1
|
Raza MA, Khatoon N, Parveen R, Nirisha V, Thakur Y, Thakur Y, Lata K, Shifana AS, Disouza J, Saoji SD, Ajazuddin. Natural macromolecules polysaccharide-based drug delivery systems targeting tumor necrosis factor alpha receptor for the treatment of cancer: A review. Int J Biol Macromol 2025:145145. [PMID: 40516724 DOI: 10.1016/j.ijbiomac.2025.145145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/27/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
Cancer is one of the most significant abnormalities in medical sciences for which a new and efficient therapeutic intervention is desired. Targeting the tumor necrosis factor-alpha receptor appears to have a critical role in both cancer development and immune modality. In this review, an attempt is made to review the potential and avenues of natural macromolecules that have augmented drug delivery systems for specific targeting of the tumor necrosis factor-alpha receptor in cancer therapy. Biological macromolecules, derived from biocompatible and biodegradable sources such as lipids, polysaccharides of natural origin, such as chitosan, hyaluronic acid, alginate, pectin, dextran, starch, cellulose, agar, carrageenan, guar gum, chondroitin sulfate, pullulan, and konjac glucomannan, have been immensely utilized in drug delivery systems for its negligible toxicity mucoadhesive properties, ability to enhance drug stability, and controlled release capabilities. Various novel drug delivery approaches are discussed in detail, including those using polysaccharides, lipids, chitosan, proteins, polymers, dendrimers, exosomes, hydrogels, albumin nanoparticles, silk nanoparticles, and cyclodextrin nanoparticles, incorporating cutting-edge engineering techniques for encapsulation of chemotherapeutic, immunomodulatory, and gene-silencing drugs for site-specific delivery at a site of a tumor. Such macromolecules can mitigate toxicity and even bypass multidrug resistance through their intrinsic property, ligand functionalization for targetability towards receptors. In the present review, an attempt is made to present an outlook for the role of natural macromolecules is being a breakthrough intervention in interfering with tumor necrosis factor-alpha receptor-dependent processes in cancer and a new direction in developing efficient, non-toxic, and personalized therapies for anti-cancer activities.
Collapse
Affiliation(s)
| | - Nousheen Khatoon
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - Rahnuma Parveen
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - V Nirisha
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - Yamini Thakur
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - Yogita Thakur
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - Kumari Lata
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - A S Shifana
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India
| | - John Disouza
- Bombay Institute of Pharmacy & Research, Dombivli East, Taluka: Kalyan 421204, Maharashtra, India
| | - Suprit D Saoji
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, Maharashtra, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India; Rungta College of Engineering & Technology, Bhilai,C.G., India.
| |
Collapse
|
2
|
Aldhubiab B, Almuqbil RM, Nair AB. Harnessing the Power of Nanocarriers to Exploit the Tumor Microenvironment for Enhanced Cancer Therapy. Pharmaceuticals (Basel) 2025; 18:746. [PMID: 40430563 PMCID: PMC12115050 DOI: 10.3390/ph18050746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
The tumor microenvironment (TME) has a major role in malignancy and its complex nature can mediate tumor survival, metastasis, immune evasion, and drug resistance. Thus, reprogramming or regulating the immunosuppressive TME has a significant contribution to make in cancer therapy. Targeting TME with nanocarriers (NCs) has been widely used to directly deliver anticancer drugs to control TME, which has revealed auspicious outcomes. TME can be reprogrammed by using a range of NCs to regulate immunosuppressive factors and activate immunostimulatory cells. Moreover, TME can be ameliorated via regulating the redox environment, oxygen content, and pH value of the tumor site. NCs have the capacity to provide site-specific delivery of therapeutic agents, controlled release, enhanced solubility and stability, decreased toxicities, and enhanced pharmacokinetics as well as biodistribution. Numerous NCs have demonstrated their potential by inducing distinct anticancer mechanisms by delivering a range of anticancer drugs in various preclinical studies, including metal NCs, liposomal NCs, solid lipid NCs, micelles, nanoemulsions, polymer-based NCs, dendrimers, nanoclays, nanocrystals, and many more. Some of them have already received US Food and Drug Administration approval, and some have entered different clinical phases. However, there are several challenges in NC-mediated TME targeting, including scale-up of NC-based cancer therapy, rapid clearance of NCs by the mononuclear phagocyte system, and TME heterogeneity. In order to harness the full potential of NCs in tumor treatment, there are several factors that need to be carefully studied, including optimization of drug loading into NCs, NC-associated immunogenicity, and biocompatibility for the successful translation of NC-based anticancer therapies into clinical practice. In this review, a range of NCs and their applications in drug delivery to remodel TME for cancer therapy are extensively discussed. Moreover, findings from numerous preclinical and clinical studies with these NCs are also highlighted.
Collapse
Affiliation(s)
- Bandar Aldhubiab
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (R.M.A.); (A.B.N.)
| | | | | |
Collapse
|
3
|
Keerikkadu M, Bangera PD, Tippavajhala VK, Rathnanand M. Dasatinib Pharmacokinetics and Advanced Nanocarrier Strategies: from Systemic Limitations to Targeted Success. AAPS PharmSciTech 2025; 26:131. [PMID: 40360939 DOI: 10.1208/s12249-025-03130-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Dasatinib (DSB) is a second-generation tyrosine kinase inhibitor widely used for treating chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph + ALL). Though clinically effective, DSB has some pharmacokinetic drawbacks evidenced by rapid systemic clearance, low oral bioavailability, and poor aqueous solubility requiring high doses for therapeutic action. Novel formulation strategies like solid dispersions, liposomal formulations, and PEGylated and hybrid nanoparticles enhance DSB's pharmacokinetic and pharmacodynamic profiles by enhancing drug solubility, stability, and controlled release. In addition, through these targeted drug-delivery systems based on ligand-functionalized nanoparticles and antibody-drug conjugates-the tumor-targeted DSB is allowed selective accumulation at the tumor site, causing fewer off-target effects and lessening systemic toxicity while maximizing effectiveness. These approaches are geared toward utilizing nanotechnology to improve intracellular drug uptake and extend the circulation time to optimize antitumor efficacy. Overall, those advances in drug delivery systems could greatly boost the therapeutic efficacy of DSB by providing better bioavailability, controlled release, and targeted distribution. Such advances would increase treatment success in CML and Ph + ALL and expand DSB's potential clinical applications toward other malignancies. Research concerning the delivery of DSB with nanocarriers and ligand-mediated targeting strategies should bear further fruits to augment DSB therapy in oncology.
Collapse
Affiliation(s)
- Mahesha Keerikkadu
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Pragathi Devanand Bangera
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vamshi Krishna Tippavajhala
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mahalaxmi Rathnanand
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
4
|
Mohanty DL, Divya N, Zafar A, Warsi MH, Parida GR, Padhi P, Khalid M, Yasir M, Mujtaba MA. Development of etoricoxib-loaded mesoporous silica nanoparticles laden gel as vehicle for transdermal delivery: optimization, ex vivo permeation, histopathology, and in vivo anti-inflammatory study. Drug Dev Ind Pharm 2025; 51:506-521. [PMID: 40192336 DOI: 10.1080/03639045.2025.2490287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/22/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE Etoricoxib (ETB) is a nonsteroidal anti-inflammatory therapeutic agent. It is poorly soluble and has various gastrointestinal side effects such as bleeding and ulcers after oral administration. The present research aimed to develop an ETB-loaded mesoporous silica nanoparticle-laden gel (ETB-MSNPs) for transdermal delivery to improve therapeutic efficacy. METHODS The ETB-MSNPs were synthesized using a precipitation and solvent evaporation technique and their optimization was performed using a Box-Behnken design. The optimized ETB-MSNPs were incorporated into a carbopol-chitosan gel and evaluated for in vitro, ex vivo, and in vivo anti-inflammatory activity. RESULTS The ETB-MSNPs displayed nanosize of particles with nanosize distribution and high entrapment efficiency of ETB. The Fourier transform infrared spectroscopy (FTIR) and differential scanning calorimetry (DSC) studies showed that ETB was encapsulated in MSNPs. The optimized ETB-MSNPs were successfully integrated into the carbopol and chitosan gel, which exhibited excellent viscosity and spreadability. The optimized ETB-MSNPs gel exhibited a significantly higher and more sustained release of ETB compared to pure ETB gel. Optimized ETB-MSNPs gel exhibited a considerably higher anti-inflammatory effect with a significant reduction in IL-1β and TNF-α levels compared to pure ETB gel. The histopathological examination confirmed that optimized ETB-MSNPs gel did not exhibit any toxicity on the skin. CONCLUSION Based on the findings, the results suggest that the MSNPs gel has the potential as a carrier for enhancing the therapeutic efficacy of ETB through topical delivery, although further studies are needed to fully confirm its effectiveness.
Collapse
Affiliation(s)
- Dibya Lochan Mohanty
- Department of Pharmaceutics, School of Pharmacy, Centre for Nanomedicine, Anurag University, Hyderabad, India
| | - Noota Divya
- Department of Pharmaceutics, School of Pharmacy, Centre for Nanomedicine, Anurag University, Hyderabad, India
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Musarrat Husain Warsi
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Gnyana Ranjan Parida
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Priyanka Padhi
- Gayatri Institute of Science and Technology, Gunupur, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohd Yasir
- Department of Pharmacy, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Md Ali Mujtaba
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Arar, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
5
|
Mugundhan SL, Mohan M. Hyaluronic acid-coated capecitabine nanostructures for CD44 receptor-mediated targeting in breast cancer therapy. RSC Adv 2025; 15:12653-12670. [PMID: 40264886 PMCID: PMC12012621 DOI: 10.1039/d5ra01275a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
Hyaluronic acid-coated capecitabine-loaded nanomicelles (HA-CAP-M) are synthesized to overcome the challenges associated with capecitabine (CAP) conventional delivery such as low permeability and systemic toxicity. Nanomicelles containing saponin, glycerol, and vitamin-E TPGS formulation of capecitabine were further encapsulated with hyaluronic acid (HA) for CD44 receptor-mediated targeting. Optimization of the formulation was carried out using a Box-Behnken design resulting in 17.8 nm particle size, 89.3% entrapment efficiency and a biphasic drug release profile. Characterization studies validated stability, spherical structure, and desirable encapsulation characteristics of the nanomicelles. Lowered critical micelle concentration (CMC) and acceptable drug release kinetics revealed improved thermodynamic stability and controlled drug release, as predicted by the Hixson-Crowell model. HA-CAP-M showed much higher permeability and cytotoxicity than the free CAP, with an IC50 of 2.964 μg mL-1 in in vitro experiments. AO/PI staining also demonstrated dose-dependent apoptosis in MCF-7 breast cancer cells and validated the highly effective active targeting of HA. In addition, the formulation demonstrated good stability during storage and dilution conditions, confirming its stability as a drug delivery platform. In conclusion, HA-functionalized nanomicelles provide a biocompatible and efficient system for the targeted breast cancer therapy, enhancing the therapeutic efficacy of capecitabine.
Collapse
Affiliation(s)
- Sruthi Laakshmi Mugundhan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology Kattankulathur-603203 Chengalpattu Tamil Nadu India
| | - Mothilal Mohan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology Kattankulathur-603203 Chengalpattu Tamil Nadu India
| |
Collapse
|