[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
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (99)

Search Parameters:
Keywords = long-acting drug delivery

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 697 KiB  
Systematic Review
Investigations of Long-Acting Formulations in Children, Adolescents, and Pregnant Women: A Systematic Review
by Lynn Bertagnolli, Zhengyi Deng, Melissa Davy-Rothwell, Elaine J. Abrams, Charles Flexner and Ethel D. Weld
Pharmaceutics 2025, 17(1), 113; https://doi.org/10.3390/pharmaceutics17010113 - 15 Jan 2025
Viewed by 379
Abstract
Background/Objectives: Long-acting and extended-release drug delivery strategies have greatly improved treatment for a variety of medical conditions. Special populations, specifically infants, children, young people, and pregnant and postpartum women, could greatly benefit from access to these strategies but are often excluded from clinical [...] Read more.
Background/Objectives: Long-acting and extended-release drug delivery strategies have greatly improved treatment for a variety of medical conditions. Special populations, specifically infants, children, young people, and pregnant and postpartum women, could greatly benefit from access to these strategies but are often excluded from clinical trials. We conducted a systematic review of all clinical studies involving the use of a long-acting intramuscular injection or implant in infants, children, young people, and pregnant and postpartum people. Methods: Pubmed, Embase, and Cochrane Library trials were searched. Studies published from 1980 through 2018 were included. After abstract review and duplication removal, full-text articles were obtained for further review, reviewed by two independent reviewers, and disagreements were resolved by a third reviewer. Results: a total of 101 studies of long-acting therapeutics were completed in these populations, and most (80%) of these had a sample size of <100 individuals. Therapeutics for only a small pool of indications were examined in these studies, with 72% of the studies investigating hormonal contraception or other types of hormonal treatments. Only 9.3% of the studies in children and 16.7% of the studies in pregnant people collected any pharmacokinetic (PK) data. Conclusions: Long-acting formulations may behave differently (both pharmacokinetically and pharmacodynamically) in childhood, adolescence, and pregnancy as compared to non-pregnant adulthood. Therefore, it is imperative to increase and improve upon the studies investigating long-acting formulations in order to close the knowledge gap and improve care and treatment in these special populations. Full article
(This article belongs to the Section Physical Pharmacy and Formulation)
Show Figures

Figure 1

Figure 1
<p>Preferred reporting items for systematic reviews and meta-analyses (PRISMA) diagram.</p>
Full article ">Figure 2
<p>Histogram of sample sizes for included studies of long-acting therapeutics.</p>
Full article ">
17 pages, 4451 KiB  
Article
Design of Nanocrystalline Suspension of Dutasteride for Intramuscular Prolonged Delivery
by Min Young Jeong, Doe Myung Shin, Min Kyeong Kwon, Ye Bin Shin, Jun Soo Park, In Gyu Yang, Jin Hyuk Myung, Dong Geon Lee, Gi Yeong Lee, Chae Won Park, Ji Won Yeo, Myoung Jin Ho, Yong Seok Choi and Myung Joo Kang
Nanomaterials 2024, 14(22), 1781; https://doi.org/10.3390/nano14221781 - 5 Nov 2024
Viewed by 996
Abstract
The aim of the study is to formulate an injectable nanocrystalline suspension (NS) of dutasteride (DTS), a hydrophobic 5α-reductase inhibitor used to treat benign prostatic hyperplasia and scalp hair loss, for parenteral long-acting delivery. A DTS-loaded NS (DTS-NS, 40 mg/mL DTS) was prepared [...] Read more.
The aim of the study is to formulate an injectable nanocrystalline suspension (NS) of dutasteride (DTS), a hydrophobic 5α-reductase inhibitor used to treat benign prostatic hyperplasia and scalp hair loss, for parenteral long-acting delivery. A DTS-loaded NS (DTS-NS, 40 mg/mL DTS) was prepared using a lab-scale bead-milling technique. The optimized DTS-NS prepared using Tween 80 (0.5% w/v) as a nano-suspending agent, was characterized as follows: rod/rectangular shape; particle size of 324 nm; zeta potential of −11 mV; and decreased drug crystallinity compared with intact drug powder. The DTS-NS exhibited a markedly protracted drug concentration-time profile following intramuscular injection, reaching a maximum concentration after 8.40 days, with an elimination half-life of 9.94 days in rats. Histopathological observations revealed a granulomatous inflammatory response at the injection site 7 days after intramuscular administration, which significantly subsided by day 14 and showed minimal inflammation by day 28. These findings suggest that the nanosuspension system is a promising approach for the sustained release parenteral DTS delivery, with a protracted pharmacokinetic profile and tolerable local inflammation. Full article
Show Figures

Figure 1

Figure 1
<p>Illustration of DTS-NS manufacturing process using dual centrifugation-based bead-milling technique.</p>
Full article ">Figure 2
<p>Influence of suspending agents on (<b>a</b>) the appearance and (<b>b</b>) particle size and homogeneity of DTS-NS formulas prepared using a lab-scale bead-milling technology. Notes: (<b>a</b>) All images were captured on glass slides within 10 min after fabrication (scale bar: 5 mm). (<b>b</b>) The concentrations of DTS and suspending agent were fixed at 40 mg/mL and 5 mg/mL, respectively. Bead milling was carried out at 1500 rpm for 1 h under conditions of −10 °C. Data represent means ± SD (n = 3).</p>
Full article ">Figure 3
<p>Effect of the concentration of Tween 80 on the crystal size of DTS-NSs prepared using a lab-scale bead-milling technology. (<b>a</b>) The appearance of drug suspension prepared with different Tween 80 concentrations. (<b>b</b>) Effect of Tween 80 concentration on mean particle size and homogeneity. Notes: (<b>a</b>) All images were captured on glass slides 10 min after preparation (scale bar: 5 mm). (<b>b</b>) Concentrations of DTS and Tween 80 were set at 40 and 0–10 mg/mL, respectively. Bead milling was performed at 1000–2000 rpm for 1 h under conditions of −10 °C. Data are presented as means ± SD (n = 3).</p>
Full article ">Figure 4
<p>Morphological and physical characteristics of DTS-NS. Scanning electron microscopic (SEM) image of (<b>a</b>) raw material and (<b>b</b>) DTS-NS. (<b>c</b>) Particle size distribution and (<b>d</b>) zeta potential of DTS-NS. Representative (<b>e</b>) XRD patterns and (<b>f</b>) DSC curves of the raw material, aqueous vehicle, and DTS-NS.</p>
Full article ">Figure 4 Cont.
<p>Morphological and physical characteristics of DTS-NS. Scanning electron microscopic (SEM) image of (<b>a</b>) raw material and (<b>b</b>) DTS-NS. (<b>c</b>) Particle size distribution and (<b>d</b>) zeta potential of DTS-NS. Representative (<b>e</b>) XRD patterns and (<b>f</b>) DSC curves of the raw material, aqueous vehicle, and DTS-NS.</p>
Full article ">Figure 5
<p>In vitro dissolution profile of DTS raw material and DTS-NS under sink condition. The sink condition was guaranteed by adding 2% <span class="html-italic">w</span>/<span class="html-italic">v</span> Cremophor EL to 10 mM sodium phosphate buffer (pH 7.4). Note: Data represent mean ± SD (n = 3).</p>
Full article ">Figure 6
<p>Multple reaction monitoring chromatograms of double blank rat plasma (<b>a</b>), 50 ng/mL of dutasteride-spiked rat plasma (<b>b</b>), rat plasma taken five minutes after dutasteride intravenous (IV) administration (<b>c</b>), and rat plasma taken eight hours after dutasteride IV administration (<b>d</b>). DTS and IS stand for dutasteride and finasteride (internal standard), respectively.</p>
Full article ">Figure 6 Cont.
<p>Multple reaction monitoring chromatograms of double blank rat plasma (<b>a</b>), 50 ng/mL of dutasteride-spiked rat plasma (<b>b</b>), rat plasma taken five minutes after dutasteride intravenous (IV) administration (<b>c</b>), and rat plasma taken eight hours after dutasteride IV administration (<b>d</b>). DTS and IS stand for dutasteride and finasteride (internal standard), respectively.</p>
Full article ">Figure 7
<p>The plasma concentration–time profiles of DTS following IM injection (5 mg/kg as DTS) of DTS-NS and IV injection (0.2 mg/kg as DTS) of DTS solution in rats. Note: Data represent mean ± SD (n = 5).</p>
Full article ">Figure 8
<p>Histological examination of gastrocnemius muscles stained with hematoxylin and eosin (H&amp;E) following intramuscular (IM) injection of negative control (normal saline) or DTS-NS in rats. Note: The depot formed by injection of DTS-NS is indicated by (*). Red double arrows indicate fibroblastic bands, and (a) denotes angiogenesis.</p>
Full article ">
14 pages, 5518 KiB  
Article
In Vitro Analysis of Aerodynamic Properties and Co-Deposition of a Fixed-Dose Combination of Fluticasone Furoate, Umeclidinium Bromide, and Vilanterol Trifenatate
by Kittipong Maneechotesuwan, Somchai Sawatdee and Teerapol Srichana
Pharmaceutics 2024, 16(10), 1334; https://doi.org/10.3390/pharmaceutics16101334 - 18 Oct 2024
Viewed by 1061
Abstract
Background/Objectives: Effective airway delivery of a fixed-dose combination of triple-aerosolized inhaled corticosteroid (ICS)/long-acting beta agonist (LABA)/long-acting muscarinic antagonist (LAMA) is likely to positively affect therapeutic responses predicted in patients with asthma and chronic obstructive pulmonary disease. This study aimed to conduct in vitro [...] Read more.
Background/Objectives: Effective airway delivery of a fixed-dose combination of triple-aerosolized inhaled corticosteroid (ICS)/long-acting beta agonist (LABA)/long-acting muscarinic antagonist (LAMA) is likely to positively affect therapeutic responses predicted in patients with asthma and chronic obstructive pulmonary disease. This study aimed to conduct in vitro fluticasone furoate, vilanterol trifenatate, and umeclidinium bromide depositions in a Next Generation Impactor. The aerodynamic properties of these inhaled medications influence the spatial distribution and drug abundance, particularly in the smaller airways, to reverse or alleviate disease pathology. Methods: The Next Generation Impactor was used to demonstrate the aerodynamic particle size distributions of fluticasone furoate, vilanterol trifenatate, and umeclidinium bromide delivered from a dry powder inhaler at different flow rates across all stages of the impactors. This in vitro study analyzed the distribution pattern of individual drug components to simulate mono-component deposition and co-deposition in the official model in the United States Pharmacopeia. An Andersen cascade impactor together with scanning electron microscope–energy-dispersive X-ray was employed to observe the drug deposition on each stage of the impactor. Results: We found that the distribution pattern of each component at the same cascade level was comparable, and the aerosol particles of the three drugs reached the in vitro representation of the lower airway compartment. The specified flow rates generated the desired fine particle fraction, fine particle dose, and mass median aerodynamic diameter. Our results also demonstrated visualized deposition patterns of the delivered drugs from different stages of the cascade impactor that may predict deposition as it occurs in vivo. Conclusions: Spatial distribution and abundance of ICS/LABA/LAMA in the same cascade levels were closely comparable, and the aerosol particles were able to reach the small aerosol-sized cascades at the lower levels to some extent. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) The schematic diagram represents the experimental design for drug deposition from a TRELEGE ELLIPTA™ DPI inhaler (fluticasone furoate, umeclidinium bromide, vilanterol trifenatate) in a Next Generation Impactor (NGI) at different air flow rates. (<b>B</b>) The experimental diagram of drug deposition in an Andersen cascade impactor (ACI) to determine the distribution patterns at each stage using scanning electron microscopy (SEM) and an energy-dispersive X-ray (EDX) analysis system. The drugs are presented by different colors in the bar chart and SEM images. The green color is fluticasone furoate, the red color is umeclidinium bromide, and the yellow/orange color is vilanterol trifenatate.</p>
Full article ">Figure 2
<p>Chromatogram representing the peaks and retention times of (<b>A</b>) standards vilanterol, umeclidinium bromide, salmeterol, and fluticasone furoate and (<b>B</b>) sample of vilanterol, umeclidinium bromide, and fluticasone furoate from a TRELEGE ELLIPTA™ DPI inhaler with the spike of the salmeterol internal standard.</p>
Full article ">Figure 3
<p>Aerodynamic particle size distributions of vilanterol trifenatate (green bar), umeclidinium bromide (red bar), and fluticasone furoate (yellow bar) from the TRELEGE ELLIPTA™ DPI inhaler operated with the Next Generation Impactor (NGI; USP Apparatus 6) at different air flow rates of (<b>A</b>) 30 L/min, (<b>B</b>) 60 L/min, and (<b>C</b>) 90 L/min. Significance by Tukey test was defined as <span class="html-italic">p</span>-value &lt; 0.05 (*) or <span class="html-italic">p</span>-value &lt; 0.01 (**).</p>
Full article ">Figure 4
<p>Aerodynamic particle size distributions: (<b>A</b>) vilanterol trifenatate; (<b>B</b>) umeclidinium bromide; and (<b>C</b>) fluticasone furoate. The TRELEGE ELLIPTA™ DPI inhaler was operated with the Next Generation Impactor (NGI; USP Apparatus 6) at different air flow rates of 30, 60, and 90 L/min. Significance by Tukey test was defined as <span class="html-italic">p</span>-value &lt; 0.05 (*) or <span class="html-italic">p</span>-value &lt; 0.01 (**).</p>
Full article ">Figure 5
<p>Aerodynamic particle size distributions of vilanterol trifenatate (green bar), umeclidinium bromide (red bar), and fluticasone furoate (yellow bar) from the TRELEGE ELLIPTA™ DPI inhaler obtained with the Andersen cascade impactor (ACI; USP Apparatus 1). (<b>A</b>) Scanning electron microscopy (SEM) images of the fixed-dose combination of ICS/LABA/LAMA particle co-deposition on each stage of the ACI under SEM and energy-dispersive X-ray (EDX) qualitative analyses. Red represents the Br atom of umeclidinium bromide, yellow represents the Cl atom of vilanterol trifenatate, and green represents the F atom of fluticasone furoate. (<b>B</b>) The bars indicate the proportions of drug deposition at each stage of the ACI by normalized EDX signal analysis.</p>
Full article ">
15 pages, 2967 KiB  
Article
Investigation of the Impact of Manufacturing Methods on Protein-Based Long-Acting Injectable Formulations: A Comparative Assessment for Microfluidics vs. Conventional Methods
by Nihan Yonet-Tanyeri, Robert S. Parker, Louis D. Falo and Steven R. Little
Pharmaceutics 2024, 16(10), 1264; https://doi.org/10.3390/pharmaceutics16101264 - 27 Sep 2024
Viewed by 853
Abstract
Background/Objectives: Microparticle-based drug delivery systems offer several advantages for protein-based drug formulations, enhancing patient compliance and therapeutic efficiency through the sustained delivery of the active pharmaceutical ingredient. Over the past few decades, the microfluidics method has emerged as a continuous manufacturing process for [...] Read more.
Background/Objectives: Microparticle-based drug delivery systems offer several advantages for protein-based drug formulations, enhancing patient compliance and therapeutic efficiency through the sustained delivery of the active pharmaceutical ingredient. Over the past few decades, the microfluidics method has emerged as a continuous manufacturing process for preparing drug-encapsulating microparticles, mainly for small molecule drugs. However, comparative assessments for the conventional batch method vs. the microfluidics method for protein-based drug formulations have been lacking. The main objective of this study was to generate immunomodulatory protein drug-loaded injectable formulations using both conventional batch and microfluidics methods. Methods: Therefore, rhCCL22-loaded poly(lactic-co-glycolic) acid (PLGA) microparticles were prepared by conventional homogenization and microfluidics methods. Results: The resulting microparticles were analyzed comparatively, focusing on critical quality attributes such as microparticle size, size distribution, morphology, drug encapsulation efficiency, release kinetics, and batch-to-batch variations in relation to the manufacturing method. Our results demonstrated that the conventional method resulted in microparticles with denser surface porosity and wider size distribution as opposed to microparticles prepared by the microfluidics method, which could contribute to a significant difference in the drug-release kinetics. Additionally, our findings indicated minimal variation within batches for the microparticles prepared by the microfluidics method. Conclusion: Overall, this study highlights the comparative assessment of several critical quality attributes and batch variations associated with the manufacturing methods of protein-loaded microparticles which is crucial for ensuring consistency in efficacy, regulatory compliance, and quality control in the drug formulation manufacturing process. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic illustration of the manufacturing methods for protein-loaded PLGA microparticles using microfluidics and conventional methods.</p>
Full article ">Figure 2
<p>rhCCL22-loaded PLGA microparticles exhibit different size distribution based on the manufacturing method. (<b>a</b>,<b>b</b>) Volume-weighted size distributions and (<b>c</b>,<b>d</b>) representative scanning electron microscopy images of rhCCL22-loaded PLGA microparticles that were prepared by (<b>a</b>,<b>c</b>) microfluidics or (<b>b</b>,<b>d</b>) conventional methods.</p>
Full article ">Figure 3
<p>Representative scanning electron microscopy images demonstrate the surface morphology and inner porosity differences for rhCCL22-loaded PLGA microparticles. PLGA microparticles were prepared by (<b>a</b>,<b>c</b>) microfluidics and (<b>b</b>,<b>d</b>) conventional methods. Arrows in (<b>d</b>) highlight some examples of the inner pore connections.</p>
Full article ">Figure 4
<p>rhCCL22 encapsulation (%) in the PLGA microparticles shows a significant difference depending on the manufacturing process: microfluidics vs. conventional methods. ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 5
<p>rhCCL22 release kinetics from PLGA microparticles demonstrates different release kinetics according to the manufacturing method. (<b>a</b>) Microparticles prepared by the microfluidics method showed a zero-order delayed (~2 days) release profile. (<b>b</b>) Microparticles prepared by the conventional method presented a zero-order release profile.</p>
Full article ">Figure 6
<p>Batch-to-batch variations in the size distribution of rhCCL22-loaded PLGA microparticles when PLGA microparticles were prepared by (<b>a</b>) microfluidics and (<b>b</b>) conventional methods.</p>
Full article ">Figure 7
<p>Batch-to-batch variations in terms of rhCCL22 encapsulation (%) in PLGA microparticles regarding the manufacturing method. rhCCL22-loaded PLGA microparticles were prepared by (<b>a</b>) microfluidics and (<b>b</b>) conventional methods. * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001, ns indicates non-significant difference.</p>
Full article ">Figure 8
<p>Batch-to-batch variations in terms of rhCCL22 release kinetics from PLGA microparticles. rhCCL22-loaded PLGA microparticles were prepared by (<b>a</b>) microfluidics and (<b>b</b>) conventional methods.</p>
Full article ">
11 pages, 4222 KiB  
Article
Design of pH/Redox Co-Triggered Degradable Diselenide-Containing Polyprodrug via a Facile One-Pot Two-Step Approach for Tumor-Specific Chemotherapy
by Yanru Hu and Peng Liu
Molecules 2024, 29(16), 3837; https://doi.org/10.3390/molecules29163837 - 13 Aug 2024
Viewed by 974
Abstract
The diselenide bond has attracted intense interest for drug delivery systems (DDSs) for tumor chemotherapy, owing to it possessing higher redox sensitivity than the disulfide one. Various redox-responsive diselenide-containing carriers have been developed for chemotherapeutics delivery. However, the premature drug leakage from these [...] Read more.
The diselenide bond has attracted intense interest for drug delivery systems (DDSs) for tumor chemotherapy, owing to it possessing higher redox sensitivity than the disulfide one. Various redox-responsive diselenide-containing carriers have been developed for chemotherapeutics delivery. However, the premature drug leakage from these DDSs was significant enough to cause toxic side effects on normal cells. Here, a pH/redox co-triggered degradable polyprodrug was designed as a drug self-delivery system (DSDS) by incorporating drug molecules as structural units in the polymer main chains, using a facile one-pot two-step approach. The proposed PDOX could only degrade and release drugs by breaking both the neighboring acid-labile acylhydrazone and the redox-cleavable diselenide conjugations in the drug’s structural units, triggered by the higher acidity and glutathione (GSH) or reactive oxygen species (ROS) levels in the tumor cells. Therefore, a slow solubility-controlled drug release was achieved for tumor-specific chemotherapy, indicating promising potential as a safe and efficient long-acting DSDS for future tumor treatment. Full article
(This article belongs to the Special Issue Exclusive Feature Papers on Molecular Structure)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p><sup>1</sup>H NMR spectrum of selenolactone.</p>
Full article ">Figure 2
<p><sup>1</sup>H NMR spectra of DOX, D-DOX<sub>ADH</sub> and PDOX.</p>
Full article ">Figure 3
<p>GPC curve of the proposed PDOX.</p>
Full article ">Figure 4
<p>Typical hydrodynamic diameter and distribution of the PDOX nanoparticles by dialysis at different concentrations (<b>a</b>) and the TEM image of the PDOX nanoparticles fabricated at 0.2 mg/mL (<b>b</b>).</p>
Full article ">Figure 5
<p>Drug release profiles of the PDOX nanoparticles in different media.</p>
Full article ">Figure 6
<p>CLSM images of the HepG2 cells after incubation with the PDOX nanoparticles (15 μg/mL) for 48 h: DOX (<b>a</b>), DAPI (<b>b</b>) and merged (<b>c</b>) (scale bar: 20 μm).</p>
Full article ">Figure 7
<p>Cell viability assay in L02 and HepG2 cells of PDOX nanoparticles (<b>a</b>) and DOX (<b>b</b>) with different concentrations for 48 h, respectively. Values are expressed as mean ± SD (<span class="html-italic">n</span> = 6) (* denotes significant difference <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Scheme 1
<p>Synthesis of the pH/redox co-triggered degradable polyprodrug (PDOX).</p>
Full article ">Scheme 2
<p>Synthesis of selenolactone.</p>
Full article ">Scheme 3
<p>pH/Reduction co-triggered degradation of PDOX to release the selenol (DOX-SeH).</p>
Full article ">Scheme 4
<p>pH/Oxidation co-triggered degradation of PDOX to release the seleninic acid (DOX-SeOOH).</p>
Full article ">
23 pages, 4341 KiB  
Article
Drug Combination Nanoparticles Containing Gemcitabine and Paclitaxel Enable Orthotopic 4T1 Breast Tumor Regression
by Jesse Yu, Xiaolin Xu, James Ian Griffin, Qingxin Mu and Rodney J. Y. Ho
Cancers 2024, 16(16), 2792; https://doi.org/10.3390/cancers16162792 - 8 Aug 2024
Viewed by 1556
Abstract
Early diagnosis, intervention, and therapeutic advancements have extended the lives of breast cancer patients; however, even with molecularly targeted therapies, many patients eventually progress to metastatic cancer. Recent data suggest that residual breast cancer cells often reside in the lymphatic system before rapidly [...] Read more.
Early diagnosis, intervention, and therapeutic advancements have extended the lives of breast cancer patients; however, even with molecularly targeted therapies, many patients eventually progress to metastatic cancer. Recent data suggest that residual breast cancer cells often reside in the lymphatic system before rapidly spreading through the bloodstream. To address this challenge, an effective drug combination composed of gemcitabine (G) and paclitaxel (T) is administered intravenously in sequence at the metastatic stage, but intravenous GT infusion may limit lymphatic GT drug accessibility and asynchronous drug exposure in cancer cells within the lymph. To determine whether co-localization of intracellular gemcitabine and paclitaxel (referred to as GT) could overcome these limitations and enhance the efficacy of GT, we have evaluated a previously reported GT drug-combination formulated in nanoparticle (referred to as GT-in-DcNP) evaluated in an orthotopic breast tumor model. Previously, with indocyanine green-labeled nanoparticles, we reported that GT-in-DcNP particles after subcutaneous dosing were taken up rapidly and preferentially into the lymph instead of blood vessels. The pharmacokinetic study showed enhanced co-localization of GT within the tumors and likely through lymphatic access, before drug apparency in the plasma leading to apparent long-acting plasma time-course. The mechanisms may be related to significantly greater inhibitions of tumor growth—by 100 to 140 times—in both sub-iliac and axillary regions compared to the equivalent dosing with free-and-soluble GT formulation. Furthermore, GT-in-DcNP exhibited dose-dependent effects with significant tumor regression. In contrast, even at the highest dose of free GT combination, only a modest tumor growth reduction was notable. Preliminary studies with MDA-231-HM human breast cancer in an orthotopic xenograft model indicated that GT-in-DcNP may be effective in suppressing human breast tumor growth. Taken together, the synchronized delivery of GT-in-DcNP to mammary tumors through the lymphatic system offers enhanced cellular retention and greater efficacy. Full article
(This article belongs to the Special Issue Drug Delivery for Cancer Therapy)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Characterization of a primary multi-site 4T1-based model of breast cancer. Mice were inoculated with 1 million 4T1-luc cells at four different sites (Panel (<b>A</b>)). Tumors located near the axillary fat pads exhibited statistically distinct growth patterns compared to tumors located near the sub-iliac fat pads (Panel (<b>B</b>)) as determined by luminescent measurements over time (mean ± SEM). ** <span class="html-italic">p</span> &lt; 0.001, Student’s <span class="html-italic">t</span>-test. On the right of Panel (<b>B</b>) is a representative bioluminescence image of a mouse with 4T1 tumors on day 7.</p>
Full article ">Figure 2
<p>Characterization of blood and lymphatic vasculature development in growing primary tumors. After 5 days of growth, 4T1 tumors harvested from the axillary and sub-iliac fat pads displayed vascularization with both blood and lymphatic vessels. CD31 was used as a marker for endothelial cells on blood vessels and is highlighted by black arrows. Podoplanin was used as a marker for the lymphatic vascular system and is highlighted by red arrows. Representative IHC slides from the tumor harvested from fad-pads of mice from axillary and sub-iliac sites are presented in Panel (<b>A</b>). ImageJ analysis was performed as a semi-quantitative measure of positive staining for CD31 (black circle) and podoplanin (black square) in the axillary (Panel (<b>B</b>)) and sub-iliac tumor (Panel (<b>C</b>)). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001, Student’s <span class="html-italic">t</span>-test. Data presented in Panels (<b>B</b>,<b>C</b>) are based on the mean ± SD of eight randomly selected 4T1 tumor areas from each site.</p>
Full article ">Figure 2 Cont.
<p>Characterization of blood and lymphatic vasculature development in growing primary tumors. After 5 days of growth, 4T1 tumors harvested from the axillary and sub-iliac fat pads displayed vascularization with both blood and lymphatic vessels. CD31 was used as a marker for endothelial cells on blood vessels and is highlighted by black arrows. Podoplanin was used as a marker for the lymphatic vascular system and is highlighted by red arrows. Representative IHC slides from the tumor harvested from fad-pads of mice from axillary and sub-iliac sites are presented in Panel (<b>A</b>). ImageJ analysis was performed as a semi-quantitative measure of positive staining for CD31 (black circle) and podoplanin (black square) in the axillary (Panel (<b>B</b>)) and sub-iliac tumor (Panel (<b>C</b>)). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001, Student’s <span class="html-italic">t</span>-test. Data presented in Panels (<b>B</b>,<b>C</b>) are based on the mean ± SD of eight randomly selected 4T1 tumor areas from each site.</p>
Full article ">Figure 3
<p>Effect of DcNP on tumor inhibition by gemcitabine and paclitaxel when compared to the same dose of free drugs. Mice were treated with either saline (black circle), GT in DMSO (20/2 mg/kg, black square), or GT in DcNP (20/2 mg/kg, black triangle) 24 h after cancer cell inoculation. Mice were monitored for 7 days, and tumor growth was evaluated by luminescence. The integrated luminescence photon counts in the axillary and sub-iliac tumors are presented in panels (<b>A</b>) and (<b>B</b>), respectively. The mean and standard deviations of luminescence (<span class="html-italic">n</span> = 4–6) are presented above. ns: non-significance, * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001, Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>Dose proportional effects of gemcitabine and paclitaxel in DcNP or free form on orthotopic 4T1 tumor inhibition. Mice with 4T1 tumors in fat-pads were treated with GT in free form at 20:2 mg/kg (∆) or GT-in-DcNP at 5:0.5 mg/kg (◊), 10:1 mg/kg (□) or 20:2 mg/kg (○) 24 h after cancer cell inoculation. Mice were monitored for 168 h (7 days) after cell inoculation, and before termination, and tumor growth was evaluated by luminescence. Panel (<b>A</b>) and (<b>B</b>) represent the luminescence signal integrations from axillary tumors and sub-iliac tumors, respectively. The mean and standard deviations of luminescence are presented (<span class="html-italic">n</span> = 8–10). DcNP vs. Free, * <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 5
<p>Effect of DcNP on the transit and accumulation of gemcitabine and paclitaxel in lymphatic vessels and tumors following subcutaneous injection. Panel (<b>A</b>): ICG infrared fluorescence-labeled GT-in-DcNP were subcutaneously injected at the lower abdomen area of tumor-bearing mice (red arrow). At 1 h, the ICG-fluorescence signal from GT-in-DcNP is detectable at the injection site (Inj) and throughout the lymphatic vessels (white arrows), but not in the blood vessel (no detectable fluorescence)—in the thoracoepigastric vein (black arrows), which is visible as a dark vessel located parallel to the fluorescent lymphatic vessels. The fluorescence signal can also be observed in the axillary node (LN) with small traces on the surface of the tumor (T). Panel (<b>B</b>): The corresponding GT drug concentrations in the tumors and plasma are presented for the 1-h and 3-h time points for mice dosed with DcNP or soluble GT at an equivalent dose. The data presented were mean <span class="underline">+</span> SD. *** indicates <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 6
<p>Time-course of GT-in-DcNP to sustain tumor gemcitabine levels as determined by tumor-to-plasma gemcitabine ratio. GT-in-DcNP administered subcutaneously resulted in consistent tumor-to-plasma concentration ratios of gemcitabine near or above 1 for 48-h in both sub-iliac and axillary 4T1 tumors implanted in fat-pads.</p>
Full article ">Figure 7
<p>Marked reduction of neoplastic 4T1 cells in tumor sites, with retention of normal mammary tissue elements following treatment with GT-in-DcNP and not free GT. The remaining tumor masses from mice inoculated with 4T1 cells were surgically resected 6 days after treatment with saline (<b>A</b>,<b>E</b>), free drugs (20:2 mg/kg) (<b>B</b>,<b>F</b>), or GT-in-DcNP (20:2 mg/kg) (<b>C</b>,<b>G</b>). The histology of healthy mammary fat-pads from non-cancer mice is presented in panel (<b>D</b>,<b>H</b>). The resected axillary site tumors or normal mammary tissue are presented in the top (<b>A</b>–<b>D</b>) panel, and sub-iliac site tumors or normal mammary tissue are presented in the bottom (<b>E</b>–<b>H</b>) panel. Tissues were fixed in 10% neutral-buffered formalin.</p>
Full article ">Figure 8
<p>Effect on GT-in-DcNP treatment on human breast cancer MDA-231-HM orthotopic mouse model. Athymic mouse inoculated with 2 million luciferase-transfected MDA-231-HM human breast cancer cells. These mice were then treated with a placebo (control) or GT-in-DcNP (test treatment product) by subcutaneous injection on days 1, 7, 14, and 21. The G:T dose given was 20:2 mg/kg on day 1 and 10:1 mg/kg on days 7, 14, and 21. The effects of GT-in-DcNP or placebo treatment were presented as two-panels. These data present the bioluminescence intensity starting on day 4 to monitor MDA-231-HM tumor growth at the axillary (Panel (<b>A</b>)) and the sub-iliac (Panel (<b>B</b>)) locations. DcNP treatment effects on tumor weights were compared to placebo treatments for MDA-231-HM tumors isolated from the axillary (Panel (<b>C</b>)) and sub-iliac (Panel (<b>D</b>)) sites on day 28. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 based on Student’s <span class="html-italic">t</span>-test. Panel (<b>E</b>) presents bioluminescence images of MDA-231-HM tumors from saline control and GT-in-DcNP groups on day 28.</p>
Full article ">Scheme 1
<p>Chemical structures, full names, molecular weights (MW), and CAS registry numbers of drugs: gemcitabine (<b>A</b>), paclitaxel (<b>B</b>), and lipid excipients: DSPC (<b>C</b>) and DSPE-mPEG<sub>2000</sub> (<b>D</b>) of the GT-in-DcNP formulation.</p>
Full article ">
23 pages, 4946 KiB  
Article
Next-Generation Contraceptive Intravaginal Ring: Comparison of Etonogestrel and Ethinyl Estradiol In Vitro and In Vivo Release from 3D-Printed Intravaginal Ring and NuvaRing
by Isabella C. Young, Allison L. Thorson, Mackenzie L. Cottrell, Craig Sykes, Amanda P. Schauer, Rani S. Sellers, Rima Janusziewicz, Kathleen L. Vincent and Soumya Rahima Benhabbour
Pharmaceutics 2024, 16(8), 1030; https://doi.org/10.3390/pharmaceutics16081030 - 2 Aug 2024
Viewed by 1562
Abstract
Intravaginal rings (IVRs) represent a well-established, woman-controlled and sustained vaginal drug delivery system suitable for a wide range of applications. Here, we sought to investigate the differences in etonogestrel (ENG) and ethinyl estradiol (EE) release from a 3D-printed IVR utilizing continuous liquid interface [...] Read more.
Intravaginal rings (IVRs) represent a well-established, woman-controlled and sustained vaginal drug delivery system suitable for a wide range of applications. Here, we sought to investigate the differences in etonogestrel (ENG) and ethinyl estradiol (EE) release from a 3D-printed IVR utilizing continuous liquid interface production (CLIP™) (referred to as CLIPLOW for low drug loading and CLIPHIGH IVRs for high drug loading) and NuvaRing, a commercially available injection molded IVR. We conducted in vitro release studies in simulated vaginal fluid to compare the release of ENG and EE from CLIPLOW IVRs and NuvaRing. CLIPLOW IVRs had a similar hormone dose to NuvaRing and exhibited slightly slower ENG release and greater EE release in vitro compared to NuvaRing. When administered to female sheep, NuvaRing demonstrated greater ENG/EE levels in plasma, vaginal tissue and vaginal fluids compared to CLIPLOW IVR despite similar drug loadings. Leveraging observed hormones levels in sheep from NuvaRing as an effective contraceptive benchmark, we developed a long-acting CLIPHIGH IVR with increased ENG and EE doses that demonstrated systemic and local hormone levels greater than the NuvaRing for 90 days in sheep. No signs of toxicity were noted regarding general health, colposcopy, or histological analysis in sheep after CLIPHIGH IVR administration. Our results provided (1) a comparison of ENG and EE release between a 3D-printed IVR and NuvaRing in vitro and in vivo, (2) a preclinical pharmacokinetic benchmark for vaginally delivered ENG and EE and (3) the generation of a 90-day CLIP IVR that will be utilized in future work to support the development of a long-acting ENG/EE IVR combined with an antiretroviral for the prevention of HIV and unplanned pregnancy. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Illustration of IVR manufacturing and loading equations for ENG and EE. (<b>A</b>) Schematic of IVR manufacturing with traditional hot-melt extrusion and injection molding. Drugs (ENG and EE) and copolymer (EVA) are poured into the hopper of the injection molding machine. Material is heated and uniformly mixed via hot-melt extrusion and injected into the final IVR mold. Once cooled, the machine ejects the final ENG/EE EVA IVR (NuvaRing). Figure made with BioRender.com. (<b>B</b>) IVR manufacturing with CLIP 3D printing (CLIP IVR). Projection of UV light onto photoactive resin (SIL30) promotes polymerization and solidification of the UV-cured IVR. Incorporation of oxygen via an oxygen-permeable window generates a region of uncured resin, known as the ‘dead zone’, to prevent part-attachment to the window. After the UV cure, the IVR undergoes a thermal cure to complete the fabrication process to produce the final product. ENG/EE was incorporated into the IVR via absorption as the IVR swells upon immersion in a drug-containing solution, promoting drug uptake. (<b>C</b>) Image of CLIP IVR (<b>left</b>) and NuvaRing (<b>right</b>). (<b>D</b>) Mass and dimensions of CLIP IVR (<span class="html-italic">n</span> = 3) and NuvaRing (<span class="html-italic">n</span> = 1). (<b>E</b>,<b>F</b>) Loading equations of ENG and EE in CLIP IVR, respectively, as previously developed and validated [<a href="#B12-pharmaceutics-16-01030" class="html-bibr">12</a>].</p>
Full article ">Figure 2
<p>In vitro release of ENG/EE from CLIP<sub>LOW</sub> IVR and NuvaRing. (<b>A</b>) Cumulative μg in vitro release of ENG/EE from CLIP<sub>LOW</sub> IVR and NuvaRing. (<b>B</b>) Cumulative μg in vitro release of ENG/EE from CLIP<sub>LOW</sub> IVR and NuvaRing after 90 days of storage under accelerated stability conditions (40 °C/75% relative humidity). (<b>C</b>,<b>D</b>) Summary of release kinetics of in vitro release of ENG/EE from CLIP<sub>LOW</sub> IVR and NuvaRing at baseline and after 90 days of storage under accelerated stability conditions, respectively. BLOD represents ‘below limit of detection’ from HPLC analysis. CLIP<sub>LOW</sub> IVR release studies were performed in triplicate (<span class="html-italic">n</span> = 3) and NuvaRing in vitro release studies were performed with <span class="html-italic">n</span> = 1. All release studies were performed in SVF + 2% Solutol at pH 4 release media at 37 °C. The maximum standard deviation for in vitro cumulative release was less than 2%.</p>
Full article ">Figure 3
<p>Pharmacokinetics of CLIP<sub>LOW</sub> IVR and NuvaRing in sheep. ENG/EE levels in sheep (average ± standard deviation) from CLIP<sub>LOW</sub> IVR (<span class="html-italic">n</span> = 4) and NuvaRing (<span class="html-italic">n</span> = 4) in (<b>A</b>) plasma, (<b>B</b>) vaginal tissue and (<b>C</b>) vaginal fluids. LLOQ of ENG in plasma, vaginal tissue and vaginal fluid is 0.2 ng/mL, 17 ng/g and 0.215 ng/swab, respectively. LLOQ of EE in plasma, vaginal tissue and vaginal fluid is 0.005 ng/mL, 4.2 ng/g and 0.108 ng/swab, respectively. Samples that were below the limit of quantification were represented as LLOQ/2. (<b>D</b>,<b>E</b>) Summary table of residual drug quantification and estimated in vivo release rates after CLIP<sub>LOW</sub> IVR and NuvaRing removal, respectively. Individual replicates are shown in <a href="#app1-pharmaceutics-16-01030" class="html-app">Figure S1</a>.</p>
Full article ">Figure 4
<p>In vitro release of ENG/EE from CLIP<sub>HIGH</sub> IVR compared to CLIP<sub>LOW</sub> IVR. (<b>A</b>) Cumulative (%) in vitro release of ENG/EE from CLIP<sub>HIGH</sub> IVR compared to CLIP<sub>LOW</sub> IVR. (<b>B</b>) Cumulative (µg) in vitro release of ENG/EE from CLIP<sub>HIGH</sub> IVR compared to CLIP<sub>LOW</sub> IVR. (<b>C</b>) Summary table of in vitro release profiles. (<b>D</b>) Summary table of in vitro release rates. All studies were carried out in triplicate in SVF pH 7 (sheep vaginal pH) release media. Each release curve represents the average ± standard deviation. Maximum standard deviation for in vitro cumulative release is less than 2%.</p>
Full article ">Figure 5
<p>Sheep pharmacokinetics of ENG/EE after 92 days of CLIP<sub>HIGH</sub> IVR administration. (<b>A</b>) Study design of a 92-day PK study with CLIP<sub>HIGH</sub> IVR in female sheep. ENG/EE concentrations in sheep (<span class="html-italic">n</span> = 3 sheep that underwent the entire 92-day study) in (<b>B</b>) plasma, (<b>C</b>) vaginal tissue and (<b>D</b>) vaginal fluids from CLIP<sub>LOW</sub> IVR, CLIP<sub>HIGH</sub> IVR and NuvaRing. Each curve represents the average ± standard deviation. Dashed lines indicated the last concentration (day 21) of ENG (dashed green line) and (dashed blue line) from the NuvaRing sheep study extended to 92 days for comparative purposes only. Individual replicates for sheep that underwent the 92-day study duration with CLIP<sub>HIGH</sub> IVR are shown in <a href="#app1-pharmaceutics-16-01030" class="html-app">Figure S2</a>. Individual replicates for CLIP<sub>LOW</sub> IVR and NuvaRing are shown in <a href="#app1-pharmaceutics-16-01030" class="html-app">Figure S1</a>.</p>
Full article ">Figure 6
<p>Assessment of PK and PK tail after CLIP<sub>HIGH</sub> IVR removal. (<b>A</b>–<b>C</b>) CLIP<sub>HIGH</sub> IVR removal at 30 days post-IVR administration in (<b>A</b>) plasma, (<b>B</b>) vaginal fluid and (<b>C</b>) vaginal tissue. (<b>D</b>–<b>F</b>) CLIP<sub>HIGH</sub> IVR removal at 60 days post-IVR administration in (<b>D</b>) plasma, (<b>E</b>) vaginal fluid and (<b>F</b>) vaginal tissue. (<b>G</b>–<b>I</b>) CLIP<sub>HIGH</sub> IVR removal at 92 days post-IVR administration in (<b>G</b>) plasma, (<b>H</b>) vaginal tissue and (<b>I</b>) vaginal fluid. Drug concentration curves are plotted as average ± standard deviation. Data from all sheep (<span class="html-italic">n</span> = 6) were pooled together for timepoints when IVRs were inserted. At day 30 post-IVR administration, <span class="html-italic">n</span> = 2 IVRs were removed from sheep (<b>A</b>–<b>C</b>) and at days 60 and 92 post-IVR administration, <span class="html-italic">n</span> = 3 IVRs were removed (<b>D</b>–<b>I</b>). Solid lines indicate when IVRs were inserted, and dashed lines indicate post-IVR removal. Black arrows indicate the time at which IVR was removed. LLOQs of ENG in plasma, vaginal tissue and vaginal fluid were 0.1 ng/mL, 5.11 ng/g and 0.215 ng/swab, respectively. LLOQs of EE in plasma, vaginal tissue and vaginal fluid were 0.005 ng/mL, 2.55 ng/g and 0.054 ng/swab, respectively. Samples that were below the limit of quantification were represented as LLOQ/2. Individual replicates are shown in <a href="#app1-pharmaceutics-16-01030" class="html-app">Figure S3</a>.</p>
Full article ">Figure 7
<p>Quantification of residual drug and estimation of in vivo release rates. (<b>A</b>) Residual drug ENG/EE after CLIP<sub>HIGH</sub> IVR removal at days 30, 60 and 92 post-administration. (<b>B</b>) % ENG/EE released in vivo after IVR removal. (<b>C</b>) Estimation of ENG and EE in vivo release rates based on residual drug content after IVR removal. Data represent average ± standard deviation for <span class="html-italic">n</span> = 2 sheep for day 30 and <span class="html-italic">n</span> = 3 sheep for days 60 and 92.</p>
Full article ">Figure 8
<p>Sheep safety analysis for NuvaRing. Histological images of vaginal biopsies from sheep administered NuvaRing (with higher magnification insert) at (<b>A</b>) day 0, (<b>B</b>) day 1, (<b>C</b>) day 3, (<b>D</b>) day 7, (<b>E</b>) day 14 and (<b>F</b>) day 21 post-IVR insertion. Red arrowheads denote examples of eosinophils. Black arrows denote parakeratosis. Scale bar = 50 µm. (<b>G</b>) Histological scoring summary based on H&amp;E images. Individual scores from each sheep are presented in <a href="#app1-pharmaceutics-16-01030" class="html-app">Figure S4</a>. Day 0 represents the baseline with no IVR use.</p>
Full article ">Figure 9
<p>Sheep safety analysis for CLIP<sub>HIGH</sub> IVR. Histological images of vaginal biopsies from sheep administered CLIP<sub>HIGH</sub> IVR (with higher magnification insert) at (<b>A</b>) day 0, (<b>B</b>) day 1, (<b>C</b>) day 3, (<b>D</b>) day 7, (<b>E</b>) day 14, (<b>F</b>) day 21, (<b>G</b>) day 30, (<b>H</b>) day 60, (<b>I</b>) day 77 and (<b>J</b>) day 92, and (<b>K</b>) day 3 post-IVR removal and (<b>L</b>) day 7 post-IVR removal. Red arrowheads denote examples of eosinophils. Black arrows denote parakeratosis. Scale bar = 50 µm. (<b>M</b>) Histological scoring summary based on H&amp;E images for 92 days. Individual scores from each sheep are presented in <a href="#app1-pharmaceutics-16-01030" class="html-app">Figures S5–S7</a>. Day 0 represents the baseline with no IVR use.</p>
Full article ">
9 pages, 247 KiB  
Brief Report
Neutrophil–Lymphocyte Ratio Values in Schizophrenia: A Comparison between Oral and Long-Acting Antipsychotic Therapies
by Antonino Messina, Fabrizio Bella, Giuliana Maccarone, Alessandro Rodolico and Maria Salvina Signorelli
Brain Sci. 2024, 14(6), 602; https://doi.org/10.3390/brainsci14060602 - 14 Jun 2024
Cited by 1 | Viewed by 1211
Abstract
Background: Schizophrenia is a mental disorder affecting approximately 0.32% of the global population, according to the World Health Organization. Antipsychotic medications are used to treat this condition by inhibiting D2 dopamine and 5HT2 serotonin receptors. The selection of the appropriate mode of delivery [...] Read more.
Background: Schizophrenia is a mental disorder affecting approximately 0.32% of the global population, according to the World Health Organization. Antipsychotic medications are used to treat this condition by inhibiting D2 dopamine and 5HT2 serotonin receptors. The selection of the appropriate mode of delivery for these drugs is based on factors such as patient adherence, clinical presentation, and patient preferences. However, additional drivers of treatment selection are required in clinical practice. Mounting evidence suggests that neuroinflammation plays a crucial role in the pathogenesis of schizophrenia. NLR, a cost-effective biomarker of inflammation, has increased in several psychiatric conditions and may represent a valid method for studying the inflammatory stage in schizophrenia, relapse, and the first episode of psychosis. The aim of this study is to evaluate whether there are any variations in NLR values between patients given oral antipsychotics and those given long-acting antipsychotics. Methods: The study included 50 individuals with schizophrenia, either acute or in the follow-up phase. NLR was obtained by calculating the ratio of absolute neutrophil count (cells/μL) and absolute lymphocyte count (cells/μL). Results: Patients on long-acting antipsychotics exhibited significantly lower mean NLR scores (1.5 ± 0.7) compared to those on oral antipsychotics (2.2 ± 1.3) (p < 0.05). Conclusions: NLR appears promising as a neuroinflammatory biomarker. This study reveals significantly lower NLR values in patients on long-acting antipsychotics, which may signify reduced systemic inflammation and improved adherence. Full article
(This article belongs to the Section Neuropsychiatry)
22 pages, 5297 KiB  
Article
Development and Evaluation of a Water-Free In Situ Depot Gel Formulation for Long-Acting and Stable Delivery of Peptide Drug ACTY116
by Yingxin Xiong, Zhirui Liu, Yuanqiang Wang, Jiawei Wang, Xing Zhou and Xiaohui Li
Pharmaceutics 2024, 16(5), 620; https://doi.org/10.3390/pharmaceutics16050620 - 5 May 2024
Viewed by 1927
Abstract
In situ depot gel is a type of polymeric long-acting injectable (pLAI) drug delivery system; compared to microsphere technology, its preparation process is simpler and more conducive to industrialization. To ensure the chemical stability of peptide ACTY116, we avoided the use of harsh [...] Read more.
In situ depot gel is a type of polymeric long-acting injectable (pLAI) drug delivery system; compared to microsphere technology, its preparation process is simpler and more conducive to industrialization. To ensure the chemical stability of peptide ACTY116, we avoided the use of harsh conditions such as high temperatures, high shear mixing, or homogenization; maintaining a water-free and oxygen-free environment was also critical to prevent hydrolysis and oxidation. Molecular dynamics (MDs) simulations were employed to assess the stability mechanism between ACTY116 and the pLAI system. The initial structure of ACTY116 with an alpha helix conformation was constructed using SYBYL-X, and the copolymer PLGA was generated by AMBER 16; results showed that PLGA-based in situ depot gel improved conformational stability of ACTY116 through hydrogen bonds formed between peptide ACTY116 and the components of the pLAI formulation, while PLGA (Poly(DL-lactide-co-glycolide)) also created steric hindrance and shielding effects to prevent conformational changes. As a result, the chemical and conformational stability and in vivo long-acting characteristics of ACTY116 ensure its enhanced efficacy. In summary, we successfully achieved our objective of developing a highly stable peptide-loaded long-acting injectable (LAI) in situ depot gel formulation that is stable for at least 3 months under harsh conditions (40 °C, above body temperature), elucidating the underlying stabilisation mechanism, and the high stability of the ACTY116 pLAI formulation creates favourable conditions for its in vivo pharmacological activity lasting for weeks or even months. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Structure of peptide ACTY116; (<b>b</b>) schematic illustration of in situ depot gel preparation and in vivo long-acting performance.</p>
Full article ">Figure 2
<p>Schematic illustration of double emulsion–solvent extraction/evaporation technique for microsphere preparation.</p>
Full article ">Figure 3
<p>CH modelling and drug administration.</p>
Full article ">Figure 4
<p>The polarizing microscope observation of ACTY116 microspheres: (<b>a</b>) 20×, (<b>b</b>) 80×; (<b>c</b>) particle size distribution.</p>
Full article ">Figure 5
<p>The CD spectra of different ACTY116 dosage forms: (<b>a</b>) ACTY116 solution (F1); (<b>b</b>) ACTY116 microspheres (F2); (<b>c</b>) ACTY116 pLAI in situ depot gel (F3).</p>
Full article ">Figure 6
<p>The results of variable evaluation: (<b>a</b>) evaluation of impact of NMP quantity on ACTY116 stability in pLAIs (F4–F7); (<b>b</b>) evaluation of impact of PLGA quantity on ACTY116 stability in pLAIs (F8–F13); (<b>c</b>) evaluation of impact of surface area of liquid–air interface (three sizes of vials with diameters of 4 mm, 10 mm, and 19 mm) on ACTY116 stability in pLAIs; (<b>d</b>) evaluation of impact of residual oxygen in the headspace on ACTY116 stability in pLAIs. Data are represented as mean ± SD (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 7
<p>Polarised microscopic observation of ACTY116 in situ depot gel: (<b>a</b>) month 0; (<b>b</b>) long-term stability: month 6; 25 °C/60% RH; (<b>c</b>) accelerated stability: month 3; 40 °C/75% RH.</p>
Full article ">Figure 8
<p>(<b>a</b>) Ensemble of ACTY116 initial state solvated in water; (<b>b</b>) MDs simulations under 373.15 K for 100 ns in water; (<b>c</b>) root mean square fluctuation (RMSF) value of ACTY116 solvated into water under 373.15 K; (<b>d</b>) ensemble of ACTY116 initial state complex of ACTY116 and PLGA solvated into NMP; (<b>e</b>) MDs simulations under 373.15 K for 100 ns in PLGA and NMP system; (<b>f</b>) root mean square fluctuation (RMSF) value of ACTY116 solvated into PLGA and NMP system under 373.15 K; (<b>g</b>) root mean square fluctuation (RMSF) value of ACTY116 solvated into water under 373.15 K for 100 ns; (<b>h</b>) RMSF value of ACTY116 solvated into PLGA and NMP system under 373.15 K for 100 ns; (<b>i</b>) visualisation of ACTY116 unit complexed with PLGA (gray part) around 10 Å distance.</p>
Full article ">Figure 9
<p>Evaluation of pharmacological activity on ACTY116 solution and pLAI formulations: (<b>a</b>) heart sizes; (<b>b</b>) heart weight to body weight (HW/BW) ratios; (<b>c</b>) heart weight to tibial length (HW/TL) ratios; (<b>d</b>) serum levels of NT-proBNP; (<b>e</b>) quantitative analysis of β-MHC expression; (<b>f</b>) quantitative analysis of myocyte cross-sectional areas in heart tissue sections with HE staining; (<b>g</b>) representative images of β-MHC (upper) and BNP (lower) expression (positive expression is brownish yellow); (<b>h</b>) representative images of H&amp;E staining of heart tissues; (<b>i</b>) representative images of WGA staining of heart tissues. <span class="html-italic">n</span> = 6 in each group, * <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 9 Cont.
<p>Evaluation of pharmacological activity on ACTY116 solution and pLAI formulations: (<b>a</b>) heart sizes; (<b>b</b>) heart weight to body weight (HW/BW) ratios; (<b>c</b>) heart weight to tibial length (HW/TL) ratios; (<b>d</b>) serum levels of NT-proBNP; (<b>e</b>) quantitative analysis of β-MHC expression; (<b>f</b>) quantitative analysis of myocyte cross-sectional areas in heart tissue sections with HE staining; (<b>g</b>) representative images of β-MHC (upper) and BNP (lower) expression (positive expression is brownish yellow); (<b>h</b>) representative images of H&amp;E staining of heart tissues; (<b>i</b>) representative images of WGA staining of heart tissues. <span class="html-italic">n</span> = 6 in each group, * <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 10
<p>Effect of PLGA copolymer on in vitro drug release from pLAI in situ depot gels: (<b>a</b>) impact of PLGA quantity on ACTY116 in vitro release; (<b>b</b>) impact of type of PLGA on ACTY116 in vitro release.</p>
Full article ">Figure 11
<p>Evaluation of different doses on ACTY116 the in vivo PK profile.</p>
Full article ">
25 pages, 3267 KiB  
Review
Long-Acting Gel Formulations: Advancing Drug Delivery across Diverse Therapeutic Areas
by Hossein Omidian and Renae L. Wilson
Pharmaceuticals 2024, 17(4), 493; https://doi.org/10.3390/ph17040493 - 12 Apr 2024
Cited by 6 | Viewed by 5423
Abstract
This multifaceted landscape of long-acting gels in diverse medical fields, aims to enhance therapeutic outcomes through localized treatment and controlled drug release. The objective involves advancements spanning cancer treatment, immunotherapy, diabetes management, neuroendocrine disorders, ophthalmic applications, contraception, HIV/AIDS treatment, chronic diseases, wound care, [...] Read more.
This multifaceted landscape of long-acting gels in diverse medical fields, aims to enhance therapeutic outcomes through localized treatment and controlled drug release. The objective involves advancements spanning cancer treatment, immunotherapy, diabetes management, neuroendocrine disorders, ophthalmic applications, contraception, HIV/AIDS treatment, chronic diseases, wound care, and antimicrobial treatments. It explores the potential of long-acting gels to offer sustained and extended drug release, targeted therapy, and innovative administration routes while addressing limitations such as scalability challenges and regulatory hurdles. Future directions focus on personalized therapies, biodegradability, combination therapies, interdisciplinary innovation, regulatory considerations, and patient-centric development. This comprehensive review highlights the pivotal role of long-acting gels in transforming therapeutic approaches and improving patient outcomes across various medical conditions. Full article
Show Figures

Figure 1

Figure 1
<p>Shows results after a one-dose administration of Tam-Gel and its ability as a long-acting agent to inhibit intrahepatic subcutaneous tumor growth of MCF-7 cells in nude mice (<b>A</b>) Photographs of intrahepatic lesions caused by MCF-7 in immunodeficient rats. (<b>B</b>) Quantitative results of the lesions’ area by total liver area (mean ± SD). (<b>C</b>) Inhibition rates calculated from the quantitative results. * <span class="html-italic">p</span> &lt; 0.05 [<a href="#B1-pharmaceuticals-17-00493" class="html-bibr">1</a>].</p>
Full article ">Figure 2
<p>Injectable thermosensitive PEG–polyester hydrogel formulation of liraglutide as a long-acting treatment for type 2 diabetes mellitus. D0, D2, D4, D7 and D10 represent the long-acting hypoglycemic effect [<a href="#B11-pharmaceuticals-17-00493" class="html-bibr">11</a>].</p>
Full article ">Figure 3
<p>A schematic presentation of the drug encapsulation and delayed drug release from optimized zinc-EXT nanoparticles encapsulated into the physical polymeric hydrogel [<a href="#B12-pharmaceuticals-17-00493" class="html-bibr">12</a>].</p>
Full article ">Figure 4
<p>A schematic representation of the preparation and in vivo nerve blockade effect of the gel-–microsphere system with bupivacaine (Gel-MS/BUP) [<a href="#B40-pharmaceuticals-17-00493" class="html-bibr">40</a>].</p>
Full article ">Figure 5
<p>Peptide hydrogelators as an enzyme-triggered, long-acting injectable delivery platform for HIV/AIDS. (<b>a</b>) Chemical structure of L-<span class="html-italic">α</span> peptide hydrogelator NapFFKY(p)G-OH and covalently conjugated antiretroviral drug (R1). (<b>b</b>) Peptide gelators, upon exposure to an enzyme, lead to the formation of a 3D network of entangled fibers that entrap water and form a hydrogel. (<b>c</b>) Demonstrates the potential application of a soluble peptide–antiretroviral conjugate administered monthly via subcutaneous injection [<a href="#B48-pharmaceuticals-17-00493" class="html-bibr">48</a>].</p>
Full article ">Figure 6
<p>(<b>A</b>) Schematic representation of HF-MAP-mediated delivery from a reservoir. Digital light microscopy images with X12.5 magnification of (<b>B</b>) MAP1 after skin deposition experiment. (<b>C</b>) Skin surface where MAP1 was applied during skin deposition experiment, (<b>D</b>) MAP2 after skin deposition experiment (X8 magnification), and (<b>E</b>) skin surface where MAP2 was applied during skin deposition experiment [<a href="#B50-pharmaceuticals-17-00493" class="html-bibr">50</a>].</p>
Full article ">Scheme 1
<p>Long-acting gels in medicine necessity and impact across diverse applications.</p>
Full article ">
12 pages, 3435 KiB  
Article
Diselenide-Bridged Doxorubicin Dimeric Prodrug: Synthesis and Redox-Triggered Drug Release
by Yanru Hu and Peng Liu
Molecules 2024, 29(8), 1709; https://doi.org/10.3390/molecules29081709 - 10 Apr 2024
Cited by 2 | Viewed by 1264
Abstract
The diselenide bond has attracted intense interest in redox-responsive drug delivery systems (DDSs) in tumor chemotherapy, due to its higher sensitivity than the most investigated bond, namely the disulfide bond. Here, a diselenide-bridged doxorubicin dimeric prodrug (D-DOXSeSe) was designed by coupling [...] Read more.
The diselenide bond has attracted intense interest in redox-responsive drug delivery systems (DDSs) in tumor chemotherapy, due to its higher sensitivity than the most investigated bond, namely the disulfide bond. Here, a diselenide-bridged doxorubicin dimeric prodrug (D-DOXSeSe) was designed by coupling two doxorubicin molecules with a diselenodiacetic acid (DSeDAA) molecule via α-amidation, as a redox-triggered drug self-delivery system (DSDS) for tumor-specific chemotherapy. The drug release profiles indicated that the D-DOXSeSe could be cleaved to release the derivatives selenol (DOX-SeH) and seleninic acid (DOX-SeOOH) with the triggering of high GSH and H2O2, respectively, indicating the double-edged sword effect of the lower electronegativity of the selenide atom. The resultant solubility-controlled slow drug release performance makes it a promising candidate as a long-acting DSDS in future tumor chemotherapy. Moreover, the interaction between the conjugations in the design of self-immolation traceless linkers was also proposed for the first time as another key factor for a desired precise tumor-specific chemotherapy, besides the conjugations themselves. Full article
(This article belongs to the Section Applied Chemistry)
Show Figures

Figure 1

Figure 1
<p><sup>1</sup>H NMR spectrum of DSeDAA.</p>
Full article ">Figure 2
<p><sup>1</sup>H NMR spectrum of D-DOX<sub>SeSe</sub>.</p>
Full article ">Figure 3
<p>Hydrodynamic diameters of D-DOX<sub>SeSe</sub> nanoparticles fabricated at different concentrations (<b>a</b>) and TEM image of D-DOX<sub>SeSe</sub> nanoparticles fabricated at 1.0 mg/mL (<b>b</b>).</p>
Full article ">Figure 4
<p>Redox-triggered drug release from D-DOX<sub>SeSe</sub> nanoparticles in different media.</p>
Full article ">Figure 5
<p>HPLC results of the D-DOX<sub>SeSe</sub> nanoparticles after treatment with GSH and H<sub>2</sub>O<sub>2</sub>.</p>
Full article ">Figure 6
<p>CLSM images of HepG2 cells after incubation with the D-DOX<sub>SeSe</sub> nanoparticles (15 μg/mL) for 24 h: (<b>a</b>) nuclei stained with DAPI, (<b>b</b>) DOX, and (<b>c</b>) the merged image.</p>
Full article ">Figure 7
<p>Cell viability assay in L02 (<b>a</b>) and HepG2 (<b>b</b>) cells of D-DOX<sub>SeSe</sub> nanoparticles and free DOX with different concentrations for 48 h, respectively. Values are expressed as mean ± SD (n = 6); * denotes significant difference <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Scheme 1
<p>Synthesis of the diselenide-bridged doxorubicin dimeric prodrug (D-DOX<sub>SeSe</sub>).</p>
Full article ">Scheme 2
<p>Reduction and oxidation-triggered degradation and drug release.</p>
Full article ">
17 pages, 4284 KiB  
Article
Nanoparticle-Based Secretory Granules Induce a Specific and Long-Lasting Immune Response through Prolonged Antigen Release
by Laia Bosch-Camós, Carlos Martínez-Torró, Hèctor López-Laguna, Jara Lascorz, Jordi Argilaguet, Antonio Villaverde, Fernando Rodríguez and Esther Vázquez
Nanomaterials 2024, 14(5), 435; https://doi.org/10.3390/nano14050435 - 27 Feb 2024
Cited by 1 | Viewed by 1711
Abstract
Developing prolonged antigen delivery systems that mimic long-term exposure to pathogens appears as a promising but still poorly explored approach to reach durable immunities. In this study, we have used a simple technology by which His-tagged proteins can be assembled, assisted by divalent [...] Read more.
Developing prolonged antigen delivery systems that mimic long-term exposure to pathogens appears as a promising but still poorly explored approach to reach durable immunities. In this study, we have used a simple technology by which His-tagged proteins can be assembled, assisted by divalent cations, as supramolecular complexes with progressive complexity, namely protein-only nanoparticles and microparticles. Microparticles produced out of nanoparticles are biomimetics of secretory granules from the mammalian hormonal system. Upon subcutaneous administration, they slowly disintegrate, acting as an endocrine-like secretory system and rendering the building block nanoparticles progressively bioavailable. The performance of such materials, previously validated for drug delivery in oncology, has been tested here regarding the potential for time-prolonged antigen release. This has been completed by taking, as a building block, a nanostructured version of p30, a main structural immunogen from the African swine fever virus (ASFV). By challenging the system in both mice and pigs, we have observed unusually potent pro-inflammatory activity in porcine macrophages, and long-lasting humoral and cellular responses in vivo, which might overcome the need for an adjuvant. The robustness of both innate and adaptive responses tag, for the first time, these dynamic depot materials as a novel and valuable instrument with transversal applicability in immune stimulation and vaccinology. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Figure 1

Figure 1
<p>Construction and characterization of RK4-P30-H6 and GFP-H6. (<b>A</b>) Architectonic principles governing the POM principle, namely microparticle generation out of nanoparticles and further nanoparticle release. His-tagged proteins tend to self-assemble, upon recombinant production and Ni<sup>2+</sup>-based purification, into oligomeric nanoparticles, assisted by divalent cations. A cationic amino acid N-terminal stretch favors this process. The addition of a molar excess of cationic Zn produces the immediate formation of microscale particles. Upon in vivo administration and upon Zn dilution, these materials release stable nanoparticles differently. (<b>B</b>) Schematic representation of RK4-P30-H6 and GFP-H6 constructs. In RK4-P30-H6, a flexible peptide linker (GGSSRSS) was incorporated. (<b>C</b>) RK4-P30-H6 characterization by H6 immunodetection in Western blot with anti-His monoclonal antibody ((<b>C</b>), left). Size of the purified protein determined by DLS. The protein size was also measured under chelating conditions (2 mM EDTA + 1% SDS) ((<b>C</b>), right). (<b>D</b>) Immunodetection of GFP-H6 by Western Blot ((<b>D</b>), left) and size of the construct analyzed by DLS ((<b>D</b>), right).</p>
Full article ">Figure 2
<p>Formation and characterization of p30 POMs and GFP POMs. Representative micrographs of p30 POMs (<b>A</b>) and GFP POMs (<b>B</b>) obtained by SEM (scale bar represents 1 µm). (<b>C</b>) Size of p30 POMs and of the soluble protein released in vitro after seven days as determined by DLS (left). The relative amount of soluble protein released from p30 POMs for seven days is also shown (right). (<b>D</b>) Size of GFP POMs and of the soluble protein released from these microparticles as determined by DLS at day seven (left). The relative amount of soluble protein released from GFP POMs for seven days is also depicted (right).</p>
Full article ">Figure 3
<p>Subcutaneous inoculation of p30 secretory granules in pigs induces ASFV-specific antibodies. ASFV-specific IgGs in sera from pigs inoculated with 50 µg of p30 Soluble (<b>A</b>) without or (<b>B</b>) with CAF01, or with 50 µg of p30 POMs (<b>C</b>) without or (<b>D</b>) with CAF01, and (<b>E</b>) the control group. (<b>F</b>) ASFV-specific IgGs in sera from pigs receiving 150 µg of p30 POMs or PBS as control from the second experiment. Arrows indicate the two vaccination days. (<b>G</b>) ASFV-specific IgG titers in pig sera two weeks after the second administration (SD35). (<b>H</b>) Statistical analyses of ASFV-specific IgG titers in pig sera two weeks after the second administration (SD35). Statistical significance was determined by one-way ANOVA followed by Tukey’s multiple comparisons test and is displayed in GraphPad style (ns <span class="html-italic">p</span> &gt; 0.05, * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, **** <span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 4
<p>Subcutaneous inoculation of p30 POMs in pigs induces an IgG2 bias and detectable IgA in serum. (<b>A</b>) ASFV-specific IgG1 and IgG2 in sera (1/100 dilution) from pigs receiving p30 or p30 POMs two weeks after the second inoculation (SD35) assessed by ELISA. (<b>B</b>) ASFV-specific IgA in sera (1/100 dilution) from pigs receiving p30 POMs two weeks after the second inoculation (SD35) determined by ELISA. Statistical significance was determined by one-way ANOVA followed by Tukey’s multiple comparisons test and is displayed in GraphPad style (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 5
<p>In vitro stimulation with POMs induces non-specific cell activation. (<b>A</b>) IFNγ-producing PBMCs from three naive pigs measured by ELISpot assay. Cells were stimulated in vitro with 5 µg/mL of p30 or p30 POMs. (<b>B</b>) Cytokine levels in culture supernatants of PAMs stimulated in vitro for 24 h with 5 µg/mL of GFP POMs or left unstimulated (RPMI) quantified by Luminex-based multiplex assay. Statistical significance was determined by unpaired two-tailed t-test for normally distributed data, or two-tailed Mann–Whitney U test for not normally distributed data and is displayed in GraphPad style (<span class="html-italic">p</span> &gt; 0.05 ns, * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 6
<p>Subcutaneous inoculation of microparticulated GFP induces long-lasting dose-dependent production of GFP-specific antibodies and CD8 T cells in mice. (<b>A</b>,<b>B</b>) GFP-specific antibody levels in sera from mice receiving GFP POMs assessed by ELISA two (SD35, (<b>A</b>)) and nine (SD85, (<b>B</b>)) weeks after the second inoculation. (<b>C</b>) GFP-specific IgG1 and IgG2a in sera from mice receiving GFP POMs nine weeks after the second inoculation. (<b>D</b>) GFP-specific IgA in sera from mice receiving GFP POMs nine weeks after the second inoculation. (<b>E</b>) Percentage of proliferating (Ki67+) CD4<sup>+</sup> and CD8<sup>+</sup> T cells in splenocytes after in vitro stimulation for five days with 5 µg/mL of GFP POMs. Percentages obtained from untreated cells were subtracted. Statistical significance was assessed by one-way ANOVA followed by Tukey’s multiple comparisons test and is displayed in GraphPad style (* <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">
13 pages, 1474 KiB  
Article
Pharmacokinetic Modeling to Guide Preclinical Development of an Islatravir-Eluting Reservoir-Style Biodegradable Implant for Long-Acting HIV PrEP
by Talisa S. Kinsale, Mackenzie L. Cottrell, Linying Li, Rhonda Brand, Greg Gatto, Ellen Luecke, Chasity Norton, Archana Krovi, Julie B. Dumond, Gauri Rao, Shekhar Yeshwante, Brian Van Horne, Ariane Van Der Straten, Angela D. M. Kashuba and Leah M. Johnson
Pharmaceutics 2024, 16(2), 201; https://doi.org/10.3390/pharmaceutics16020201 - 30 Jan 2024
Viewed by 1920
Abstract
Long-acting injectable cabotegravir is more effective than daily oral PrEP at preventing HIV transmission due to improved adherence, but requires bi-monthly large-volume intramuscular injections. Subcutaneous (SC) contraceptive implants can be formulated with antiretrovirals for extended-duration HIV PrEP. Islatravir (ISL) is a first-in-class, investigational [...] Read more.
Long-acting injectable cabotegravir is more effective than daily oral PrEP at preventing HIV transmission due to improved adherence, but requires bi-monthly large-volume intramuscular injections. Subcutaneous (SC) contraceptive implants can be formulated with antiretrovirals for extended-duration HIV PrEP. Islatravir (ISL) is a first-in-class, investigational antiretroviral with pharmacologic properties well-suited for implant delivery. We performed preclinical studies for the development of a reservoir-style, poly(ε-caprolactone) ISL-eluting implant by conducting a single-dose SC ISL dose-ranging pharmacokinetic (PK) study of 0.1, 0.3, and 1 mg/kg in adult Wistar rats. Non-compartmental analysis was conducted, and dose proportionality assessed for ISL plasma and intracellular islatravir-triphosphate (ISL-tp). Population PK models estimated ISL’s unit impulse response to deconvolve ISL-implant in vivo absorption rate (mg/day) and cumulative mass (mg) from published rat plasma PK (n = 10). Drug release was interpreted using four kinetic models. Dose proportionality was affirmed for ISL and ISL-tp. A first-order, two-compartment model fitted the SC ISL bolus data. Mean (SD) absorption rate from 0 to 154 days was 0.072 ± 0.024 mg/day, and cumulative mass at 154 days was 8.67 ± 3.22 mg. ISL absorption was well-described by zero-order (r2 = 0.95) and Ritger–Peppas (r2 = 0.98). Our zero-order ISL-release poly(ε-caprolactone) implant is projected to achieve clinical PK above ISL-tp’s PrEP efficacy threshold. Continued development for HIV PrEP applications is warranted. Full article
Show Figures

Figure 1

Figure 1
<p>Semi-log concentration–time plot of (<b>a</b>) Islatravir (ISL) plasma concentration (ng/mL) and (<b>b</b>) ISL-triphosphate (ISL-tp) concentration (pmol/million cells) following a single SC administration at 0.1, 0.3, or 1.0 mg/kg. Plasma and PBMCs was collected from 3 rats per time point, with 24 rats per dosing level. Markers are individual plasma concentration with median (line) categorized by dose (color). The grey reference line (dash) is clinical threshold of 0.05 pmol/million cells.</p>
Full article ">Figure 2
<p>Two-compartment population pharmacokinetic (PopPK) model with multiplicative error was fit to ISL plasma concentrations observed in rats following single SC administration at 0.1, 0.3 and 1 mg/kg doses. Macro-parameter constants (A, B) and exponential rates (α, β) are described in the inset table.</p>
Full article ">Figure 3
<p>Two-compartment population PK diagnostics showing observed (open circles) and predicted (black lines) for (<b>a</b>) individual predicted concentrations vs. residuals with loess fit (blue) and absolute trend lines (red), (<b>b</b>) individual predicted vs. observed log-concentrations, and (<b>c</b>) quantile–quantile normal plot.</p>
Full article ">Figure 4
<p>(<b>a</b>) Absorption rate, and (<b>b</b>) cumulative mass absorbed of subcutaneous ISL-eluting implants in rats over 154 days. Red represents animals receiving ISL only, while blue received ISL alongside a hormone implant (ISL plus). Lines and markers (shape) represent each individual animal sample collected at approximately 15-day increments. Three implants were removed after 100 days in the ISL only group.</p>
Full article ">
42 pages, 6282 KiB  
Review
Polymer Delivery Systems for Long-Acting Antiretroviral Drugs
by Mohammad Ullah Nayan, Sudipta Panja, Ashrafi Sultana, Lubaba A. Zaman, Lalitkumar K. Vora, Brady Sillman, Howard E. Gendelman and Benson Edagwa
Pharmaceutics 2024, 16(2), 183; https://doi.org/10.3390/pharmaceutics16020183 - 28 Jan 2024
Cited by 4 | Viewed by 2761
Abstract
The success of long-acting (LA) drug delivery systems (DDSs) is linked to their biocompatible polymers. These are used for extended therapeutic release. For treatment or prevention of human immune deficiency virus type one (HIV-1) infection, LA DDSs hold promise for improved regimen adherence [...] Read more.
The success of long-acting (LA) drug delivery systems (DDSs) is linked to their biocompatible polymers. These are used for extended therapeutic release. For treatment or prevention of human immune deficiency virus type one (HIV-1) infection, LA DDSs hold promise for improved regimen adherence and reduced toxicities. Current examples include Cabenuva, Apretude, and Sunlenca. Each is safe and effective. Alternative promising DDSs include implants, prodrugs, vaginal rings, and microarray patches. Each can further meet patients’ needs. We posit that the physicochemical properties of the formulation chemical design can optimize drug release profiles. We posit that the strategic design of LA DDS polymers will further improve controlled drug release to simplify dosing schedules and improve regimen adherence. Full article
Show Figures

Figure 1

Figure 1
<p>Timeline for LA drug delivery formulations for the treatment and prevention of HIV-1.</p>
Full article ">Figure 2
<p>Chemical structures of the polymers used in LA delivery systems.</p>
Full article ">Figure 3
<p>Different types of LA DDSs that are in either clinical or preclinical development. (<b>A</b>) Different types of solid implants. Reproduced with permission from Ref [<a href="#B165-pharmaceutics-16-00183" class="html-bibr">165</a>]. Copyright 2018 Antimicrobial Agents and Chemotherapy. (<b>B</b>) Different types of Vaginal rings—(<b>i</b>) over-molded metal spring design, (<b>ii</b>) matrix-type VR, (<b>iii</b>) Full-length reservoir-type VR, (<b>iv</b>) sandwich-shaped VR, (<b>v</b>) Partial reservoir-type VR, (<b>vi</b>) Insertable reservoir-type VR. Images were adapted under the terms of Creative Commons Attribution—Non-commercial from ref. [<a href="#B166-pharmaceutics-16-00183" class="html-bibr">166</a>]. (<b>C</b>) Nanocrystal formulations, polymeric nanoformulations, liposomes, and hydrogels. (<b>D</b>) Different types of microarray patches. Reproduced with permission [<a href="#B21-pharmaceutics-16-00183" class="html-bibr">21</a>].</p>
Full article ">Figure 4
<p>Different types of non-erodible implants are illustrated. (<b>A</b>–<b>C</b>) TAF implants contain silicone and PVA. Reproduced with permission from [<a href="#B15-pharmaceutics-16-00183" class="html-bibr">15</a>]. (<b>D</b>) PCL reservoir-style implant for TAF delivery, which comprises a formulated drug core (Reproduced under the Creative Commons CC-BY license from [<a href="#B12-pharmaceutics-16-00183" class="html-bibr">12</a>]). (<b>E</b>,<b>F</b>) Cross-sectional depiction of nanochannel-containing implant showing drug refill needles through the loading ports with resealable silicon plugs. TAF and FTC implants of two different sizes and shapes. Reprinted with permission [<a href="#B15-pharmaceutics-16-00183" class="html-bibr">15</a>].</p>
Full article ">Figure 5
<p>Microstructure in PLGA-based ISFIs: (<b>A</b>) Cumulative release of CAB ISFI PLGA formulations. (<b>B</b>) Effect of drug loading on CAB release. (<b>C</b>) PLGA molecular weight affects CAB release. To compare the drug release from different formulation at different timepoints in (<b>B</b>,<b>C</b>), two-way ANOVA with tukey’s multiple comparison test were performed. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, and ns (not significant) when p &gt; 0.05. (<b>D</b>–<b>L</b>) Effect of PLGA degradation on implant microstructure. SEM cross-section images of placebo ISFIs (1:2 <span class="html-italic">w</span>/<span class="html-italic">w</span> PLGA/NMP, PLGA MW 27 kDa) over a 30-day period. (<b>D</b>–<b>F</b>) Low-magnification image (100×) of the entire implant (scale bar = 100 µm). (<b>G</b>–<b>I</b>) Higher magnification (200×) of the implant shell (shell thickness was measured using SEM scale; scale bar = 50 µm). (<b>J</b>–<b>L</b>) Higher (500×) magnification of the center of the implant (scale bar = 20 µm). (<b>D</b>,<b>G</b>,<b>J</b>) Implants imaged at day 3 postincubation in 0.01 M PBS pH 7.4 at 37 °C. (<b>E</b>,<b>H</b>,<b>K</b>) Implants imaged at day 14 postincubation. (<b>F</b>,<b>I</b>,<b>L</b>) Implants imaged at day 30 postincubation. Symbol (*) represents implant shell. Symbols (^) represents the central pore of the implant. Images were reproduced under the terms of the Creative Commons CC-BY license [<a href="#B12-pharmaceutics-16-00183" class="html-bibr">12</a>,<a href="#B180-pharmaceutics-16-00183" class="html-bibr">180</a>].</p>
Full article ">Figure 6
<p>Fast-dissolving MNs have been made for ARV delivery. (<b>A</b>) Schematic depiction of programmed dissolving bilayer MAP with a high drug loading of LA microdepots for potential monthly human application. (<b>B</b>) Schematic presentation of the MN preparation process. The figure was reproduced under the terms of the Creative Commons CC-BY license [<a href="#B22-pharmaceutics-16-00183" class="html-bibr">22</a>].</p>
Full article ">Figure 7
<p>The illustration shows the in vivo fate of LASER ART prodrug nanoformulation. (<b>A</b>) Chemical synthesis of prodrug from parent ARV. (<b>B</b>) IM injection of LASER ART nanoformulation illustrates the muscle as the primary drug depot. (<b>C</b>,<b>D</b>) Macrophages phagocytose and store the nanocrystal from the site of injection. (<b>E</b>) Biodistribution of prodrug and active drug in tissues including HIV-1 reservoir site. (<b>F</b>) Slow prodrug release in the low-pH microenvironment in macrophages and then hydrolysis to release active drug. (<b>G</b>,<b>H</b>) Slow dissolution and hydrolysis of prodrug in the blood and tissues. The figure was reproduced under the terms of the Creative Commons CC-BY license [<a href="#B269-pharmaceutics-16-00183" class="html-bibr">269</a>].</p>
Full article ">
23 pages, 4095 KiB  
Article
A Bilayer Microarray Patch (MAP) for HIV Pre-Exposure Prophylaxis: The Role of MAP Designs and Formulation Composition in Enhancing Long-Acting Drug Delivery
by Lalitkumar K. Vora, Ismaiel A. Tekko, Fabiana Volpe Zanutto, Akmal Sabri, Robert K. M. Choy, Jessica Mistilis, Priscilla Kwarteng, Courtney Jarrahian, Helen O. McCarthy and Ryan F. Donnelly
Pharmaceutics 2024, 16(1), 142; https://doi.org/10.3390/pharmaceutics16010142 - 20 Jan 2024
Cited by 10 | Viewed by 2125
Abstract
Microarray patches (MAPs) have shown great potential for efficient and patient-friendly drug delivery through the skin; however, improving their delivery efficiency for long-acting drug release remains a significant challenge. This research provides an overview of novel strategies aimed at enhancing the efficiency of [...] Read more.
Microarray patches (MAPs) have shown great potential for efficient and patient-friendly drug delivery through the skin; however, improving their delivery efficiency for long-acting drug release remains a significant challenge. This research provides an overview of novel strategies aimed at enhancing the efficiency of MAP delivery of micronized cabotegravir sodium (CAB Na) for HIV pre-exposure prophylaxis (PrEP). The refinement of microneedle design parameters, including needle length, shape, density, and arrangement, and the formulation properties, such as solubility, viscosity, polymer molecular weight, and stability, are crucial for improving penetration and release profiles. Additionally, a bilayer MAP optimization step was conducted by diluting the CAB Na polymeric mixture to localize the drug into the tips of the needles to enable rapid drug deposition into the skin following MAP application. Six MAP designs were analyzed and investigated with regard to delivery efficiency into the skin in ex vivo and in vivo studies. The improved MAP design and formulations were found to be robust and had more than 30% in vivo delivery efficiency, with plasma levels several-fold above the therapeutic concentration over a month. Repeated weekly dosing demonstrated the robustness of MAPs in delivering a consistent and sustained dose of CAB. In summary, CAB Na MAPs were able to deliver therapeutically relevant levels of drug. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Schematic illustrating the fabrication and manufacture of MAP molds via two-photon polymerization 3D printing. (<b>b</b>) MAP array designs developed for two-photon polymerization 3D printing (except D5).</p>
Full article ">Figure 2
<p>Schematic representation of the bilayer MAP preparation method.</p>
Full article ">Figure 3
<p>Schematic diagram illustrating the experimental procedures used to evaluate CAB Na deposition ex vivo.</p>
Full article ">Figure 4
<p>(<b>a</b>) Drug loading per patch (µg) for MAPs of different designs and geometries. Data are shown as the mean ± SD (n ≥ 3). (<b>b</b>) Amount and (<b>c</b>) delivery efficiency of CAB Na into the ex vivo porcine skin following MAP application. Data are shown as the mean ± SD (n ≥ 4). (<b>d</b>) Amount and (<b>e</b>) delivery efficiency of CAB Na into female Sprague Dawley rats. Data are shown as the mean ± SD (n ≥ 8).</p>
Full article ">Figure 5
<p>(<b>a</b>) Representative light microscope images of MAPs manufactured using diluted CAB Na-hydrogel formulations F1 through F5, with MAP design D1 showing the localization of the drug into the tip (scale bar = 500 µm). Red arrow shows the drug loaded part in the MAP tips (<b>b</b>) Drug loading per patch (µg) for MAP-D1 with different hydrogel compositions. Data are shown as the mean ± SD (n ≥ 3). (<b>c</b>) Mechanical strength of MAP-D1 prepared by using formulations F1 through F5 (reported as %MAP shaft height reduction after applying a compression force of 32 N against aluminum block following a standardized test using a texture analyzer. Data are reported as mean ± SD (n = 3).</p>
Full article ">Figure 6
<p>(<b>a</b>) Representative light microscope images for the three MAP designs prepared by using formulation F3. (<b>b</b>) Comparison of drug content between F1 and F3 for different MAP designs. Data are shown as the mean ± SD (n ≥ 3).</p>
Full article ">Figure 7
<p>(<b>a</b>) Amount and (<b>b</b>) delivery efficiency of CAB Na into the ex vivo skin following MAP application. Data are shown as the mean ± SD (n = 5). (<b>c</b>) Amount and (<b>d</b>) delivery efficiency of CAB Na into female Sprague Dawley rats. Data are shown as the mean ± SD (n = 8).</p>
Full article ">Figure 8
<p>(<b>a</b>) Schematic of the pharmacokinetic study design consisting of three different experimental cohorts: (i) rats administered an IM injection at a dose of 2.5 mg/rat; (ii) rats administered MAP-D1-F3 at a dose of 4.34 mg/rat; and (iii) rats administered MAP-D5-F3 at a dose of 4.56 mg/rat. (<b>b</b>) Plasma profile of CAB following different experimental strategies. (<b>c</b>) Pharmacokinetic parameters for the different experimental groups. Data are shown as the mean ± SD (n ≥ 6).</p>
Full article ">Figure 9
<p>(<b>a</b>) Schematic of the pharmacokinetic study design for repeated dosing consisting of three different experimental cohorts: (i) rats administered an IM injection at a loading dose of 2.5 mg/rat, followed by a maintenance dose of 1.25 mg/rat via IM injection once a week; (ii) rats administered a loading dose of 2.5 mg/rat via IM injection, followed by a maintenance dose using a MAP at a dose of 4.34 mg/rat once a week; (iii) rats administered a MAP at a loading dose and a maintenance dose of 4.34 mg/rat once a week. (<b>b</b>) Plasma profile of CAB following different administration strategies. (<b>c</b>) Pharmacokinetic parameters for the different experimental groups. Data are shown as the mean ± SD (n ≥ 5). C<sub>max-1</sub> and C<sub>max-2</sub>: first and second maximum plasma concentration, respectively. T<sub>max-1</sub> and T<sub>max-2</sub>: first and second minimum time for C<sub>max</sub>, respectively.</p>
Full article ">
Back to TopTop