively charged cardiolipin headgroups. CL/Chol/DOPC liposomes, [CL] = 1.5 × 10<sup>−4</sup> mol/L, total lipid concentration of 1 mg/mL. 10<sup>−3</sup> M phosphate buffer, pH 7.2.</p>
Full article ">Figure 4
<p>Dependence of concentration of unbound dendrimers on ratio of ionic groups [+]/[−] for D<sub>3</sub><sup>50+</sup> (1), D<sub>2</sub><sup>29+</sup> (2), D<sub>2</sub><sup>15+</sup> (3), and D<sub>1</sub><sup>6+</sup> (4), where [+] is the molar concentration of the pyridinium groups of dendrimers, and [−] is the molar concentration of negatively charged cardiolipin headgroups. CL/Chol/DOPC liposomes, [CL] = 1.5 × 10<sup>−4</sup> mol/L, total lipid concentration of 1 mg/mL. 10<sup>−3</sup> M phosphate buffer, pH 7.2.</p>
Full article ">Figure 5
<p>Time-dependent changes in relative conductivity of dendrimer–NaCl-loaded liposome complexes for D<sub>3</sub><sup>50+</sup> (1), D<sub>2</sub><sup>29+</sup> (2), D<sub>2</sub><sup>15+</sup> (3), and D<sub>1</sub><sup>6+</sup> (4). The ratio of the ionic groups was [+]/[−] = 2. CL/Chol/DOPC liposomes, [CL] = 1.5 × 10<sup>−4</sup> mol/L, total lipid concentration of 1 mg/mL. 10<sup>−3</sup> M phosphate buffer, pH 7.2.</p>
Full article ">Figure 6
<p>Schematic representation of interactions of liposomes with dendrimers. D<sub>3</sub><sup>50+</sup> and D<sub>2</sub><sup>29+</sup> did not display a disruptive effect toward the liposomes, while the complexation of liposomes with D<sub>2</sub><sup>29+</sup> was followed by minor defect formation. D<sub>1</sub><sup>6+</sup> induced significant destruction of liposomal membranes.</p>
Full article ">Figure 7
<p>Relative fluorescence intensity of label (DOPE-CF) in dendrimer–liposome complexes as a function of ratios of ionic groups [+]/[−] for D<sub>3</sub><sup>50+</sup> (1, 5), D<sub>2</sub><sup>29+</sup> (2, 6), D<sub>2</sub><sup>15+</sup> (3, 7), and D<sub>1</sub><sup>6+</sup> (4, 8) before (1–4) and after (5–8) NaCl addition. [NaCl] = 0.4 mol/L. CL/Chol/DOPC liposomes, [CL] = 1.5 × 10<sup>−4</sup> mol/L, total lipid concentration of 1 mg/mL. 10<sup>−3</sup> M phosphate buffer, pH 7.2.</p>
Full article ">Figure 8
<p>Diagrams of distribution of dendrimers between labeled (Lip*) and unlabeled (Lip) liposomes in 1.5 h for D<sub>3</sub><sup>50+</sup> (<b>a</b>), D<sub>2</sub><sup>29+</sup> (<b>b</b>), and D<sub>2</sub><sup>15+</sup> (<b>c</b>). The ratios of the ionic groups were [+]/[−] = 0.5 (1) and 1 (2). CL/Chol/DOPC liposomes, [CL] = 1.5 × 10<sup>−4</sup> mol/L, total lipid concentration of 1 mg/mL. 10<sup>−3</sup> M phosphate buffer, pH 7.2.</p>
Full article ">Figure 9
<p>Schematic representation of the migration experiment for D<sub>3</sub><sup>50+</sup> and D<sub>2</sub><sup>29+</sup> (<b>a</b>), as well as for D<sub>2</sub><sup>15+</sup> (<b>b</b>). D<sub>3</sub><sup>50+</sup> and D<sub>2</sub><sup>29+</sup> were able to migrate between liposomes since the binding was due to electrostatic forces (<b>a</b>). For D<sub>2</sub><sup>15+</sup>, hydrophobic interactions were observed, along with electrostatic complexation; as a result, defects were formed in the membrane that led to irreversible interactions with no migration of D<sub>2</sub><sup>15+</sup> between liposomes (<b>b</b>). The dendrimer molecules are indicated as circles, and the fluorescent labels are indicated as green stars.</p>
Full article ">
19 pages, 4139 KiB  
Article
Effect of Ionization Degree of Poly(amidoamine) Dendrimer and 5-Fluorouracil on the Efficiency of Complex Formation—A Theoretical and Experimental Approach
by Magdalena Szota, Pawel Wolski, Cristina Carucci, Flaminia Cesare Marincola, Jacek Gurgul, Tomasz Panczyk, Andrea Salis and Barbara Jachimska
Int. J. Mol. Sci. 2023, 24(1), 819; https://doi.org/10.3390/ijms24010819 - 3 Jan 2023
Cited by 7 | Viewed by 2841
Abstract
Due to their unique structure, poly(amidoamine) (PAMAM) dendrimers can bind active ingredients in two ways: inside the structure or on their surface. The location of drug molecules significantly impacts the kinetics of active substance release and the mechanism of internalization into the cell. [...] Read more.
Due to their unique structure, poly(amidoamine) (PAMAM) dendrimers can bind active ingredients in two ways: inside the structure or on their surface. The location of drug molecules significantly impacts the kinetics of active substance release and the mechanism of internalization into the cell. This study focuses on the effect of the protonation degree of the G4PAMAM dendrimer and the anticancer drug 5-fluorouracil (5FU) on the efficiency of complex formation. The most favorable conditions for constructing the G4PAMAM-5FU complex are a low degree of protonation of the dendrimer molecule with the drug simultaneously present in a deprotonated form. The fluorine components in the XPS spectra confirm the formation of the stable complex. Through SAXS and DLS methods, a decrease in the dendrimer’s molecular size resulting from protonation changes at alkaline conditions was demonstrated. The gradual closure of the dendrimer structure observed at high pH values makes it difficult for the 5FU molecules to migrate to the interior of the support structure, thereby promoting drug immobilization on the surface. The 1H NMR and DOSY spectra indicate that electrostatic interactions determine the complex formation process. Through MD simulations, the localization profile and the number of 5FU molecules forming the complex were visualized on an atomic scale. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The initial evaluation of cytotoxicity of dendrimers and their complexes with 5FU against mouse fibroblasts (L929), glioblastoma (SNB-19), prostate cancer (Du-145), and colon adenocarcinoma (HT-29) determined via metabolic activity assay of the cells cultured for 7 days in the presence of G4PAMAM dendrimers, G4-5FU complexes, and 5FU, with concentration corresponding to its concentration in complex [<a href="#B15-ijms-24-00819" class="html-bibr">15</a>].</p>
Full article ">Figure 2
<p>SAXS curves of G4PAMAM dendrimer 2.5% in 1 × 10<sup>−2</sup> NaCl 10 at different pH.</p>
Full article ">Figure 3
<p><sup>1</sup>H NMR spectra of G4PAMAM (bottom), 5FU (top), and dendrimer–drug mixtures at different molar ratios and pH (G4PAMAM = 2 mg/mL; 50:50 vol% H<sub>2</sub>O/D<sub>2</sub>O; 30 °C). Labels: a–d denote the methylene protons in the core of dendrimer; a’–d’ denote the methylene proton in the outmost layer of the dendrimer; e denotes the hydrogen occupying position 6 in 5FU. The dotted lines serve to guide the eye only.</p>
Full article ">Figure 4
<p>The XPS spectra of G4PAMAM dendrimer, 5-fluorouracil, and G4-5FU layers deposited on Au surface in the BE regions of C 1s (<b>a</b>), N 1s (<b>b</b>), O 1s (<b>c</b>), and F 1s (<b>d</b>).</p>
Full article ">Figure 5
<p>Final structures of the mixed systems: dendrimer (blue) and 100 5FU molecules (yellow).</p>
Full article ">Figure 6
<p>The instantaneous number of 5FU molecules bound to the dendrimer as a function of time obtained in the last 10 ns of simulations.</p>
Full article ">Figure 7
<p>Radial distribution functions determined for the dendrimer–drug molecule distances.</p>
Full article ">Figure 8
<p>(<b>A</b>) Schematic illustration of chemical linkages in the first-generation PAMAM dendrimers. Higher-level generations can be built by repeating inner monomer units. Red sticks denote the linkage between core, inner monomers, and terminal groups, while red N atoms denote nitrogen atoms which can be protonated. (<b>B</b>) Chemical structure of the anionic form of the 5-fluorouracil molecule used in simulations.</p>
Full article ">
17 pages, 1687 KiB  
Article
Mitigating Cardiotoxicity of Dendrimers: Angiotensin-(1-7) via Its Mas Receptor Ameliorates PAMAM-Induced Cardiac Dysfunction in the Isolated Mammalian Heart
by Saghir Akhtar, Fawzi Babiker, Usman A. Akhtar and Ibrahim F. Benter
Pharmaceutics 2022, 14(12), 2673; https://doi.org/10.3390/pharmaceutics14122673 - 1 Dec 2022
Cited by 5 | Viewed by 1737
Abstract
Aim: The influence of the physiochemical properties of dendrimer nanoparticles on cardiac contractility and hemodynamics are not known. Herein, we investigated (a) the effect of polyamidoamine (PAMAM) dendrimer generation (G7, G6, G5, G4 and G3) and surface chemistry (-NH2, -COOH and [...] Read more.
Aim: The influence of the physiochemical properties of dendrimer nanoparticles on cardiac contractility and hemodynamics are not known. Herein, we investigated (a) the effect of polyamidoamine (PAMAM) dendrimer generation (G7, G6, G5, G4 and G3) and surface chemistry (-NH2, -COOH and -OH) on cardiac function in mammalian hearts following ischemia-reperfusion (I/R) injury, and (b) determined if any PAMAM-induced cardiotoxicity could be mitigated by Angiotensin-(1-7) (Ang-(1-7), a cardioprotective agent. Methods: Hearts isolated from male Wistar rats underwent regional I/R and/or treatment with different PAMAM dendrimers, Ang-(1-7) or its MAS receptors antagonists. Thirty minutes of regional ischemia through ligation of the left anterior descending coronary artery was followed by 30 min of reperfusion. All treatments were initiated 5 min prior to reperfusion and maintained during the first 10 min of reperfusion. Cardiac function parameters for left ventricular contractility, hemodynamics and vascular dynamics data were acquired digitally, whereas cardiac enzymes and infarct size were used as measures of cardiac injury. Results: Treatment of isolated hearts with increasing doses of G7 PAMAM dendrimer progressively exacerbated recovery of cardiac contractility and hemodynamic parameters post-I/R injury. Impairment of cardiac function was progressively less on decreasing dendrimer generation with G3 exhibiting little or no cardiotoxicity. Cationic PAMAMs (-NH2) were more toxic than anionic (-COOH), with neutral PAMAMs (-OH) exhibiting the least cardiotoxicity. Cationic G7 PAMAM-induced cardiac dysfunction was significantly reversed by Ang-(1-7) administration. These cardioprotective effects of Ang-(1-7) were significantly revoked by administration of the MAS receptor antagonists, A779 and D-Pro7-Ang-(1-7). Conclusions: PAMAM dendrimers can impair the recovery of hearts from I/R injury in a dose-, dendrimer-generation-(size) and surface-charge dependent manner. Importantly, PAMAM-induced cardiotoxicity could be mitigated by Ang-(1-7) acting through its MAS receptor. Thus, this study highlights the activation of Ang-(1-7)/Mas receptor axis as a novel strategy to overcome dendrimer-induced cardiotoxicity. Full article
(This article belongs to the Special Issue Engineering and Characterisation of Novel Nanomedicine Formulations)
Show Figures

Figure 1

Figure 1
<p>Schematic representation showing the experimental protocols used in the study (n = 8). A: Untreated ischemia-reperfusion control (C). B: Dose response relationship for the G7 PAMAM dendrimer. C: Effect of the dendrimer size (generation) on heart subjected to ischemia and reperfusion. D: Effect of the surface charge/chemistry of dendrimers on hearts subjected to ischemia and reperfusion. E: Effect of the G7 PAMAM dendrimer in the presence or absence of Ang-(1-7) and its Mas receptors antagonists on the effects of ischemia and reperfusion.</p>
Full article ">Figure 2
<p>Dose-dependent recovery of cardiac function parameters following I/R upon acute administration of different doses of G7 cationic PAMAM (1.0 through 20 µg/mL). Percent recovery of cardiac function data (<b>a</b>–<b>f</b>) following I/R for left ventricle function (DPmax (<b>a</b>) and LVEDP (<b>b</b>)), contractility indices (+dP/dt (<b>c</b>) and −dP/dt (<b>d</b>) and coronary vascular dynamics (CF (<b>e</b>) and CVR (<b>f</b>)) are shown. The data were computed after 30 min reperfusion and expressed as the mean ± SEM. DPmax: maximum developed pressure; LVEDP: left ventricular end-diastolic pressure; CF: coronary flow; CVR: coronary vascular resistance. Control hearts, C = I/R alone. N = 8. Mean ± SEM. Asterix (*) indicates significant difference (<span class="html-italic">p</span> &lt; 0.05) from controls.</p>
Full article ">Figure 3
<p>PAMAM-induced impairment in cardiac function is dependent on physicochemical properties of molecular size (generation) and surface charge of PAMAM dendrimers. Post I/R recovery in the left ventricle function (DPmax (<b>a</b>) and LVEDP (<b>b</b>)), contractility indices (+dP/dt (<b>c</b>) and –dP/dt (<b>d</b>)) and coronary vascular dynamics (CF (<b>e</b>) and CVR (<b>f</b>)) after treatment with various PAMAM dendrimer generations with variable molecular sizes (G3, G4, G5, G6, G7) or variable surface charge (cationic G6, anionic G5.5 or neutral G6). The data were computed after 30 min reperfusion and expressed as the mean ± SEM. DPmax: maximum developed pressure; LVEDP: left ventricular end-diastolic pressure; CF: coronary flow; CVR: coronary vascular resistance; G3: third generation PAMAM dendrimer; G4: fourth generation PAMAM; G5: fifth generation PAMAM; G6: sixth generation PAMAM; G7: seventh generation PAMAM G4. Control hearts, C = I/R alone. N = 8. Asterix (*) indicates significant difference (<span class="html-italic">p</span> &lt; 0.05) from controls.</p>
Full article ">Figure 4
<p>Ang-(1-7) via its Mas Receptor rescues cationic G7 PAMAM-induced impairment of cardiac function (<b>a</b>–<b>f</b>). Post I/R recovery in the left ventricle function (DPmax (<b>a</b>) and LVEDP (<b>b</b>)), contractility indices (+dP/dt (<b>c</b>) and −dP/dt (<b>d</b>)) and coronary vascular dynamics (CF (<b>e</b>) and CVR (<b>f</b>)) after treatment with G7 PAMAM in presence or absence of Ang-(1-7) and its Mas receptor blockers D-Pro and A779. The data were computed after 30 min reperfusion and expressed as the mean ± SEM. DPmax: maximum developed pressure; LVEDP: left ventricular end-diastolic pressure; CF: coronary flow; CVR: coronary vascular resistance; Ang-(1-7): angiotensin-91-7); D-Pro: Ang-(1-7) selective antagonist; A779: Ang-(1-7) selective antagonist (see <a href="#sec2-pharmaceutics-14-02673" class="html-sec">Section 2</a>). Double Asterix ** refers to significant difference (<span class="html-italic">p</span> &lt; 0.05) compared to control I/R alone and single Asterix * refers to significant difference (<span class="html-italic">p</span> &lt; 0.05) compared G7 values. Dollar sign (<span>$</span>) indicates significant difference (<span class="html-italic">p</span> &lt; 0.05) compared to G7 + Ang-(1-7).</p>
Full article ">Figure 5
<p>Ang-(1-7) via its Mas Receptor rescues cationic G7 PAMAM-induced myocardial infarction. Infarct size post-I/R injury was determined after treatment with G7 PAMAM in the presence or absence of Ang-(1-7) and its Mas receptor blockers D-Pro and A779 (n = 4). Top panel: representative 2,3,5-triphenyl-2H-tetrazolium chloride-stained heart slices for each treatment condition. Bottom Panel: measured infarct size, normalized to the LV area, in isolated rat hearts at the end of reperfusion. C: control; G7: seventh generation cationic PAMAM dendrimer; Ang-(1-7): angiotensin-1-7; D-Pro: Ang-(1-7) selective antagonist; A779: Ang-(1-7) selective antagonist. Single Asterix * refers to significant difference (<span class="html-italic">p</span> &lt; 0.05) compared to control I/R alone and double Asterix ** to significant difference (<span class="html-italic">p</span> &lt; 0.05) compared to G7 values.</p>
Full article ">Figure 6
<p>A schematic summary of the impact of different PAMAMs in the mammalian heart and the ability of Ang-(1-7) to mitigate their cardiotoxicity. In isolated rat hearts subjected to I/R injury, administration of PAMAM dendrimers exacerbated recovery of cardiac function in terms of LV contractility and hemodynamics parameters as well by increasing infarct size and cardiac enzyme levels (LDH and CK)—hallmarks of cardiac damage and toxicity. These effects of PAMAMs were dependent on dendrimer generation (G7 &gt; G6 &gt; G5 &gt; G4 &gt; G3) and surface charge ((-NH<sub>2</sub> (cationic) &gt; -COOH (anionic) &gt; -OH (neutral)). The adjunct administration of Ang-(1-7) rescued the cardiotoxicity caused by cationic PAMAM dendrimers. The beneficial effects of Ang-(1-7) were revoked by two Mas receptor (MasR) antagonists (A779 and D-Pro), confirming that Ang-(1-7) actions were, at least in part, mediated through MasR. Thus, Ang-(1-7) may represent a viable strategy to mitigate the cardiotoxicity of PAMAM dendrimers.</p>
Full article ">
14 pages, 1799 KiB  
Review
A Review on Increasing the Targeting of PAMAM as Carriers in Glioma Therapy
by Xingyue Li, Wenjing Ta, Ruochen Hua, Jihong Song and Wen Lu
Biomedicines 2022, 10(10), 2455; https://doi.org/10.3390/biomedicines10102455 - 1 Oct 2022
Cited by 8 | Viewed by 2403
Abstract
Glioma is an invasive brain cancer, and it is difficult to achieve desired therapeutic effects due to the high postoperative recurrence rate and limited efficacy of drug therapy hindered by the biological barrier of brain tissue. Nanodrug delivery systems are of great interest, [...] Read more.
Glioma is an invasive brain cancer, and it is difficult to achieve desired therapeutic effects due to the high postoperative recurrence rate and limited efficacy of drug therapy hindered by the biological barrier of brain tissue. Nanodrug delivery systems are of great interest, and many efforts have been made to utilize them for glioma treatment. Polyamidoamine (PAMAM), a starburst dendrimer, provides malleable molecular size, functionalized molecular structure and penetrable brain barrier characteristics. Therefore, PAMAM-based nanodrug delivery systems (PAMAM DDS) are preferred for glioma treatment research. In this review, experimental studies on PAMAM DDS for glioma therapy were focused on and summarized. Emphasis was given to three major topics: methods of drug loading, linkers between drug/ligand and PAMAM and ligands of modified PAMAM. A strategy for well-designed PAMAM DDS for glioma treatment was proposed. Purposefully understanding the physicochemical and structural characteristics of drugs is necessary for selecting drug loading methods and achieving high drug loading capacity. Additionally, functional ligands contribute to achieving the brain targeting, brain penetration and low toxicity of PAMAM DDS. Furthermore, a brilliant linker facilitates multidrug combination and multifunctional PAMAM DDS. PAMAM DDS show excellent promise as drug vehicles and will be further studied for product development and safety evaluation. Full article
(This article belongs to the Special Issue Targeted Drug Delivery to Brain)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic diagram of the structure of PAMAM. Schematic showing the chemical structure of PAMAM, including the core, terminal groups and different generations. EDA: ethylenediamine; DAB: diaminobutane; DAH: 1,5-diaminohexane.</p>
Full article ">Figure 2
<p>Methods of loading drugs in PAMAM DDS. Drugs can be embedded in the PAMAM’s cavity or complexed on the surface of PAMAM or covalently bound in the arm of PAMAM. SiRNA: small interfering RNA; miRNA-7: microRNA-7; TRAIL: tumor-necrosis-factor-related apoptosis-inducing ligand; IFN-β: interferon-β.</p>
Full article ">Figure 3
<p>Schematic diagram of the formation process of different linkers between drugs and PAMAM. Different background colors represent the synthesis of linkers with different release properties: pink for acid-sensitive, blue for non-acid-sensitive and purple for GSH-sensitive. DOX is taken as an example of a drug. DOX: doxorubicin; BMPH: N-(β-maleimidopropionic acid) hydrazide; SPDP: N-succinimidyl-3-(2-pyridyldithio) proprionate.</p>
Full article ">Figure 4
<p>Schematic representation of targeting ligands and their receptors or targeting sites on the PAMAM DDS. NRP-1: neuropilin 1; MMP-2: matrix metalloproteinase-2; METr: mesenchymal transition factor receptor; Tfr: transferrin receptor; FAr: folic acid receptor; CTX: chlorotoxin; Tf: transferrin; FA: folic acid; PMPC: poly (2-methacryloyloxyethyl phosphorylcholine); BMP: bacterial magnetic nanoparticles; LRP 1: low-density lipoprotein receptor-related protein 1.</p>
Full article ">
21 pages, 3575 KiB  
Article
Retinal Delivery of the Protein Kinase C-β Inhibitor Ruboxistaurin Using Non-Invasive Nanoparticles of Polyamidoamine Dendrimers
by Rehab A. Alshammari, Fadilah S. Aleanizy, Amal Aldarwesh, Fulwah Y. Alqahtani, Wael A. Mahdi, Bushra Alquadeib, Qamraa H. Alqahtani, Nazrul Haq, Faiyaz Shakeel, Hosam G. Abdelhady and Ibrahim A. Alsarra
Pharmaceutics 2022, 14(7), 1444; https://doi.org/10.3390/pharmaceutics14071444 - 11 Jul 2022
Cited by 13 | Viewed by 2142
Abstract
Ruboxistaurin (RBX) is an anti-vascular endothelial growth factor (anti-VEGF) agent that is used in the treatment of diabetic retinopathy and is mainly given intravitreally. To provide a safe and effective method for RBX administration, this study was designed to develop RBX nanoparticles using [...] Read more.
Ruboxistaurin (RBX) is an anti-vascular endothelial growth factor (anti-VEGF) agent that is used in the treatment of diabetic retinopathy and is mainly given intravitreally. To provide a safe and effective method for RBX administration, this study was designed to develop RBX nanoparticles using polyamidoamine (PAMAM) dendrimer generation 5 for the treatment of diabetic retinopathy. Drug loading efficiency, and in vitro release of proposed complexes of RBX: PAMAM dendrimers were determined and the complexation ratio that showed the highest possible loading efficiency was selected. The drug loading efficiency (%) of 1:1, 2.5:1, and 5:1 complexes was 89.2%, 96.4%, and 97.6%, respectively. Loading capacities of 1:1, 2.5:1, and 5:1 complexes were 1.6%, 4.0%, and 7.2% respectively. In comparison, the 5:1 complex showed the best results in the aforementioned measurements. The in vitro release studies showed that in 8 h, the RBX release from 1:1, 2.5:1, and 5:1 complexes was 37.5%, 35.9%, and 77.0%, respectively. In particular, 5:1 complex showed the highest drug release. In addition, particle size measurements showed that the diameter of empty PAMAM dendrimers was 214.9 ± 8.5 nm, whereas the diameters of loaded PAMAM dendrimers in 1:1, 2.5:1, 5:1 complexes were found to be 461.0 ± 6.4, 482.4 ± 12.5, and 420.0 ± 7.1 nm, respectively. Polydispersity index (PDI) showed that there were no significant changes in the PDI between the free and loaded PAMAM dendrimers. The zeta potential measurements showed that the free and loaded nanoparticles possessed neutral charges due to the presence of anionic and cationic terminal structures. Furthermore, the safety of this formulation was apparent on the viability of the MIO-M1 cell lines. This nanoformulation will improve the therapeutic outcomes of anti-VEGF therapy and the bioavailability of RBX to prevent vision loss in patients with diabetic retinopathy. Full article
(This article belongs to the Special Issue Novel Approaches for Overcoming Biological Barriers)
Show Figures

Figure 1

Figure 1
<p>In vitro release profiles of different complexes (mean ± SD, <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 2
<p>Scanning electron microscope (SEM) overview image of (<b>A</b>) G4.5 complex 25:1 and (<b>B</b>) G5 complex 25:1.</p>
Full article ">Figure 3
<p>Transmission electron microscope (TEM) overview images of (<b>A</b>) empty PAMAM dendrimers G4.5 and (<b>B</b>) G4.5 complex 25:1.</p>
Full article ">Figure 4
<p>TEM overview images of (<b>A</b>) empty PAMAM dendrimers G5 and (<b>B</b>) G5 complex 25:1.</p>
Full article ">Figure 5
<p>Effect of RBX after 24 h exposure on the cell viability of MIO-M1 cells under controlled conditions (mean ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Effect of (<b>A</b>) PAMAM dendrimers G4.5 and (<b>B</b>) PAMAM dendrimers G5 after 24 h. Exposure on the cell viability of MIO-M1 cells under controlled conditions (mean ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 7
<p>Effect of G4.5 complex 25:1 (<b>A</b>) and G5 complex 25:1 (<b>B</b>) after 24 h exposure on the cell viability of MIO-M1 cells under controlled conditions (mean ± SD; <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 8
<p>Effect of the tested compounds after (<b>A</b>) 24 h and (<b>B</b>) 48 h. Exposure on the cell viability of MIO-M1 cells under controlled and high glucose mediums (mean ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 9
<p>TEER measurements of monolayer cell membrane 24 h after exposure to treatments. Data represent % of TEER mean of the control ± SEM (<span class="html-italic">n</span> = 3–4).</p>
Full article ">Figure 10
<p>Apparent permeability coefficient (Papp) of the MIO-M1 monolayer cell after 24 treatments * <span class="html-italic">p</span> value &lt; 0.01.</p>
Full article ">
29 pages, 3480 KiB  
Review
Applications of Various Types of Nanomaterials for the Treatment of Neurological Disorders
by Abdul Waris, Asmat Ali, Atta Ullah Khan, Muhammad Asim, Doaa Zamel, Kinza Fatima, Abdur Raziq, Muhammad Ajmal Khan, Nazia Akbar, Abdul Baset and Mohammed A. S. Abourehab
Nanomaterials 2022, 12(13), 2140; https://doi.org/10.3390/nano12132140 - 22 Jun 2022
Cited by 49 | Viewed by 7943
Abstract
Neurological disorders (NDs) are recognized as one of the major health concerns globally. According to the World Health Organization (WHO), neurological disorders are one of the main causes of mortality worldwide. Neurological disorders include Alzheimer’s disease, Parkinson′s disease, Huntington′s disease, Amyotrophic lateral sclerosis, [...] Read more.
Neurological disorders (NDs) are recognized as one of the major health concerns globally. According to the World Health Organization (WHO), neurological disorders are one of the main causes of mortality worldwide. Neurological disorders include Alzheimer’s disease, Parkinson′s disease, Huntington′s disease, Amyotrophic lateral sclerosis, Frontotemporal dementia, Prion disease, Brain tumor, Spinal cord injury, and Stroke. These diseases are considered incurable diseases because no specific therapies are available to cross the blood-brain barrier (BBB) and reach the brain in a significant amount for the pharmacological effect in the brain. There is a need for the development of strategies that can improve the efficacy of drugs and circumvent BBB. One of the promising approaches is the use of different types of nano-scale materials. These nano-based drugs have the ability to increase the therapeutic effect, reduce toxicity, exhibit good stability, targeted delivery, and drug loading capacity. Different types and shapes of nanomaterials have been widely used for the treatment of neurological disorders, including quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These nanoparticles have unique characteristics, including sensitivity, selectivity, and the ability to cross the BBB when used in nano-sized particles, and are widely used for imaging studies and treatment of NDs. In this review, we briefly summarized the recent literature on the use of various nanomaterials and their mechanism of action for the treatment of various types of neurological disorders. Full article
(This article belongs to the Special Issue Nanotechnology and Nanomaterials in Biological Systems and Medicine)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic illustration of drug delivery system for the treatment of neurological disorders.</p>
Full article ">Figure 2
<p>Nanomaterials mediated drug delivery of therapeutic agents targeting the brains of patients suffering from Alzheimer’s disease to improve clinical outcomes. Adopted from [<a href="#B99-nanomaterials-12-02140" class="html-bibr">99</a>].</p>
Full article ">Figure 3
<p>Schematic diagram of the possible mechanisms for the treatment of PD. Adopted from [<a href="#B111-nanomaterials-12-02140" class="html-bibr">111</a>]. Copyright Elsevier 2021.</p>
Full article ">Figure 4
<p>Schematic diagram of the neuroprotective effect of nano-Se in a model of <span class="html-italic">C. elegans</span> HD. Reproduced with permission [<a href="#B116-nanomaterials-12-02140" class="html-bibr">116</a>]. Copyright 2019 The American Chemical Society.</p>
Full article ">Figure 5
<p>(<b>A</b>) Rabies virus mimicking silica-coated gold nanorods bypass the BBB via neuronal pathways to treat brain disease. (<b>B</b>) Delivery of therapeutic siRNA to the mouse brain by systemic injection of exosomes. (<b>a</b>) Schematic illustration of the preparation of exosomes; (<b>b</b>) gene silencing efficiency by different vehicles. Adopted from Wang et al., 2020 [<a href="#B123-nanomaterials-12-02140" class="html-bibr">123</a>].</p>
Full article ">Figure 6
<p>Different nanomaterials as drug delivery systems for the treatment of SCI. One of their most important features is their small size (nanoscale), which makes them capable of crossing the blood spinal cord barrier. Adopted from [<a href="#B151-nanomaterials-12-02140" class="html-bibr">151</a>], with permission from Elsevier Limited 2019.</p>
Full article ">Figure 7
<p>Schematic diagram of different nanomaterials for the treatment of stroke. Adopted from Chao Li et al., 2020 [<a href="#B163-nanomaterials-12-02140" class="html-bibr">163</a>].</p>
Full article ">Figure 8
<p>Nanomaterials for the treatment and diagnosis of brain tumors. Adopted from Simona et al., 2020 [<a href="#B183-nanomaterials-12-02140" class="html-bibr">183</a>].</p>
Full article ">
37 pages, 1117 KiB  
Review
Safety Challenges and Application Strategies for the Use of Dendrimers in Medicine
by Xiang Li, Abid Naeem, Shanghua Xiao, Lei Hu, Jing Zhang and Qin Zheng
Pharmaceutics 2022, 14(6), 1292; https://doi.org/10.3390/pharmaceutics14061292 - 17 Jun 2022
Cited by 39 | Viewed by 4245
Abstract
Dendrimers are used for a variety of applications in medicine but, due to their host–guest and entrapment characteristics, are particularly used for the delivery of genes and drugs. However, dendrimers are intrinsically toxic, thus creating a major limitation for their use in biological [...] Read more.
Dendrimers are used for a variety of applications in medicine but, due to their host–guest and entrapment characteristics, are particularly used for the delivery of genes and drugs. However, dendrimers are intrinsically toxic, thus creating a major limitation for their use in biological systems. To reduce such toxicity, biocompatible dendrimers have been designed and synthesized, and surface engineering has been used to create advantageous changes at the periphery of dendrimers. Although dendrimers have been reviewed previously in the literature, there has yet to be a systematic and comprehensive review of the harmful effects of dendrimers. In this review, we describe the routes of dendrimer exposure and their distribution in vivo. Then, we discuss the toxicity of dendrimers at the organ, cellular, and sub-cellular levels. In this review, we also describe how technology can be used to reduce dendrimer toxicity, by changing their size and surface functionalization, how dendrimers can be combined with other materials to generate a composite formulation, and how dendrimers can be used for the diagnosis of disease. Finally, we discuss future challenges, developments, and research directions in developing biocompatible and safe dendrimers for medical purposes. Full article
(This article belongs to the Special Issue Recent Advances in Dendrimer Nanomedicine)
Show Figures

Figure 1

Figure 1
<p>The structures of four major monomeric dendrimers: (<b>a</b>) Poly (amidoamine); (<b>b</b>) Poly (propylene imine); (<b>c</b>) Carbosilane; (<b>d</b>) Chiral dendrimers.</p>
Full article ">Figure 2
<p>Schematic representation of a PAMAM core–shell TECTO dendrimer.</p>
Full article ">Figure 3
<p>Typical structure of a peptide dendrimer.</p>
Full article ">Figure 4
<p>Different types of glyco-dendrimers.</p>
Full article ">
Back to TopTop