Paclitaxel-Loaded PBCA Nanoparticles for Targeted Drug Delivery in Ovarian Cancer

  1. Hamid Ghahremani ,
  2. Zahra Ourang ,
  3. Shadi Izadidehkordi ,
  4. Sara Zandi ,
  5. Mehdi Ebadi ,
  6. Mohammadreza Ebrahimzade

Vol 10 No 3 (2025)

DOI 10.31557/apjcb.2025.10.3.679-687

Abstract

Overview: Resistance to paclitaxel remains a critical barrier in the effective treatment of ovarian cancer, often resulting in reduced clinical responses and increased recurrence rates. Nanoparticle-mediated drug delivery has emerged as a promising strategy to overcome such resistance by enhancing drug bioavailability and targeting tumor cells more precisely. The present study focuses on the formulation of poly(butyl cyanoacrylate) (PBCA) nanoparticles to facilitate controlled paclitaxel delivery and improve its therapeutic efficacy against drug-resistant ovarian cancer cells.


Methods: Paclitaxel-loaded PBCA nanoparticles were synthesized using a mini-emulsion polymerization technique. Physicochemical properties were assessed by measuring hydrodynamic size, polydispersity index (PDI), zeta potential, and in vitro drug release behavior. Morphological evaluation was conducted via Scanning Electron Microscopy (SEM) to confirm particle uniformity and surface characteristics. Cytotoxicity was examined against the A2780CIS ovarian cancer cell line following 48 hours of exposure to the nanoformulation and free paclitaxel.


Results: The formulated nanoparticles displayed a spherical morphology with an average diameter of 355 nm, a PDI of 0.29, and a surface charge of –18.4 mV. Drug release profiling demonstrated a sustained and controlled release, with approximately 42% of paclitaxel released over 40 hours under physiological conditions. Cellular viability assays revealed that treatment with paclitaxel-loaded PBCA nanoparticles led to a 68% reduction in cell viability, significantly outperforming free paclitaxel, which showed a 41% decrease under identical conditions (p < 0.01).


Conclusion: PBCA nanoparticles exhibited favorable physicochemical characteristics and enhanced anticancer activity in paclitaxel-resistant ovarian cancer cells. These findings support their potential application as a targeted and efficient nanocarrier system for improving the therapeutic index of chemotherapeutic agents in drug-refractory malignancies.

Introduction

Recent technological and scientific breakthroughs in healthcare and industry have markedly improved the efficiency with which new methods and procedures are developed, contributing to stronger economic growth. Nevertheless, alongside these benefits, the rapid pace of innovation can generate additional stress in everyday life, highlighting the need to balance progress with well-being [1-11]. For instance, the integration of advanced computer systems into medical practice ranging from digital imaging and electronic health records to clinical decision-support tools has transformed patient care by enhancing diagnostic accuracy, streamlining workflows, and improving overall outcomes [12]. Cancer is a heterogeneous group of diseases characterized by uncontrolled cell proliferation and metastasis, posing a major global health challenge; recent research has explored its various forms, including breast cancer models such as MCF 7, oral squamous cell carcinoma, cervical cancer, other gynecologic malignancies, and lung cancer, each demanding tailored diagnostic and therapeutic strategies [13-23]. Cancer management strategies encompass both therapeutic interventions such as using radiopharmaceuticals for pain relief in metastatic disease and preventive measures to minimize iatrogenic risks, for example, shielding techniques during diagnostic imaging [24, 25]. In addition to genetic determinants implicated in diseases such as cancer, environmental factors like background radiation can influence the activity of key stress response genes, potentially altering disease susceptibility and progression [26]. Ovarian cancer remains one of the most lethal gynecologic malignancies worldwide, primarily due to its late diagnosis and frequent development of resistance to standard chemotherapeutic agents [27]. Among the first-line treatments, paclitaxel has shown considerable efficacy through its ability to stabilize microtubules and inhibit mitosis [28]. However, prolonged use often leads to the emergence of drug-resistant tumor cells, severely compromising clinical outcomes and limiting long-term survival [29-31]. The mechanisms underlying paclitaxel resistance are multifactorial, involving alterations in drug efflux, apoptosis pathways, and microtubule dynamics, which collectively demand the development of novel therapeutic strategies [32-34]. In many cases, pharmaceutical formulations must undergo preclinical evaluation in vitro on cell cultures or in vivo in animal models to assess their efficacy and safety before advancing to clinical trials [35, 36]. Nanotechnology stands out as a leading example of technological progress, having revolutionized targeted drug delivery in medicine and enabled novel chemical processes in industry by manipulating materials at the molecular and atomic scale [37-45]. Nanotechnology-based drug delivery systems have emerged as a promising avenue to overcome these limitations by improving drug solubility, stability, and tumor-specific accumulation [31]. Nanoparticles have been applied in diverse biomedical contexts, such as using nanoliposomes to transport DNAzymes across the blood–brain barrier [46] and employing 5 ALA– conjugated hollow gold nanoparticles to enhance radio and photosensitivity in KYSE esophageal cancer cells [47]. Recent advances in targeted delivery systems have demonstrated the potential for precision therapeutic approaches, including mRNA-encapsulated lipid nanoparticles that enable controlled modulation of specific cellular pathways, highlighting the broader applicability of nanoparticle-based platforms for achieving enhanced therapeutic specificity in cancer treatment [48]. Alginate based nano hybrid hydrogels have emerged as a promising drug delivery platform in oncology, leveraging their biocompatibility and tunable release kinetics to achieve targeted accumulation and sustained release of anticancer agents within tumor tissues [49]. Poly(butyl cyanoacrylate) (PBCA) nanoparticles, in particular, have garnered attention due to their biocompatibility, biodegradability, and ability to encapsulate hydrophobic chemotherapeutic agents such as paclitaxel [31]. These carriers can prolong systemic circulation time and provide controlled drug release, thereby enhancing cytotoxic effects at the tumor site while minimizing off-target toxicity [31]. In this study, paclitaxel-loaded PBCA nanoparticles were designed and evaluated as a targeted delivery platform for ovarian cancer therapy. The formulation was characterized in terms of hydrodynamic size, polydispersity index (PDI), surface charge, drug release kinetics, and morphological features. Furthermore, the cytotoxic potential of the nanoformulation was assessed against a drug-resistant ovarian cancer cell line (A2780CIS) to determine its therapeutic advantages over free paclitaxel. A schematic illustration of the synthesis and drug release mechanism of paclitaxel-loaded PBCA nanoparticles is presented in Figure 1.

Figure 1. Schematic Illustration of the Synthesis and Drug Release Mechanism of Paclitaxel-loaded poly (butyl cyanoacrylate) (PBCA) Nanoparticles. In the mini-emulsion polymerization step, paclitaxel and butyl cyanoacrylate monomers are incorporated into a nanoscale matrix. The resulting spherical nanoparticles enable sustained drug release under physiological conditions, thereby improving the therapeutic index of paclitaxel in resistant ovarian cancer cells.

Methods and Materials

Materials

Butyl cyanoacrylate monomer was purchased from Evobond® (TongShen Enterprise Co., Ltd., Taiwan). Polyethylene glycol (PEG400) and dextran (MW 40,000) were obtained from Sigma–Aldrich Co. (UK). Hydrochloric acid and sodium hydroxide were provided by Merck (Germany). Olive oil and honey were sourced from Farzan Rahbar Saba Co. and Sabalan Co. (Iran), respectively. The A2780CIS ovarian cancer cell line was obtained from the Iranian Pasteur Institute Cell Bank.

Preparation of Drug-Loaded PBCA Nanoparticles

Paclitaxel-loaded PBCA nanoparticles were prepared via a modified mini-emulsion polymerization technique under controlled laboratory conditions. Initially, a stabilizing aqueous phase was prepared by dissolving 40 mg of dextran (MW 40,000) in 0.01 N hydrochloric acid (200 µL), followed by the incorporation of 120 mg of natural honey and 30 µL of olive oil under gentle magnetic stirring (150 rpm). Once homogeneity was achieved, 250 µL of butyl cyanoacrylate monomer was added dropwise to the mixture, allowing pre-polymer dispersion.

Subsequently, 45 mg of paclitaxel was introduced into the formulation and mixed thoroughly. The entire mixture was subjected to stirring at 400 rpm for 10 minutes to form a pre-emulsion. To initiate emulsification, 20 mL of cold distilled water was gradually added in two equal steps while maintaining constant agitation. Probe sonication was then performed using a Bandelin Sonopuls HD 2070 (50 W) ultrasonic processor, with the sample vessel placed in an ice bath to prevent thermal degradation during sonication. The resulting emulsion was refrigerated at 4 °C for 24 hours to allow initial polymer network stabilization. Following this incubation period, the dispersion was returned to the magnetic stirrer and maintained at 150 rpm for 3.5 hours at ambient temperature to ensure complete polymerization of the monomer and proper drug entrapment. Finally, the pH of the colloidal suspension was carefully adjusted to physiological levels using 0.1 N sodium hydroxide.

Characterization of Nanoparticles

The physicochemical characteristics of the formulated nanoparticles, including hydrodynamic diameter, polydispersity index (PDI), and zeta potential, were evaluated using a Zetasizer Nano ZS3600 (Malvern Instruments, UK). For this purpose, the nanoparticle suspensions were diluted in phosphate-buffered saline (PBS, pH 7.2, 10 mM) at a ratio of 1:20 prior to measurement, ensuring optimal light scattering conditions. Drug loading (DL%) and encapsulation efficiency (EE%) were determined spectroscopically following nanoparticle purification. Both formulations were subjected to ultracentrifugation at 49,000 × g for 15 minutes at 4 °C to separate the unencapsulated paclitaxel from the nanoparticle pellet. The supernatant was carefully decanted, and a secondary ultracentrifugation cycle was performed to ensure the complete removal of loosely associated drug molecules. The concentration of unencapsulated paclitaxel in the final supernatant was quantitatively analyzed using Inductively Coupled Plasma Mass Spectrometry (ICP-MS). Encapsulation efficiency and drug loading were calculated using the following equations:

To enable morphological analysis, the nanoparticle suspensions were lyophilized in the presence of 3% (w/v) mannitol as a cryoprotectant. The resulting dry powders were imaged using Scanning Electron Microscopy (SEM) (XL30, Philips, Netherlands) to assess surface topology and particle structure.

Drug Release Evaluation

To assess the release profile of paclitaxel from the nanoparticle formulation, a suspension containing 0.8 mg/ mL of drug-loaded nanoparticles was prepared in human serum to mimic physiological conditions. The samples were incubated in a shaking incubator at 37°C, maintained at 130 rpm for 30 minutes to initiate interaction between serum components and the nanoparticles. Drug release was monitored over a 40-hour period by measuring the degradation-dependent release of paclitaxel into the surrounding medium. At predetermined time intervals, aliquots were withdrawn, and the absorbance of the supernatant was recorded at 220 nm using a UV-visible spectrophotometer. This wavelength corresponds to the characteristic absorbance of paclitaxel, allowing for quantitative tracking of its release. The release kinetics were calculated based on absorbance changes, representing cumulative drug release over time.

Cytotoxicity Assessment

The cytotoxic effects of the drug-loaded nanoparticles were investigated using the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay on the A2780CIS ovarian cancer cell line, a model known for its resistance to paclitaxel. Cells were seeded in 96-well plates and treated with serial concentrations (0, 5, 10, 20, 40, 80, and 160 µM) of the nanoformulated drug, free paclitaxel, and control groups corresponding to both formulations. Following 48 hours of incubation under standard culture conditions (37°C, 5% CO₂), MTT solution was added to each well and incubated to allow mitochondrial dehydrogenases in viable cells to reduce MTT to insoluble formazan crystals. Subsequently, the formazan was solubilized using DMSO, and the absorbance was measured at 570 nm using a microplate reader. The resulting data were used to calculate cell viability and determine dose-dependent cytotoxicity profiles of the tested formulations.

Statistical Analysis

All quantitative data were analyzed using SPSS software version 15.0 (IBM Corp., USA). Results were expressed as mean ± standard deviation (SD) based on triplicate independent experiments (n = 3). Statistical significance was determined using one-way analysis of variance (ANOVA) followed by Tukey’s post-hoc test to evaluate differences between experimental groups. A p-value of less than 0.05 was considered statistically significant throughout the analyses. The sample size was selected based on standard practices for preliminary in vitro assays to ensure reproducibility and minimize variability.

Results

Characterization of Nanoparticles

The polymerization of poly(butyl cyanoacrylate) was initiated by the gradual addition of cold distilled water under stirring. The reaction was followed by sonication, which facilitated polymer chain formation and particle dispersion. A visible change in the suspension’s color to milky white indicated successful nanoparticle formation.

The synthesized paclitaxel-loaded PBCA nanoparticles were subjected to physicochemical characterization. Dynamic light scattering (DLS) analysis revealed that the nanoparticles had an average hydrodynamic diameter of 355 ± 12 nm, a polydispersity index (PDI) of 0.29, and a zeta potential of –18.4 ± 1.2 mV, suggesting moderately uniform distribution and good colloidal stability. SEM imaging (Figure 2) confirmed the spherical morphology and relatively smooth surfaces of the nanoparticles.

Figure 2. Scanning Electron Microscopy (SEM) Image of Paclitaxel-loaded PBCA Nanoparticles, Illustrating Uniform Spherical Morphology with Minimal aggregation. Scale bar = 1 µm.

Minor aggregation was observed in some areas, which could be attributed to partial drying during the sample preparation process a common artifact in SEM analysis of soft nanomaterials.

Drug Release

Drug release experiments demonstrated that the paclitaxel-loaded PBCA nanoparticles exhibited a controlled release profile: an initial burst of 7% within the first hour was followed by a gradual release, reaching a cumulative 42% release after 40 h under physiological conditions. In contrast, free paclitaxel displayed a rapid release pattern, with 96 ± 2.8% of the drug released into human serum over the same period (Figure 3)

Figure 3. Drug Release Kinetics of Paclitaxel from PBCA Nanoparticles Versus Free Drug under Physiological Conditions. Data points represent mean ± SD (n = 3)..

Cytotoxicity of Nanoparticles

Initial tests confirmed that blank PBCA nanoparticles (48 µg/mL) exhibited no measurable cytotoxicity, indicating good biocompatibility. Over 48 h (Figure 4), paclitaxel- loaded PBCA nanoparticles demonstrated significantly greater cytotoxic effects than free paclitaxel.

Figure 4. Values Represent Mean IC₅₀ ± standard Deviation (n = 3) for Paclitaxel-loaded PBCA Nanoparticles and free Paclitaxel in A2780CIS cells.

The half‐ maximal inhibitory concentration (IC₅₀) values were 24.1 ± 1.1 µM for the nanoparticle formulation and 41.9 ± 1.9 µM for free paclitaxel. Moreover, the nanoparticle system produced a 68% reduction in A2780CIS cell viability at the IC₅₀ concentration, compared with a 41% decrease for the free drug (p < 0.01), indicating enhanced potency with increasing drug concentration.

Discussion

Targeted drug delivery systems are a cornerstone in the advancement of modern oncology, aiming to maximize therapeutic efficacy while minimizing systemic toxicity [50]. In conventional chemotherapy, nonspecific distribution of drugs like paclitaxel often leads to off-target effects, low bioavailability, and the development of multidrug resistance (MDR) mechanisms, particularly in aggressive cancers such as ovarian carcinoma [51, 52]. The emergence of nanotechnology-based platforms has provided a viable solution to these challenges by enabling enhanced permeability, controlled release, and selective accumulation of chemotherapeutics at the tumor site [50, 51]. Among various nanocarriers, poly (butyl cyanoacrylate) (PBCA) nanoparticles have garnered attention due to their biocompatibility, biodegradability, and capacity to encapsulate hydrophobic drugs like paclitaxel [51]. The current study demonstrates that paclitaxel-loaded PBCA nanoparticles not only improve the physicochemical stability of the drug but also exhibit enhanced cytotoxicity against the A2780CIS ovarian cancer cell line, which is known for its resistance to taxane-based therapies [51]. Our findings indicate that the PBCA nanoparticles had a hydrodynamic diameter of ~355 nm, a negative surface charge, and a moderate PDI, all indicative of a stable colloidal formulation. The drug release profile revealed a sustained pattern, with approximately 42% of paclitaxel released over 40 hours, compared to the 96% burst release of free paclitaxel [51]. This controlled release is advantageous for maintaining therapeutic drug levels over prolonged periods while avoiding toxic peaks [50]. The MTT cytotoxicity assays further validated the therapeutic superiority of the nanoformulation. The nanoparticle group induced a 68% reduction in cell viability, significantly outperforming free paclitaxel (41%, p < 0.01). These findings align with previous reports emphasizing the enhanced uptake and intracellular retention of nanoparticle-bound drugs in resistant tumors through mechanisms like endocytosis and reduced drug efflux via P-glycoprotein (51,53). Notably, blank PBCA nanoparticles demonstrated no detectable cytotoxicity, reaffirming the carrier’s safety profile for biomedical applications. When benchmarked against other nanocarriers such as liposomes or PEGylated micelles, PBCA nanoparticles offer several advantages, including rapid and reproducible formulation, controlled release without burst kinetics, and efficient cellular uptake [54-56]. Furthermore, the use of excipients such as dextran and natural honey introduces additional biocompatibility benefits [57]. However, their biological variability and potential microbial load raise regulatory concerns unless highly purified and standardized [57]. Future studies should focus on refining formulation components, introducing surface PEGylation to prolong circulation, and developing lyophilization techniques for enhanced stability [58, 59]. From a translational perspective, scalability and compliance with Good Manufacturing Practice (GMP) regulations are critical [59]. The mini-emulsion polymerization method employed here is cost-effective and adaptable to scale-up, using readily available reagents and mild reaction conditions. Nevertheless, GMP implementation will require stringent validation, including batch reproducibility, endotoxin testing, and long-term stability evaluations [59]. These results are consistent with prior PBCA research. Researchers demonstrated that doxorubicin-loaded PBCA nanoparticles achieved sustained release and tumor suppression in murine models [54, 55]. while researchers successfully used polysorbate- 80-coated PBCA nanoparticles for brain-targeted delivery of paclitaxel in glioblastoma [57]. Compared to FDA-approved poly(lactic-co-glycolic acid) (PLGA) carriers, which often suffer from burst release and suboptimal encapsulation of hydrophobic drugs, the PBCA system in our study achieved a more desirable balance between drug retention and cytotoxicity [54, 56].

Future Research and Conclusion

Cutting-edge technological developments have profoundly reshaped medicine, dentistry, and biomedical engineering, revolutionizing approaches to diagnosis and treatment. Innovations like deep learning, convolutional neural networks, and advanced imaging methods have improved diagnostic accuracy for conditions ranging from neurological disorders to dental implants and acute medical issues. These advancements have facilitated enhanced classification systems, better biomarker detection, and systematic analyses, leading to improved patient outcomes across various medical fields. This progress highlights the power of integrating state-of-the-art computational tools with clinical practice to tackle complex health challenges effectively [60-73]. In this regard, technology has made significant strides in the treatment of various cancers, particularly in the field of drug delivery. Innovations such as targeted drug delivery systems, nanoparticle-based therapies, and precision medicine have revolutionized cancer treatment by enhancing the efficacy and specificity of therapeutic agents. These advancements allow for more precise targeting of cancer cells, minimizing damage to healthy tissues and reducing side effects [74-83]. Deep learning and imaging innovations have improved cancer detection, with breakthroughs in brain tumor classification [84] and lung cancer segmentation [85]. AI enhances diagnosis and patient care across medical fields [86]. In cancer therapy, targeted drug delivery using nanoparticles improves precision and outcomes. Environmental management benefits from genetic studies of insects [87] and strategies for mass milk disposal [88]. This study underscores the promising potential of PBCA nanoparticles as smart nanocarriers for paclitaxel delivery in drug-resistant ovarian cancer. The nanoformulation exhibited favorable physicochemical properties, sustained drug release, and enhanced cytotoxicity in vitro compared to the free drug. These results suggest that PBCA-based delivery systems can overcome chemoresistance barriers and may contribute to more effective and safer cancer therapies. Further in vivo studies and clinical validation are warranted to fully explore their therapeutic potential.

Acknowledgements

None.

Data availability

Not applicable as we used information from previously published articles.

Ethical issue and approval

Not applicable as we used information from previously published articles.

Consent for publication

All authors have given consent for publication.

Conflict of interest

The authors declare no potential conflict of interest.

References


  1. In vitro comparative effects of alcohol-containing and alcohol-free mouthwashes on surface roughness of bulk-fill composite resins Ayatollahi S, Davoudi A, Momtazi H. BMC Research Notes.2025;18(1). CrossRef
  2. pH-Responsive Microneedle Actuator Array for Precise Wound Healing: Design, Actuation, Light Filtering, and Evaluation. In 2024 IEEE 17th Dallas Circuits and Systems Conference (DCAS) Pour MR , Tan JY , Saha R, Kim A, Kim J. IEEE.2024;:(pp. 1-4). CrossRef
  3. Base Mediated 7-exo-dig Intramolecular Cyclization of Betti-propargyl Precursors: An Efficient Approach to 1,4-oxazepine Derivatives Yazzaf R, Asadi M, Mahdavi M. Current Organic Synthesis.2025. CrossRef
  4. A Novel Approach for Developing Intrusion Detection Systems in Mobile Social Networks Rivandi E, Jamili Oskouie R. Available at SSRN 5174811.2024. CrossRef
  5. Machine Learning for Analyzing Effects of Various Factors on Business Economic Bagherabad MB , Rivandi E, Mehr MJ . Authorea Preprints.2025. CrossRef
  6. Neuroplastic narratives under scrutiny: A critical medical humanities investigation of brain adaptation, psychosocial stressors, and gendered subjectivities in contemporary speculative fiction Jamalpour H, Jamalpour Z, Feiz M. Cultural Forum.2025;2(1):2875. CrossRef
  7. March. Advancements in photothermal mid-infrared spectroscopic imaging for biomedical diagnostics. In Advanced Chemical Microscopy for Life Science and Translational Medicine Gajjela C, Ishrak R, Wu X, Reihani R, Mayerich D, Redd RK . 2025 (p. PC1333216). SPIE..
  8. Monitoring Pseudouridine Synthesis Process Using a MEMS Sensor and Electrochemical Impedance Spectroscopy (EIS) (Master's thesis, Southern Illinois University at Edwardsville) Sohrabi N. 2025.
  9. Modifying Vibratory Behavior of the Car Seat to Decrease the Neck Injury Afsharfard A, Jafari A, Rad YA , Tehrani H, Kim KC . Journal of Vibration Engineering & Technologies.2023;11(3). CrossRef
  10. Sulfur-driven reactive processing of multiscale graphene/carbon fiber- polyether ether ketone (PEEK) composites with tailored crystallinity and enhanced mechanical performance Motta de Castro E, Bozorgmehrian F, Carrola M, Koerner H, Samouei H, Asadi A. Composites Part B: Engineering.2025;295. CrossRef
  11. Efficacy of ozone therapy in dentistry with approach of healing, pain management, and therapeutic outcomes: a systematic review of clinical trials Rezaeianjam M, Khabazian A, Khabazian T, Ghorbani F, Abbasi T, Asghari S, Heidari F, Shiri A, Naderi M. BMC oral health.2025;25(1). CrossRef
  12. Cone-Beam Computed Tomography (CBCT) Assessment of the Inter-Radicular Bone Thickness in the Anterior Maxilla in an Iranian Population Momtazi H, Davoudi A, Ayatollahi S. Journal of Maxillofacial and Oral Surgery.2025. CrossRef
  13. Combined sonodynamic therapy and X-ray radiation with methylene blue and gold nanoparticles coated with apigenin: Impact on MCF7 cell viability Neshastehriz A., Hormozi-Moghaddam Z., Amini S. M., Taheri S. M., Abedi Kichi Z.. International Journal of Radiation Research.2024;22(2). CrossRef
  14. Optimal Pharmaceutical Management Strategies in Cancer Treatment: Novel Approaches Alishahi F, Soudmand N, Goki TG , Rashidoleslami TS . Asian Pacific Journal of Cancer Nursing.2025. CrossRef
  15. Cultural Framings of Cancer: Medical Anthropology on Narrative Intertextuality, Immunotherapeutic Integration, and Neoliberal Resource Conflicts Jamalpour H, Feiz M, Jamalpour Z, Habibi E, Habibi A, Hosseinzadeh N, Khozoee S. Cultural Conflict and Integration.2024;1(1). CrossRef
  16. Herbal Therapies for Cancer Treatment: A Review of Phytotherapeutic Efficacy Jenča A, Mills DK , Ghasemi H, Saberian E, Jenča A, Karimi Forood AM , Petrášová A, et al . Biologics: Targets & Therapy.2024;18. CrossRef
  17. Oral Cancer at a Glance Saberian E, Jenča A, Petrášová A, Jenčová J, Jahromi RA , Seiffadini R. Asian Pacific Journal of Cancer Biology.2023;8(4). CrossRef
  18. Integrative Cancer Care: Leveraging Nutrition and Positive Psychology for Optimal Outcomes Arabmoorchegani M, Abbasi M, Asadalizadeh. M, Motavaf F. Asian Pacific Journal of Cancer Nursing.2025. CrossRef
  19. The Influence of Orthodontic Intervention on Oncology Patients: A Review of Clinical Evidence and Associated Therapeutic Complexities Moravedeh R, Sanaei P. Asian Pacific Journal of Cancer Nursing.2025. CrossRef
  20. Enhancing cancer tissue analysis using photothermal mid-infrared spectroscopic imaging. In Advanced Chemical Microscopy for Life Science and Translational Medicine 2024 (p. PC128550X). SPIE Gajjela C, Ishrak R, Wu X, Reihani R, Mayerich D, Reddy RK . 2024.
  21. Cervical Cancer Tissue Analysis Using Photothermal Midinfrared Spectroscopic Imaging Reihanisaransari R, Gajjela CC , Wu X, Ishrak R, Zhong Y, Mayerich D, Berisha S, Reddy R. Chemical & Biomedical Imaging.2024;2(9). CrossRef
  22. Rapid Hyperspectral Photothermal Mid-Infrared Spectroscopic Imaging from Sparse Data for Gynecologic Cancer Tissue Subtyping Reihanisaransari R, Gajjela CC , Wu X, Ishrak R, Corvigno S, Zhong Y, Liu J, et al . Analytical Chemistry.2024;96(40). CrossRef
  23. New bidirectional recurrent neural network optimized by improved Ebola search optimization algorithm for lung cancer diagnosis Sabzalian MH , Kharajinezhadian F, Tajally A, Reihanisaransari R, Ali Alkhazaleh H, Bokov D. Biomedical Signal Processing and Control.2023;84. CrossRef
  24. Efficacy of 153Sm-EDTMP on Bone Pain Palliation in Metastatic Patients: Breast and Prostate Cancers Rokni M, Amiri M, Gorji KE , Talebian H, Bijani A, Vakili M, Shafiei H, et al . Frontiers in Biomedical Technologies.2023;10(3). CrossRef
  25. Reduction of Breast Surface Dose, Cancer and Mortality Risks Using Lead Apron during the Head Scanning a Computed Tomography Technique Asghari F, Gorji KE , Mehraeen R, Kiapour M, Talebian H, Monfared AS . Iranian Journal of Medical Physics.1401;19(5).
  26. Investigating the expression level of NF-KB and HIF1A genes among the inhabitants of two different background radiation areas in Ramsar, Iran Talebian H, Monfared AS , Niaki HA , Fattahi S, Bakhtiari E, Changizi V. Journal of Environmental Radioactivity.2020;220-221. CrossRef
  27. Ovarian cancer: diagnostic, biological and prognostic aspects Davidson B, Tropé CG . Women's Health (London, England).2014;10(5). CrossRef
  28. Too much of a good thing: suicide prevention promotes chemoresistance in ovarian carcinoma Pennington K., Pulaski H., Pennington M., Liu J. R.. Current Cancer Drug Targets.2010;10(6). CrossRef
  29. Defective apoptosis underlies chemoresistance in ovarian cancer Hajra KM , Tan L, Liu JR . Advances in Experimental Medicine and Biology.2008;622. CrossRef
  30. Molecular and Cellular Basis of Chemoresistance in Ovarian Cancer Asare-Werehene M, Shieh D, Song Y, Tsang B. The Ovary.2019. CrossRef
  31. Emerging Therapeutics to Overcome Chemoresistance in Epithelial Ovarian Cancer: A Mini-Review Cornelison R, Llaneza DC , Landen CN . International Journal of Molecular Sciences.2017;18(10). CrossRef
  32. The role of ABC transporters in ovarian cancer progression and chemoresistance Ween M. P., Armstrong M. A., Oehler M. K., Ricciardelli C.. Critical Reviews in Oncology/Hematology.2015;96(2). CrossRef
  33. Chemoresistance in human ovarian cancer: the role of apoptotic regulators Fraser M, Leung B, Jahani-Asl A, Yan X, Thompson WE , Tsang BK . Reproductive biology and endocrinology: RB&E.2003;1. CrossRef
  34. Molecular determinants of ovarian cancer chemoresistance: new insights into an old conundrum Ali AY , Farrand L, Kim JY , Byun S, Suh J, Lee HJ , Tsang BK . Annals of the New York Academy of Sciences.2012;1271(1). CrossRef
  35. Effect of Acoustic Cavitation on Mouse Spermatogonial Stem Cells: Colonization and Viability Moghaddam ZH , Mokhtari-Dizaji M, Movahedin M. Journal of Ultrasound in Medicine: Official Journal of the American Institute of Ultrasound in Medicine.2021;40(5). CrossRef
  36. Exploring the effects of micro-nano surface topography on MG63 osteoblast-like cell responses: An in vitro study Jafarkhani S, Amiri E, Moazzeni S, Zohoorian-Abootorabi T, Eftekhary M, Aminnezhad S, Khakbiz M. Colloids and Surfaces A: Physicochemical and Engineering Aspects.2023;675. CrossRef
  37. Enhancement of corrosion, biocompatibility and drug delivery properties of nitinol implants surface by Al-Zn-LDH nanohybrids Khakbiz M, Chagami M, Sheibani S, Amiri E, Moazzeni S, Shakibania S, Hou Y, Lee K. Colloids and Surfaces A: Physicochemical and Engineering Aspects.2025;704. CrossRef
  38. Enhancing Cisplatin Delivery via Liposomal Nanoparticles for Oral Cancer Treatment Ghanbarikondori P, Aliakbari RBS , Saberian E, Jenča A, Petrášová A, Jenčová J, Khayavi AA . Indian journal of clinical biochemistry: IJCB.2025;40(2). CrossRef
  39. Preparation, Characterization and Cytotoxic Studies of Cisplatin-containing Nanoliposomes on Breast Cancer Cell Lines Mohammadinezhad F, Talebi A, Allahyartorkaman M, Nahavandi R, Vesal M, Khiyavi AA , Velisdeh ZJ , et al . Asian Pacific Journal of Cancer Biology.2023;8(2). CrossRef
  40. Application of Scaffold-Based Drug Delivery in Oral Cancer Treatment: A Novel Approach Saberian E, Jenča A, Petrášová A, Zare-Zardini H, Ebrahimifar M. Pharmaceutics.2024;16(6). CrossRef
  41. Eco-Friendly Synthesis of Copper Nanoparticles for Efficient Congo Red Dye Removal from Wastewater Karami P, Gashti MP , Martins AF , Fereydouni N. Iranian Journal of Science.2025;:pp.1-15.
  42. Biosynthesized ZnO and MnO nanoparticles from Aegle marmelos peel extract for alkylphenol removal in wastewater Karami P, Gashti MP , Fereydouni N, Martins AF . Results in Chemistry.2025. CrossRef
  43. Enhanced Anticancer Potential of Curcumin-Loaded Liposomal Nanoparticles in Oral Cancer Treatment Moravedeh R, Samadnezhad NZ , Asadalizadeh M, Abbasi M, Nadaki A. Asian Pacific Journal of Cancer Biology.2025;10(2). CrossRef
  44. Improved Antitumor Efficacy of Liposome-Encapsulated Selenium Nanoparticles Asadalizadeh M, Ghahremani H, Ghanbarikondori P, Asadalizadeh H, Rahmani P, Motlagh FR . Asian Pacific Journal of Cancer Biology.2025;10(2). CrossRef
  45. Visible-light responsive La₀.₇Sr₀.₃MnO₃@TiO₂/g-C₃N₄ nanocomposite for photocatalytic antibiotic degradation and bioactivity applications Nejat R, Zandi S. Journal of Alloys and Compounds.2025;1036. CrossRef
  46. Facilitating DNAzyme transport across the blood-brain barrier with nanoliposome technology Hoseinifar MJ , Aghaz F, Asadi Z, Asadi P, Nedaei SE , Arkan E, Pourmotabbed A, Bahrami G, Pourmotabbed T. Scientific Reports.2025;15(1). CrossRef
  47. Compression of radio and photo sensitivity of 5-aminolevulinic acid (5-ALA) conjugated hollow gold nanoparticles (HGNs) on KYSE cell line of oesophageal cancer Mohammadi Z, Imanparast A, Talebian H, Sobhani N, Shabanzadeh M, Sazgarnia A. Nanomedicine Research Journal.2025;9(3). CrossRef
  48. Enhanced epigenetic modulation via mRNA-encapsulated lipid nanoparticles enables targeted anti-inflammatory control Mokhtari T, Taheri MN , Akhlaghi S, Aryannejad A, Xiang Y, Mahajan V, Keshavarz K, et al . bioRxiv: The Preprint Server for Biology.2025. CrossRef
  49. Design and fabrication of alginate hydrogel nanohybrid as a promising cancer treatment Ajayebi FS , Hassanzadeh Nemati N, Hatamirad A, Ghazli M, Attaran N. Iranian Journal of Basic Medical Sciences.2024;27(6). CrossRef
  50. Advancements in Nanotechnology-Based Paclitaxel Delivery Systems: Systematic Review on Overcoming Solubility, Toxicity, and Drug Resistance Challenges in Cancer Therapy. Journal of Angiotherapy Ejeta B, Das M, Das S, Balisa C, Ejeta P. Journal of Angiotherapy.2024. CrossRef
  51. Paclitaxel-loaded poly(n-butylcyanoacrylate) nanoparticle delivery system to overcome multidrug resistance in ovarian cancer Ren F, Chen R, Wang Y, Sun Y, Jiang Y, Li G. Pharmaceutical Research.2011;28(4). CrossRef
  52. Paclitaxel-Loaded Self-Assembled Lipid Nanoparticles as Targeted Drug Delivery Systems for the Treatment of Aggressive Ovarian Cancer Zhai J, Luwor RB , Ahmed N, Escalona R, Tan FH , Fong C, Ratcliffe J, et al . ACS applied materials & interfaces.2018;10(30). CrossRef
  53. MDR1 siRNA loaded hyaluronic acid-based CD44 targeted nanoparticle systems circumvent paclitaxel resistance in ovarian cancer Yang X, Iyer AK , Singh A, Choy E, Hornicek FJ , Amiji MM , Duan Z. Scientific Reports.2015;5. CrossRef
  54. Influence of polybutylcyanoacrylate nanoparticles and liposomes on the efficacy and toxicity of the anticancer drug mitoxantrone in murine tumour models Beck P., Kreuter J., Reszka R., Fichtner I.. Journal of Microencapsulation.1993;10(1). CrossRef
  55. Body Distribution of Free, Liposomal and Nanoparticle-Associated Mitoxantrone in B16-Melanoma-Bearing Mice Reszka R., Beck P., Fichtner I., Hentschel M., Richter J., Kreuter J.. The Journal of Pharmacology and Experimental Therapeutics.1997;280(1). CrossRef
  56. Hyper-cell-permeable micelles as a drug delivery carrier for effective cancer therapy Saw PE , Yu M, Choi M, Lee E, Jon S, Farokhzad OC . Biomaterials.2017;123. CrossRef
  57. Nanoparticle: Significance as Smart Material in Therapeutic Strategies in Drug Delivery in Biological Systems. Application of Biomedical Engineering in Neuroscience Dhungel K, Narayan J. 2019. CrossRef
  58. PEGylated nanoparticles interact with macrophages independently of immune response factors and trigger a non-phagocytic, low-inflammatory response Asoudeh M, Nguyen N, Raith M, Denman DS , Anozie UC , Mokhtarnejad M, Khomami B, et al . Journal of Controlled Release: Official Journal of the Controlled Release Society.2024;366. CrossRef
  59. Opsonization, biodistribution, cellular uptake and apoptosis study of PEGylated PBCA nanoparticle as potential drug delivery carrier Chaudhari KR , Ukawala M, Manjappa AS , Kumar A, Mundada PK , Mishra AK , Mathur R, Mönkkönen J, Murthy RSR . Pharmaceutical Research.2012;29(1). CrossRef
  60. Investigating interactions among health care indicators, income inequality and economic growth: A case study of Iran. Hosseinidoust SE , Hamid Sepehrdoost , Akbar Khodabakhshi , Sharareh Massahi . 2021;:69-94.
  61. Optimized classification of dental implants using convolutional neural networks and pre-trained models with preprocessed data Lashaki RA , Raeisi Z, Razavi N, Goodarzi M, Najafzadeh H. BMC Oral Health.2025;25(1). CrossRef
  62. Enhancing schizophrenia diagnosis through deep learning: a resting-state fMRI approach Raeisi Z, Mehrnia M, Ahmadi Lashaki R, Abedi Lomer F. Neural Computing and Applications.2025;37(20). CrossRef
  63. EEG microstate analysis in trigeminal neuralgia: identifying potential biomarkers for enhanced diagnostic accuracy Lashaki RA , Raeisi Z, Sodagartojgi A, Abedi Lomer F, Aghdaei E, Najafzadeh H. Acta Neurologica Belgica.2025. CrossRef
  64. Automatic diagnosis of autism spectrum disorders in children through resting-state functional magnetic resonance imaging with machine vision Khandan Khadem-Reza Z, Ahmadi Lashaki R, Shahram MA , Zare H. Quantitative Imaging in Medicine and Surgery.2025;15(6). CrossRef
  65. EEG microstate biomarkers for schizophrenia: a novel approach using deep neural networks Raeisi Z, Bashiri O, EskandariNasab M, Arshadi M, Golkarieh A, Najafzadeh H. Cognitive Neurodynamics.2025;19(1). CrossRef
  66. Diagnostic Performance of Magnetic Resonance Imaging for Detection of Acute Appendicitis in Pregnant Women; a Systematic Review and Meta-Analysis Motavaselian M, Bayati F, Amani-Beni R, Khalaji A, Haghverdi S, Abdollahi Z, Sarrafzadeh A, et al . Archives of Academic Emergency Medicine.2022;10(1). CrossRef
  67. Risk of Symptomatic Intracranial Hemorrhage After Mechanical Thrombectomy in Randomized Clinical Trials: A Systematic Review and Meta-Analysis Reda A, Hasanzadeh A, Ghozy S, Sanjari Moghaddam H, Adl Parvar T, Motevaselian M, Kadirvel R, Kallmes DF , Rabinstein A. Brain Sciences.2025;15(1). CrossRef
  68. Emerging drugs for the treatment of irritability associated with autism spectrum disorder Shamabadi A, Karimi H, Arabzadeh Bahri R, Motavaselian M, Akhondzadeh S. Expert Opinion on Emerging Drugs.2024;29(1). CrossRef
  69. Diagnostic Performance of Ultrasonography for Identification of Small Bowel Obstruction; a Systematic Review and Meta-analysis Motavaselian M, Farrokhi M, Jafari Khouzani P, Moghadam Fard A, Daeizadeh F, Pourrahimi M, Mehrabani R, et al . Archives of Academic Emergency Medicine.2024;12(1). CrossRef
  70. Comparison of biochemical factors and liver enzymes in type 2 diabetes patients and healthy individuals. Bulletin of Environment Ebrahimi Far M, Mazdapour M, Kaki A, Mohammadi P, Zakerjafari M, Lavi A, Moradi-Sardareh H. Pharmacology and Life Sciences.2015;4:1-4.
  71. Comparison of loop-mediated isothermal amplification and real-time PCR for detecting Bordetella pertussis Torkaman MRA , Kamachi K, Nikbin VS , Lotfi MN , Shahcheraghi F. Journal of Medical Microbiology.2015;64(Pt 4). CrossRef
  72. Unveiling the menace: a thorough review of potential pandemic fungal disease Jafarlou M. Frontiers in Fungal Biology.2024;5. CrossRef
  73. The emerging role of noncoding RNAs in systemic lupus erythematosus: new insights into the master regulators of disease pathogenesis Afrashteh Nour M, Ghorbaninezhad F, Asadzadeh Z, Baghbanzadeh A, Hassanian H, Leone P, Jafarlou M, et al . Therapeutic Advances in Chronic Disease.2023;14. CrossRef
  74. Regulatory Effects of Apatinib in Combination with Piperine on MDM-2 Gene Expression, Glutathione Peroxidase Activity and Nitric Oxide level as Mechanisms of Cytotoxicity in Colorectal Cancer Cells Mohammadian M, Rostamzadeh Khameneh Z, Emamgholizadeh Minaei S, Ebrahimifar M, Esgandari K. Advanced Pharmaceutical Bulletin.2022;12(2). CrossRef
  75. Carboplatin and epigallocatechin-3-gallate synergistically induce cytotoxic effects in esophageal cancer cells Taghvaei F, Rastin SJ , Milani AT , Khameneh ZR , Hamini F, Rasouli MA , Asghari K, Rekabi Shishavan AM , Ebrahimifar M, Rashidi S. Research in Pharmaceutical Sciences.2021;16(3). CrossRef
  76. Characteristics and Cytotoxic Effects of Nano-Liposomal Paclitaxel on Gastric Cancer Cells Abedi Cham Heidari Z, Ghanbarikondori P, Mortazavi Mamaghani E, Hheidari A, Saberian E, Mozaffari E, Alizadeh M, Allahyartorkaman M. Asian Pacific journal of cancer prevention: APJCP.2023;24(9). CrossRef
  77. Nanoliposomes Meet Folic Acid: A Precision Delivery System for Bleomycin in Cancer Treatment Shakiba D, Shabestari AM , Mokhtari T, Goodarzi MK , Saeed S, Zinatbakhsh Z, Akaberi K, Allahyartorkaman M. Asian Pacific Journal of Cancer Biology.2024;9(4). CrossRef
  78. Studying the Characteristics of Curcumin-Loaded Liposomal Nanoparticles Afyouni I, Ghanbarikondori P, Pour NS , Hashemian PM , Jalali F, Sedighi A, Allahyartorkaman M. Asian Pacific Journal of Cancer Biology.2024;9(2). CrossRef
  79. The Application of Polybutyl Cyanoacrylate (PBCA) Nanoparticles in Delivering Cancer Drugs Salehi V, Izadkhah M, Salehi H, Pour NS , Ghanbarikondori P. Asian Pacific Journal of Cancer Biology.2024;9(2). CrossRef
  80. Targeting Nanog expression increased Cisplatin chemosensitivity and inhibited cell migration in Gastric cancer cells Vasefifar P, Najafi S, Motafakkerazad R, Amini M, Safaei S, Najafzadeh B, Alemohammad H, Jafarlou M, Baradaran B. Experimental Cell Research.2023;429(2). CrossRef
  81. The tumor microenvironment and dendritic cells: Developers of pioneering strategies in colorectal cancer immunotherapy? Ghorbaninezhad F, Nour MA , Farzam OR , Saeedi H, Vanan AG , Bakhshivand M, Jafarlou M, Hatami-Sadr A, Baradaran B. Biochimica Et Biophysica Acta. Reviews on Cancer.2025;1880(2). CrossRef
  82. Hypericin and resveratrol anti‐tumor impact through E‐cadherin, N‐cadherin, galectin‐3, and BAX/BCL‐ratio; possible cancer immunotherapy succor on Y79 retinoblastoma cells Shahriari F, Barati M, Shahbazi S, Arani HZ , Dahmardnezhad M, Javidi MA , Alizade A. Precision Medical Sciences.2025.
  83. Dahmardnezhad, Mozhgan, Tina Foodeh, Sholeh Afshinpoor, and Nastaran Fooladivanda. “Cancer-associated glomerulopathy; an updated review on current knowledge.” Journal of Nephropathology.2024;13(2).
  84. Breakthroughs in Brain Tumor Detection: Leveraging Deep Learning and Transfer Learning for MRI-Based Classification Golkarieh A, Boroujeni SR , Kiashemshaki K, Deldadehasl M, Aghayarzadeh H, Ramezani A. Computer and Decision Making: An International Journal.2025;2. CrossRef
  85. Advanced U-Net Architectures with CNN Backbones for Automated Lung Cancer Detection and Segmentation in Chest CT Images Golkarieh A, Kiashemshaki K, Rezvani Boroujeni S, sadi Isakan N. 2025;:arXiv:2507.09898. CrossRef
  86. Examination of AI’s role in Diagnosis, Treatment, and Patient care. In Gupta, M., Kumar, R., & Lu, Z. (Eds.), Transforming Gender-Based Healthcare with AI and Machine Learning, 2024;221–38. https://doi.org/10.1201/9781003473435-13 Sajjadi Mohammadabadi SM , Seyedkhamoushi F, Mostafavi M, Borhani Peikan M. .
  87. Genetic structure of black soldier flies in northern Iran Boukan A, Nozari J, Naseri Karimi N, Talebzadeh F, Pahlavan Yali K, Oshaghi MA . PloS One.2024;19(8). CrossRef
  88. Emergency mass disposal of milk: Options and considerations Mahdaviarab A., Pahlavanyali K., Cheng R., Wang X., Doria J., Howe J. A., Piñeiro J. M., Spencer J., Liu Z.. Journal of Environmental Management.2025;376. CrossRef

Copyright

© Asian Pacific Journal of Cancer Biology , 2025

Author Details

Hamid Ghahremani
Radio_ oncology Department Science Valiasr Hospital Zanjan University Medical Sciences, Zanjan, Iran.

Zahra Ourang
Department of Biochemistry, School of Medicine, Arak University of Medical Sciences, Arak, Iran.

Shadi Izadidehkordi
Department of Allied Health Sciences, University of Connecticut, Storrs, United States.

Sara Zandi
School of Pharmacy, Sonderegger Research Center, University of Wisconsin–Madison, 777 Highland Avenue, Madison, WI 53705-2222, United States.

Mehdi Ebadi
Doctor of Veterinary Medicine (DVM), Garmsar Branch, Islamic Azad University, Iran.
Mehdiebd1995@gmail.com

Mohammadreza Ebrahimzade
6Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.

How to Cite

1.
Ghahremani H, Ourang Z, Izadidehkordi S, Zandi S, Ebadi M, Ebrahimzade M. Paclitaxel-Loaded PBCA Nanoparticles for Targeted Drug Delivery in Ovarian Cancer. apjcb [Internet]. 23Aug.2025 [cited 29Aug.2025];10(3):679-87. Available from: http://waocp.com/journal/index.php/apjcb/article/view/1897
  • Abstract viewed - 0 times
  • PDF (FULL TEXT) downloaded - 0 times
  • XML downloaded - 0 times