1. Introduction
Melanoma is the most lethal form of skin cancer, and the global occurrence of these cancers continues to increase. In the United States, approximately 100,640 new melanoma cases occurred in 2024, resulting in 8290 deaths. The 5-year relative survival rate of melanoma ranges significantly depending on the stage progression. In the early stage, when the cancer is localized, the survival rate exceeds 99%. However, the rate drops markedly to 35% following metastasis to a distant area [
1].
Recent studies have highlighted that resistance to immune checkpoint inhibitors remains a substantial clinical hurdle in melanoma treatment [
2]. Similarly, while targeted therapies have improved patient outcomes, the development of resistance mechanisms continues to impede their long-term efficacy. These findings underscore the necessity for ongoing research to overcome resistance and enhance the effectiveness of current melanoma therapies [
3]. Against these backdrops, research into PTX-based formulations could offer promising opportunities for melanoma treatment. Notably, PTX, a widely used chemotherapeutic agent, has demonstrated efficacy across various cancer types, including melanoma [
4,
5]. Indeed, a phase III randomized controlled trial showed that nanoparticle albumin-bound paclitaxel (nab-PTX) significantly improved progression-free survival and disease control rates in chemotherapy-naive patients with metastatic melanoma compared to dacarbazine [
6]. Additionally, research has indicated that ultralow, non-toxic doses of PTX can elicit a notable antitumor effect in vivo, as evidenced by prolonged survival in melanoma-bearing mice and a reduced tumor burden [
7].
PTX administration is fundamentally limited by its extremely poor aqueous solubility (0.3 μg/mL), which necessitates the use of cremophor EL and ethanol for intravenous (IV) injection. However, cremophor EL is associated with a significant risk of severe hypersensitivity reactions, including anaphylaxis, dyspnea, hypotension, angioedema, and generalized urticaria [
8]. Therefore, to mitigate these risks, patients must receive premedication with corticosteroids and antihistamines before each infusion [
9]. Subcutaneous (SC) administration of PTX represents a transformative approach to overcoming the critical limitations of IV PTX injections [
10]. By leveraging polymer-based prodrug formulations, such as polyacrylamide conjugates, SC delivery achieves a 70% reduction in peak plasma concentration (C
max) compared to IV, while maintaining therapeutic efficacy and mitigating dose-limiting toxicities, including neutropenia [
11]. That is, SC administration of the PTX formulation sustained tumor drug levels for >72 h post-injection, ensuring prolonged therapeutic exposure [
11]. Moreover, SC formulations eliminate cremophor EL, thereby reducing systemic neurotoxicity and hypersensitivity reactions [
12]. This administration route also simplifies treatment logistics, enabling potential self-administration and reducing hospital visits, which could significantly improve long-term patient compliance. Collectively, these pharmacokinetic and safety advantages position SC administration of PTX as a promising candidate for redefining the paradigms of outpatient cancer therapy.
Meanwhile, studies have indicated that PTX can inadvertently activate nuclear factor-kappa B (NF-κB), leading to the upregulation of matrix metalloproteinase-9 (MMP-9), which is associated with increased tumor invasiveness and metastasis. Specifically, PTX-induced NF-κB activation promotes the elevated expression of MMP-9, which contributes to the degradation of the extracellular matrix and facilitates cancer cell migration. This paradoxical effect imposes a significant limitation on PTX monotherapy, as it may promote cancer progression in certain contexts. Therefore, combining PTX with agents that inhibit NF-κB activation could enhance its therapeutic efficacy by mitigating these adverse effects [
13].
Curcumin (CUR) is also a promising adjunctive agent to PTX, as it enhances the therapeutic efficacy of PTX and addresses some of the limitations associated with chemotherapy, including drug resistance and toxicity [
14,
15]. Furthermore, the ability of CUR to inhibit MMP-9 and upregulate the tissue inhibitor of metalloproteinases-2 (TIMP-2) significantly suppresses tumor angiogenesis, metastasis, and cell proliferation, thereby complementing the antitumor mechanisms of PTX [
16,
17]. In breast cancer cells (MCF-7), the combination of CUR and PTX has been shown to synergistically enhance growth inhibition and induce significant apoptosis [
17]. Furthermore, CUR acts as a chemosensitizer by inhibiting the nuclear factor kappa B (NF-κB) pathway-mediated multidrug-resistance, thereby improving the cytotoxic activity of PTX, a substrate for P-glycoprotein (P-gp) [
18]. Studies also demonstrated that CUR-loaded lipid nanoparticles inhibited P-gp-mediated drug efflux in multidrug-resistant cancer cells, restoring the susceptibility of cancer cells to PTX. Additionally, the incorporation of PTX and CUR into novel drug delivery systems, such as cationic PEGylated niosomes and solid lipid nanoparticles, enhances their bioavailability, cellular uptake, and sustained release, resulting in improved anticancer efficacy while minimizing toxicity to normal cells [
18,
19]. These findings underscore the potential of CUR-PTX combination therapy as an effective strategy for overcoming drug resistance and enhancing therapeutic outcomes in cancer treatment.
Tocopherol polyethylene glycol succinate (TPGS), a water-soluble derivative of vitamin E, serves as a solubilizing agent and has been extensively studied for its role in enhancing the delivery and efficacy of hydrophobic drugs such as PTX when incorporated into mesoporous silica nanoparticles [
20]. Additionally, TPGS has been shown to inhibit P-gp, a key player in multidrug resistance, thereby increasing the intracellular concentration of P-gp substrates, including various chemotherapeutic drugs [
21]. The amphiphilic nature of TPGS also facilitates enhanced cellular uptake of nanoparticles, promoting improved interactions with cancer cell membranes [
22]. Furthermore, TPGS stabilizes CUR and promotes its sustained release, enhancing its synergistic anticancer properties alongside PTX, such as apoptosis induction and NF-κB inhibition [
17,
23]. While specific studies on TPGS in B16F10 melanoma models are limited, the general benefits in drug delivery and potential to enhance therapeutic efficacy make TPGS a promising component in PTX-based treatments for melanoma [
24].
Therefore, this study aimed to develop a nano-based delivery system comprising PTX-loaded mesoporous silica nanoparticles for SC administration. Mesoporous silica nanoparticles have been approved by the United States Food and Drug Administration (U.S. FDA) for clinical trials of cancer formulations due to their adjustable porous structure, ability to induce surface modification, high loading efficiency, excellent biocompatibility, and biodegradability [
25]. This study demonstrated that the insertion of PTX into mesoporous silica nanoparticles with co-administration of CUR and TPGS could efficiently deliver PTX to B16F10 by diminishing fatal problems associated with PTX in vivo and in vitro, yielding PSC (i.e., PS combined with CUR) and PSCT (i.e., PS combined with CUR and TPGS) formulations, respectively. These formulations are designed to enhance therapeutic efficacy by overcoming the inherent drawbacks associated with PTX through synergistic chemosensitization and modulation of P-gp activity.
2. Materials and Methods
2.1. Materials
Mesoporous silica nanoparticles, named Smart Mesoporous Ball 7 (SMB7; Patent no. KR-10-2240246) and PTX encapsulated SMB7 (PS), were provided by Dr. Sang-Cheol Han (CEN Co., Ltd.; Miryang-si, Republic of Korea). SMB7 contains SiO
2 and ZnO (atomic composition: Si 30.4%, O 68.0%, and Zn 1.6%), while the average particle size of SMB7 was 200 nm, with pore diameter and volume of 2.54 nm and 0.81 cm
3/g, respectively. PS was prepared by the solvent evaporation method. Briefly, 2 g of PTX was dissolved in a methanol: acetone = 1:1 (
v/
v) mixture in a round-bottom flask, then 8 g of SMB7 was added to the PTX solution and stirred for 1 h; the solvent was evaporated using a vacuum evaporator [
26]. PS was purified through a multi-step washing process to remove unincorporated or surface-adsorbed PTX. Specifically, PS was dispersed in 100 mL of ethanol for 2 h, followed by centrifugation at 10,000×
g for 10 min, and this process was repeated three times. Subsequently, the PS particles were dispersed in 100 mL of distilled water for 4 h and centrifuged at 10,000×
g for 10 min, and this process was repeated twice to remove residual solvent and free drug molecules. The final nanoparticle product was recovered by centrifugation and dried under low vacuum conditions to obtain a dry powder suitable for biological experiments.
B16F10 cells were obtained from the Korean Cell Line Bank (Seoul, Republic of Korea). CUR, TPGS, berberine chloride (internal standard; IS), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), dimethyl sulfoxide (DMSO), verapamil, PTX, bovine serum albumin (BSA), and formic acid were purchased from Sigma–Aldrich (St. Louis, MO, USA). Dulbecco’s Modified Eagle Medium (DMEM), phosphate-buffered solution (PBS), fetal bovine serum (FBS), and penicillin–streptomycin were purchased from Corning Life Sciences (Oneonta, NY, USA). Meanwhile, 4′,6-diamidino-2-phenylindole (DAPI) was purchased from Chem Cruz (Dallas, TX, USA). Goat anti-mouse IgG conjugated with FSD 594 was purchased from BioActs (Incheon, Republic of Korea). All other reagents were of cell culture grade or analytical grade.
2.2. Physicochemical Characteristics of PS
2.2.1. Solubility
PTX loading in the mesoporous nanosilicate was measured as follows: 5 mg of the powder was placed in 50 mL of methanol and completely dissolved using a thermo micro-mixer (Finepcr; Gunpo-si, Republic of Korea) at 37 °C for 2 h. After 1 min of centrifugation, the supernatant was filtered through a 0.45 μm membrane filter. The filtrate was diluted 100-fold with the mobile phase and injected into the LC–MS/MS system to measure the PTX concentration. PTX loading (%) was calculated as the percentage of PTX amount in the total weight of the PS formulation.
To evaluate the solubility of the formulation, PTX and PS powder (2 mg as PTX) were weighed, and then, added in 2 mL of simulated body fluid (SBF; pH 7.4) consisting of 135 mM sodium chloride, 4.2 mM sodium bicarbonate, 4 mM potassium chloride, 1 mM potassium phosphate dibasic trihydrate, 1.5 mM magnesium chloride, 2.5 mM calcium chloride, 0.5 mM sodium sulfate 0.071 g, tris(hydroxymethyl)aminomethane 50 mM, and 40 mM hydrochloride. The mixtures were shaken using a thermo-micro mixer at 37 °C for 2 h. After 1 min of centrifugation, the supernatant was filtered through a 0.45 μm membrane filter. The filtrate was diluted 100-fold with the mobile phase and injected into the LC–MS/MS to measure the PTX concentration.
2.2.2. Release Test
The in vitro drug release of PTX from the formulation was measured as follows: PTX and PS (all containing 2 mg PTX) were placed in the cellulose membrane pouch (SnakeskinTM Dialysis Tubing 10,000 MWCO; Thermo Fisher Scientific Inc.; Waltham, MA, USA), and 5 mL of SBF (pH 7.4) was added inside the cellulose membrane pouch. The cellulose membrane pouch was put into 50 mL of SBF containing 0.5% Tween 80 in a 250 mL beaker and shaken at 100 rpm in a dual-motion shaker (Finepcr; Gunpo-si, Republic of Korea). Samples (100 μL aliquots) were collected for 48 h, and then diluted 100-fold with mobile phase and injected into the LC-MS/MS system to measure the PTX concentration.
Although the intrinsic solubility of PTX in aqueous media is low (0.3 µg/mL), SBF containing 0.5% Tween 80 used as a release medium significantly increases PTX solubility (i.e., PTX solubility > 10–100 µg/mL at 0.1–3% Tween 80) [
27]. In this study, PS in 5 mL SBF was placed in a dialysis pouch, while the exterior release medium consisted of 50 mL of SBF + 0.5% Tween 80. This large external volume and surfactant content allowed the PTX released from the pouch to diffuse into the external medium and remain dissolved.
2.2.3. Scanning Electron Microscopy (SEM)
The particle size and surface morphology of PTX, SMB7, and PS were observed using a SU8220 Scanning Electron Microscope (Hitachi; Tokyo, Japan). Samples were coated under conditions of 5.0 kV, 15 mA, and 10 min using a sputter coater (EMI Tech, K575k, West Sussex, UK).
2.2.4. Differential Scanning Calorimetry (DSC)
The thermograms for PTX, SMB7, and PS were obtained using a thermal analysis instrument (DSC Q2000, V24.4 Build 116; TA Instruments, New Castle, DE, USA). Then, 5 mg of PTX, SMB7, or PS was placed in a Tzero aluminum pan. The instrument was operated under a nitrogen atmosphere (50.0 mL/min), and the following thermal program was applied: equilibration at 10 °C, followed by an isothermal step for 1 min, a temperature ramp of 5 °C/min up to 300 °C, and a final isothermal step for 1 min.
2.2.5. X-Ray Diffraction (XRD)
An Empyrean X-ray diffractometer equipped with a copper anode (Malvern Panalytical Ltd.; Malvern, UK) was used to analyze the X-ray diffraction pattern of PTX, SMB7, and PS under the generator voltage of 40 kV and tube current of 30 mA. XRD patterns were recorded using Cu Kα radiation (λ = 1.54 Å) over a 2θ range of 5–70° with a scanning speed of 0.05°/s. Major diffraction peaks were identified and assigned Miller indices (hkl) based on matching observed 2θ values with literature-reported peaks for crystalline PTX.
2.2.6. Fourier Transform Infrared Spectroscopy (FT-IR)
FT-IR pattern for PTX, SMB7, and PS was obtained using an FT-IR spectrophotometer (Frontier, PerkinElmer, Norwalk, CT, USA) with a scan range of 400–4000 cm−1
2.2.7. Degradation of PS
Each 2 mg sample of SMB7 and PS was suspended in 1 mL of SBF (pH 7.4) and placed in a 2 mL tube. The tube was stirred in a Confido-S20H micro mixer incubator (Finepcr; Gunpo-si, Republic of Korea) at 37 °C at 700 rpm. The degradation profile of each formulation was monitored for 5 days. Every 24 h, a carbon-coated copper grid (Formvar/carbon 200 mesh; Ted Pella Inc.; Redding, CA, USA) was gently immersed in each 2 mL sample tube to allow the adsorption of the particles onto the grid surface. After approximately 1 min of soaking, the grid was removed, air-dried, and observed using an HT 7700 Bio-transmission electron microscope (Hitachi; Tokyo, Japan) to assess the structural integrity of the formulation over time.
2.3. Cell Viability
B16F10 cells were seeded at a density of 4 × 104 cells per well in a 96-well plate and incubated at 37 °C in a 5% CO2 atmosphere for 24 h to allow cell attachment and stabilization before drug administration. After 24 h, the medium was replaced with fresh culture media containing various PTX formulations (i.e., PTX, PS, PS with CUR, PS with TPGS, and PS with verapamil) at increasing concentrations (0, 10, 20, 50, 100, 200, 500, and 1000 nM) of PTX or its equivalent amount.
After 48 h of drug exposure, MTT reagent (0.5 mg/mL in PBS, 15 μL) was added to each well, followed by an additional 4 h incubation to allow mitochondrial dehydrogenase enzymes in viable cells to reduce MTT into insoluble formazan crystals. The resulting formazan was then solubilized using 150 μL DMSO, and the absorbance was measured at 592 nm using an ultraviolet (UV) spectrophotometer. Cell viability was calculated relative to the untreated control group. Data were fitted using the inhibitory effect model . Emax and IC50 represent uninhibited cell viability (%) and the concentration of PTX (nM) at which half-maximal inhibition occurs. [I] represents the concentration of PTX.
2.4. Cellular Accumulation of PTX Formulation in B16F10 Cells
B16F10 cells were seeded at a density of 3 × 105 cells per well in a 6-well plate and incubated at 37 °C with 5% CO2 overnight. The cells were treated with PTX formulations (i.e., PTX, PS, PS with CUR, PS with TPGS, and PS with verapamil) at 1 μM of PTX or its equivalent. After 1 h of incubation, cells were washed with PBS and harvested. Cells were added to 150 μL of IS solution (1 ng/mL berberine in 80% MeOH) and vortexed for 5 min, followed by centrifugation at 16,000× g and 4 °C for 5 min. A 120 μL aliquot of supernatant was transferred to an autosampler vial, and 1 μL was injected into the LC–MS/MS system to measure the intracellular PTX concentration.
2.5. Cytotoxicity of PTX Formulation in 3D Cultured B16F10 Spheroid
B16F10 cells were seeded at a density of 3 × 105 cells per well in an ultra-low attachment 96-well plate to facilitate spheroid formation. Immediately after seeding, the plate was centrifuged at 15,000 rpm for 15 min to promote cell aggregation. Following centrifugation, the spheroids were allowed to stabilize for 24 h before drug treatment. The spheroids were treated with the following conditions and grown for 5 days: control, 200 μg/mL of SMB7, 200 μg/mL of curcumin, 25 μg/mL of PTX, PS (equivalent to 25 μg/mL PTX), PS with curcumin (equivalent to 25 μg/mL PTX + 200 μg/mL curcumin), and PS with TPGS (equivalent to 25 μg/mL PTX + 600 μg/mL TPGS).
During the culture period, the spheroid radius was measured daily to assess tumor growth inhibition across different treatment groups. After five days, the spheroids were harvested, disrupted into a single-cell suspension, and analyzed for intracellular PTX and curcumin concentrations using LC–MS/MS. Additionally, the culture medium was collected from each well and analyzed to quantify the remaining extracellular concentrations of PTX and curcumin, providing further insights into drug uptake and retention within the spheroids.
2.6. Apoptosis Analysis in B16F10 Cells Using Flow Cytometry
B16F10 cells were seeded at a density of 8 × 104 cells in a cell culture dish and cultured until a confluency of 80% was reached. Upon reaching the desired confluency, the culture medium was aspirated and replaced with fresh medium containing one of the following treatments: control, PTX (25 μg/mL), PS (equivalent to 25 μg/mL PTX), PSC (equivalent to 25 μg/mL PTX + 200 μg/mL curcumin), and PSCT (equivalent to 25 μg/mL PTX + 200 μg/mL curcumin + 600 μg/mL TPGS).
After 24 h of treatment, the culture supernatant was collected into a 15 mL conical tube. Cells were then detached using 1.5 mL of trypsin, and the resulting cell suspension—including detached cells and debris—was combined with the previously collected medium in the same tube. The samples were centrifuged at 1000 rpm for 3 min, and the supernatant was carefully discarded. The cell pellet was resuspended in 1 mL of DMEM supplemented with 10% FBS and 1% penicillin–streptomycin. Subsequently, 100 μL of the Muse Annexin V and Dead Cell kit (Cytek Biosciences Inc., Fremont, CA, USA) was added to the cell suspension, and the samples were incubated at room temperature in the dark for 20 min. Apoptosis and cell viability were then assessed using the Guava® Muse® Cell Analyzer (Cytek Biosciences Inc., Fremont, CA, USA).
2.7. Confocal Staining
B16F10 cells were seeded at a density of 4 × 104 cells per well in a 24-well plate and cultured until a confluency of 80% was reached. Once the desired confluency was achieved, the culture medium was replaced with fresh media containing the following respective treatments: control, 200 μg/mL of SMB7, 200 μg/mL of curcumin, 25 μg/mL of PTX, PS (equivalent to 25 μg/mL PTX), PS with curcumin (equivalent to 25 μg/mL PTX + 200 μg/mL curcumin), and PS with TPGS (equivalent to 25 μg/mL PTX + 600 μg/mL TPGS).
Cells were incubated post-treatment for 24 h and then fixed and stained to visualize intracellular structures. DAPI was used for DNA staining to highlight nuclear morphology, and goat anti-Mouse IgG conjugated with FSD 594 was employed for β-tubulin staining to observe microtubule organization. As PTX primarily acts by stabilizing microtubules, fluorescence images were acquired using an LSM700 confocal laser scanning microscope (Carl Zeiss GmbH, Oberkochen, Germany). Confocal images were acquired at 100 × magnification to enhance the visualization of DNA and β-tubulin structures, enabling the assessment of PTX-induced cytoskeletal disruption and apoptosis-related nuclear changes.
2.8. Antitumor Activity and Pharmacokinetic Study of PTX Formulation in B16F10 Xenograft Mice
C57BL/6 mice (male, 6 weeks old) were purchased from Samtaco (Osan, Republic of Korea) and used as B16F10 tumor xenograft models. Mice were maintained under pathogen-free conditions.
B16F10 melanoma cells (1 × 10
6 cells in 200 µL PBS) were subcutaneously implanted into the dorsal flank of forty C57BL/6 mice. Tumor progression was monitored daily, and when the tumor volume reached approximately 100 mm
3, each of the ten mice was randomly assigned to four treatment groups. Each group received SC injections of the assigned formulations daily for seven consecutive days (
Figure 1). The minimum number of mice required to achieve adequate study power (significance level of 0.05 and statistical power of 80%) to monitor a 30% reduction in tumor volume change was estimated to be 10 mice per group based on our previous results of Kim et al. [
26] and sample size calculators (clincalc.com). Based on these estimates, we conducted an antitumor activity study on a total of 40 B16F10 xenograft mice (10 mice per group).
During treatment, tumor volume and body weight were measured daily throughout this study using the following formula: V = 0.5 × A × B2. Here, A denotes length and B represents width. Mice were excluded if they showed signs of abnormal behavior, rapid tumor necrosis, tumor volume exceeding 3000 mm3, or weight loss exceeding 20% during the study period. Two mice from the control groups died on the 5th and 6th days of vehicle treatment, and two mice were excluded because of tumor volume exceeding 3000 mm3 on the 7th day. Finally, six mice from the control group were included in the data analysis. No mice in the PTX + CUR, PSC, and PSCT groups did not meet the exclusion criteria during the experiment; thus, all animals were included in the final analysis, and pharmacokinetic and biodistribution analyses were performed.
After the final injection, the mice from PTX + CUR, PSC, and PSCT groups were randomly subdivided into two time points (n = 5 for each subgroup; 2 h and 8 h post-administration) for pharmacokinetic and biodistribution analyses. At the designated time points (0.25, 1, and 2 h from the 2 h group; 0.5, 4, and 8 h from the 8 h group), blood samples (approximately 80 µL) were collected from the retro-orbital plexus of mice. For the PTX and CUR analyses, 30 µL of plasma aliquots were stored at −80 °C. At 2 h, the mice were anesthetized, and the organs (heart, lung, liver, kidney, pancreas, and tumor) were harvested, weighed, and homogenized using a MM400 Laboratory Ball Mixer Mill (Retsch, Haan, Germany). Aliquots (50 µL) of 20% tissue homogenates prepared using 10% MeOH were stored at −80 °C for subsequent analyses.
Aliquots (30 μL) of plasma and aliquots (50 μL) of tissue homogenate samples were added with 120 and 150 μL of IS solution (1 ng/mL berberine in MeOH), respectively, and vortexed for 5 min, followed by 5 min centrifuged at 16,000× g at 4 °C. A 120 μL aliquot of supernatant was transferred to an autosampler vial and 1 μL was injected into the LC-MS/MS system.
2.9. SMB7 Toxicity Evaluation
Fourteen C57BL/6 mice (male, 6 weeks old) were randomly divided into two groups (
n = 7 per group) and received daily subcutaneous injection either vehicle (saline 4 mL/kg) or SMB7 (100 mg/kg suspended in 4 mL saline) for seven consecutive days. After the completion of the treatment period, mice were humanely euthanized according to approved protocols. Major organs (heart, lung, kidney, pancreas, and liver) were harvested, gently blotted to remove excess blood, and weighed. Body weights were recorded daily throughout the experiment and at the final endpoint. The minimum number of mice required for adequate study power (significance level of 0.05 and statistical power of 80%) to monitor a 20% increase or decrease in organ weight change was estimated to be 3~7 mice per group for major organs based on our previous results of Kim et al. [
26] and sample size calculators (clincalc.com). Based on these estimates, we conducted a toxicity assessment on a total of 14 mice (7 mice per group).
2.10. LC-MS/MS Analysis
The concentrations of PTX and CUR in the biological samples were analyzed simultaneously using a TSQ Altis Plus LC-MS/MS system (Thermo Fisher Scientific Inc.; Waltham, MA, USA). The separation was performed on a Luna C18 column (2.0 × 150 mm, 5 μm particle size; Phenomenex, Torrance, CA, USA) using a mobile phase consisting of water and methanol (20:80, v/v) with 0.1% formic acid at a flow rate of 0.2 mL/min. The column temperature was maintained at 40 °C, and the injection volume was 1 μL. Quantification was performed using multiple reaction monitoring (MRM) at m/z 876.4 → 308.0 for PTX (retention time TR 3.09 min), at m/z 369.1 → 285.0 for CUR (TR 3.16 min), and at m/z 336.2 → 320.1 for berberine (IS; TR 2.34 min). The fragmentor voltage was 25 V, and collision energies for 16–27 eV for PTX, CUR, and berberine in a positive ionization mode.
2.11. Data Analysis
Non-compartmental methods (WinNonlin version 5.1, Pharsight Co., Mountain View, CA, USA) were used to estimate the pharmacokinetic parameters.
The biodistribution of PTX in various mouse tissues was quantified and expressed as a percentage of the injected dose per gram of tissue (%ID/g). This parameter reflects the proportion of the administered drug accumulated in a specific organ, normalized by the weight of the organ. The following formula was used to calculate the drug concentration in the tissue, which is measured (e.g., ng), normalized to the total injected dose, and further corrected based on the weight of the harvested tissue to standardize drug accumulation across different organs as follows: .
An analysis of variance (ANOVA) and student’s t-test were used to analyze statistical significance using SPSS Statistics for Windows, version 27 (IBM Corp., Armonk, NY, USA).
4. Discussion
After approval by the US FDA as a medication in 1993, PTX has been established as a first-line therapy for ovarian, breast, lung cancers, and melanoma, demonstrating excellent efficacy, especially when used in combination with other chemotherapeutic agents [
34]. PTX functions as a mitotic inhibitor by disrupting microtubule spindle assembly, thereby inhibiting chromosome segregation and cell division. However, poor solubility and low tumor accumulation of PTX due to P-gp-mediated efflux have limited its clinical application [
35]. To overcome these limitations and enhance solubility while inhibiting P-gp, we introduced PTX-loaded mesoporous silica nanoparticles, co-administered with CUR and TPGS.
In our previous study, CUR was fully embedded within mesoporous nanosilicate SMB7, as confirmed through SEM and FT-IR analyses. Additionally, data from XRD and DSC revealed that the physicochemical properties of CUR-loaded SMB7 promoted an amorphous state [
26]. Similarly, the irregular square-shaped crystalline structures of PTX were not observed in the PS formulation during SEM analysis. Considering the broad halo pattern in XRD, the absence of a distinct melting peak in DSCs with a thermal shift from approximately 220 °C to ~100 °C, and the broadening of FT-IR peaks, it can be concluded that PS exists predominantly in an amorphous state. Moreover, the PS formulation exhibited natural biodegradation, initiating within 2 days and achieving complete degradation within 5 days (
Figure 2). A previous study investigated the IC
50 of SMB7 on B16F10 cells and found that SMB7 did not cause cytotoxicity [
26]. Additionally, our research, including both in vitro cytotoxicity assays and an in vivo study on SMB7 cytotoxicity, confirmed no observed toxicity in biological systems (
Figure 5D,E).
Next, we investigate the in vitro anticancer effect of PTX, PS, PSC, and PSCT in melanoma cells (
Figure 3). Unlike traditional 2D monolayer cultures, 3D spheroid cytotoxicity assays provide a more physiologically relevant model that closely mimics the tumor microenvironment [
36]. In 2D cultures, the absence of an extracellular matrix limits cell–cell and cell–matrix interactions, leading to simplified drug responses and exaggerated sensitivity. Conversely, 3D spheroids enable the formation of extracellular matrix-like structures, promoting natural cell adhesion, signaling pathways, and the development of nutrient and oxygen gradients. This architecture creates drug penetration barriers, contributing to enhanced resistance mechanisms that reflect in vivo conditions [
37]. Additionally, B16F10 melanoma spheroid models capture tumor heterogeneity, enabling the evaluation of long-term drug efficacy and apoptotic responses [
38]. Thus, B16F10 spheroid assays offer superior predictive value for anticancer drug performance compared to conventional 2D assays. This was evidenced by the higher correlation coefficient obtained from B16F10 spheroids than from conventional 2D assays (
Figure 3B,F). In addition, the elevated PTX accumulation in PSCT-treated spheroids suggests that CUR and TPGS effectively inhibit the P-gp-mediated efflux, resulting in sustained drug retention and prolonged cytotoxic effects. Moreover, the enhanced permeation and retention (EPR) effect facilitates the entry of nanosized silicate nanoparticles into the tumor microenvironment, as well as their cellular accumulation [
39].
The improved cytotoxicity and enhanced intracellular accumulation observed in the PSCT-treated B16F10 2D cultures and 3D spheroids suggest that CUR and TPGS not only enhanced the intracellular accumulation of PTX by inhibiting P-gp activity but also facilitated mitochondrial dysfunction, leading to apoptosis (
Figure 4). The combined data from 3D spheroid cytotoxicity, MTT assays, and PTX accumulation studies highlight the therapeutic potential of PSCT as an optimized drug delivery system, offering improved drug retention, enhanced cytotoxicity, and superior tumor growth inhibition compared to conventional PTX formulations. The interplay between PTX uptake, microtubule disruption, and apoptosis induction was demonstrated through uptake studies, confocal imaging, and flow cytometry (
Figure 3E and
Figure 4).
PTX functions as a mitotic inhibitor by disrupting microtubule spindle assembly, thereby inhibiting chromosome segregation and cell division. This mechanism leads to G2/M phase arrest and triggers apoptosis [
35]. This process is tightly linked to the generation of reactive oxygen species and the activation of caspase cascades, which ultimately lead to apoptosis [
40]. It is known that PTX-induced microtubule dysfunction initiates apoptosis via both c-Jun N-terminal kinase (JNK)-dependent and -independent pathways in many cancer cells, leading to the activation of caspase-3 and PARP cleavage, which are key markers of the apoptotic process [
41,
42]. In this study, the downstream effect of microtubule disruption by PTX was reflected in the flow cytometry apoptosis analysis. While late apoptosis percentages were similar across PS, PSC, and PSCT, early apoptosis showed substantial differences. The early apoptosis rate was increased from ~5.4% in PS to ~11.0% in PSC, highlighting the role of CUR as a P-gp inhibitor that enhances intracellular drug retention. Notably, PSCT, which includes both CUR and TPGS, exhibited an early apoptosis rate of ~41.75%, nearly 4-fold higher than PSC (
Figure 4A,B).
CUR, while known for its P-gp inhibitory properties, also interferes with tubulin dynamics. CUR binds to tubulin, disrupts microtubule polymerization, reduces GTPase activity, and promotes tubulin dimer aggregation, leading to cell cycle arrest at the G2/M phase and subsequent apoptosis [
43]. Confocal imaging revealed that CUR reduces microtubule network density compared to the control and SMB7, although not to the extent observed with PTX. PSC demonstrated enhanced anticancer effects, while the addition of TPGS further amplified these effects due to its strong P-gp inhibition (
Figure 4C).
TPGS directly binds to the ATP-binding site of P-gp, inhibiting ATPase activity and inducing structural changes that impair drug efflux function [
21]. In contrast, CUR indirectly suppresses ATPase activity and reduces P-gp expression through the PI3K/Akt/NF-κB signaling pathway [
44]. Due to these differences, TPGS is more effective in directly inhibiting P-gp function, while CUR excels in regulating expression and suppressing energy metabolism. Thus, utilizing these distinct mechanisms may provide a synergistic effect in P-gp inhibition. The combined results from confocal microscopy and uptake assays confirm the dual role of CUR in inhibiting tubulin polymerization and P-gp activity; meanwhile, TPGS primarily enhances drug accumulation by inhibiting P-gp. This suggests a synergistic effect of CUR and TPGS in inhibiting P-gp, resulting in sustained PTX accumulation and the rapid, early activation of apoptotic signaling. Moreover, the combination of CUR and TPGS likely enhances mitochondrial membrane destabilization, thereby further promoting the initiation of apoptosis [
45,
46]. In summary, improved PTX uptake via P-gp inhibition (by CUR and TPGS) leads to greater microtubule disruption, ultimately accelerating early apoptosis. This mechanistic pathway underscores the potential of the PSCT formulation to overcome drug resistance and improve anticancer efficacy through optimized drug delivery and apoptotic induction.
Similar previous studies have reported enhanced tumor accumulation and efficacy using PTX-loaded mesoporous silica carriers. Fu et al. demonstrated a 6.5-fold increase in tumor PTX accumulation and a 3.2-fold extended peritoneal residence time compared to free PTX [
47]. Our PSCT system exhibited a 135-fold higher %ID/g compared with free PTX + CUR and a 20.8-fold tumor-to-liver PTX concentration ratio. Considering the %ID/g metric offers critical insight into the selectivity and efficiency of tumor targeting, PSCT treatment achieved markedly higher tumor-specific accumulation of PTX compared to all treatment groups, highlighting its superior delivery efficiency and specificity (
Figure 6C). This is particularly significant when considering off-target organ accumulation, especially in the liver, a common site of clearance for nanoparticles. The tumor-to-liver PTX ratio was highest in PSCT, indicating a favorable biodistribution profile and minimized hepatic burden (
Figure 6C).
Regarding the PTX release profile, Jia et al. reported that spherical mesoporous silica nanoparticles with a small pore diameter (3.03 nm) yielded sustained PTX release and showed higher AUC and longer MRT values compared to larger pore size (9.68 nm) [
48]. Additionally, Fang et al. demonstrated that PTX-loaded mesoporous silica nanoparticles shaped as hexagonal plates exhibited higher AUC and longer MRT values and superior antitumor efficacy compared to spherical or rod-shaped nanoparticles [
49]. These results suggest that a nanosilicate carrier with a sustained release profile of PTX may increase bioavailable PTX and tumor targeting and, thereby, have therapeutic potential. Our PSCT formulation is based on spherical mesoporous silica nanoparticles with a small pore diameter (2.54 nm), which retains several important advantages, including sustained PTX release (57.6% for 48 h) and increased tumor-specific PTX accumulation. Notably, spherical mesoporous silica nanoparticles can achieve efficient tumor penetration when properly sized (e.g., 100–200 nm) and offer well-established in vivo safety and formulation scalability [
50]. The PSCT and SMB7 nanosilicate carriers in this study exhibited limited systemic toxicity during the treatment period. Therefore, our findings highlight that appropriately engineered spherical mesoporous silica nanoparticles with small pore diameters, such as PSCT in this study, can prolong drug release, enhance intracellular accumulation, exhibit potent anticancer effects, and reduce systemic toxicity.
Despite advancements in subcutaneous injection of PTX formulations, significant challenges remain in balancing injectability with therapeutic efficacy. While improving drug stability, high-viscosity formulations can cause necrosis at the injection site or inconsistent absorption due to various tissue characteristics. This phenomenon is exacerbated in individuals with a higher body mass index or compromised skin integrity. For instance, studies have shown that flow rates exceeding 10 mL/min at viscosities > 50 cP can induce tissue fracturing, thereby altering drug distribution and bioavailability [
51]. Future investigations should emphasize the co-development of devices and formulations, such as thinner polymers or hyaluronidase analogs, to reduce viscosity without compromising payload capacity [
51,
52]. Additionally, long-term local toxicity profiles remain unexplored. While preclinical models have reported manageable profiles, the impact of chronic subcutaneous injection on tissue fibrosis or immune cell activation in humans remains unclear [
53]. Rigorous pharmacokinetic–pharmacodynamic studies are necessary to correlate tumor exposure with therapeutic efficacy and tolerability [
52,
54].