ABSTRACT
This review focuses on the advancements in liquid chromatographic techniques to do both qualitative and quantitative research on cyclosporine and gingerol, with particular emphasis on their incorporation into Novel Drug Delivery Systems (NDDS) to improve the management of Rheumatoid Arthritis (RA). The review underscores the importance of robust analytical methods for ensuring the precision, stability, and efficacy of these therapeutic agents in the context of RA. Despite the availability of conventional therapies, challenges such as poor bioavailability, rapid metabolism, and systemic toxicity persist. The combination of cyclosporine, a potent immunosuppressant, and gingerol, a natural anti-inflammatory compound, presents a promising approach to addressing these challenges and enhancing patient management in RA. Furthermore, this review explores the pathophysiology of RA, highlighting the need for specialised medication administration systems, such as nanocarriers, that optimize drug bioavailability and specifically target inflamed joints, thereby improving therapeutic outcomes. The role of liquid chromatography in the analysis and quality control of cyclosporine and gingerol is also discussed, with an emphasis on the development of analytical methods to support the advancement of NDDS for RA therapy.
INTRODUCTION
A chronic inflammatory and autoimmune disease, Rheumatoid Arthritis (RA) is typified by progressive joint damage due to the immune cells infiltrating the synovial tissue. This immune dysregulation leads to synovial inflammation, cartilage destruction, and bone erosion, eventually leading to joint deformities, reduced quality of life, and systemic complications such as cardiovascular and renal involvement (Rai et al., 2023). Effective management of RA aims to control inflammation, alleviate pain, and prevent progressive joint destruction. Current therapeutic approaches comprise corticosteroids, Disease-Modifying Anti-Rheumatic Drugs (DMARDs), and Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) (Tenti et al., 2023). Despite advancements, these therapies face limitations such as high costs, adverse effects due to non-specific immunosuppression, short drug half-lives, and incomplete disease control in many patients (Siddique, 2023).
Recent innovations in targeted drug delivery, such as polymeric micelles and lipid-based nanocarriers, show great promise for enhancing the effectiveness and stability of Rheumatoid Arthritis (RA) treatments (He et al., 2020). Cyclosporine, a potent immunosuppressive agent, has demonstrated efficacy in modulating immune responses and reducing inflammation in RA. Similarly, gingerol, a bioactive compound derived from ginger, has emerged as a natural anti-inflammatory agent capable of mitigating RA symptoms by inhibiting pro-inflammatory cytokines. Combining these therapeutic agents with advanced delivery systems can enhance bioavailability, improve stability, and enable site-specific delivery to inflamed joints, thereby minimizing adverse effects commonly associated with conventional treatments (Prasad et al., 2023).
Combining cyclosporine and gingerol offers a promising approach for RA treatment, leveraging their immunosuppressive and anti-inflammatory properties. Advanced drug delivery systems enable targeted delivery to inflamed joints, enhancing efficacy and reducing side effects (Meyer et al., 2007). Liquid chromatography ensures accurate and reproducible analysis. This study aims to develop an analytical method for the simultaneous evaluation of cyclosporine and gingerol in novel delivery systems, ensuring formulation precision, stability, and improved treatment outcomes.
Drug Delivery Systems in Treating Rheumatoid Arthritis
Rheumatoid Arthritis (RA) is a chronic autoimmune disorder that requires long-term management. Traditional treatments, such as DMARDs, corticosteroids, and NSAIDs, are used to control inflammation; however, they often prove inadequate due to systemic side effects, limited target specificity, and incomplete disease control. Although intra-articular injections can provide localized symptom relief, repeated use may increase the risk of infection, joint tissue damage, and reduced patient compliance (Dhule et al., 2023).
A key focus in Rheumatoid Arthritis (RA) management is the integration of drugs with potent anti-inflammatory properties into advanced delivery systems. Cyclosporine, an immunosuppressive agent, has demonstrated potential in alleviating inflammation and preventing joint damage by modulating various aspects of the immune response. Similarly, gingerol a bioactive compound extracted from ginger is gaining attention for its ability to inhibit inflammatory cytokines, thereby offering a complementary mechanism in RA therapy (Garcês et al., 2018). The refinement of drug delivery methods and analytical techniques contributes to the development of more effective and patient-centered approaches for managing RA (Thakur et al., 2018) (Figure 1).

Figure 1:
Pathophysiology of Rheumatoid Arthritis.
Integration of Gingerol and Cyclosporine in Rheumatoid Arthritis Management
6-Gingerol, found in ginger, has anti-inflammatory, antioxidant, and analgesic effects, making it a potential treatment for Rheumatoid Arthritis (RA) (Lehár et al., 2009). RA is a chronic autoimmune disease causing joint inflammation and degeneration (Szentesi et al., 2019). Conventional treatments often have side effects and limited long-term success (Meyer et al., 2007). Due to poor bioavailability and stability, gingerol’s clinical use is limited. Nanostructured Lipid Carriers (NLCs) improve its solubility, stability, and controlled release (Al-Ziyadi et al., 2024). Combining gingerol with cyclosporine may offer a synergistic approach to RA by addressing both inflammation and immune dysfunction.
Cyclosporine modulates T-cell activation and reduces pro-inflammatory cytokines, while gingerol complements this by alleviating oxidative stress and inflammation (Alsahli et al., 2021). Nanostructured Lipid Carriers (NLCs) improve the pharmacokinetics of compounds like gingerol and provide sustained release, maintaining consistent therapeutic levels (Rosli et al., 2024). This reduces dosing frequency and enhances patient compliance, which is a key factor in managing chronic conditions such as RA.
Furthermore, the lipid-based structure of NLCs enhances gingerol’s penetration into inflamed synovial tissues, allowing it to exert its anti-inflammatory effects more effectively (Shinde et al., 2021). Incorporating these advancements into RA therapy has the potential to significantly improve disease management by offering treatments that are safer, more effective, and better tolerated by patients. By addressing current therapeutic gaps, this multidisciplinary approach contributes to improved quality of life for individuals living with RA (Mucke et al., 2022).
Analytical Method Development for Cyclosporine and Gingerol in Rheumatoid Arthritis
This study presents the development and validation of an HPLC method for the simultaneous analysis of 6-, 8-, and 10-gingerols and 6-shogaol in ginger extracts. The method follows ICH guidelines, ensuring specificity, precision, accuracy, linearity, and appropriate detection and quantification limits (Raclariu et al., 2018). Advances in liquid chromatography now support multi-compound analysis, aiding comprehensive drug formulation assessments (Zhu et al., 2017). An online extraction technique was used, achieving complete analysis in 2.5 min with high efficiency and reliability.
Building on validated methods for the simultaneous quantification of immunosuppressive agents, the inclusion of both cyclosporine and gingerol in analytical techniques presents a novel strategy to enhance treatment approaches for Rheumatoid Arthritis (RA) (Zhang et al., 2021). Neha Desai et al., (2019) reported the development and validation of an RP-HPLC method for the simultaneous estimation of curcumin and cyclosporine in combined dosage formulations (Peng et al., 2016). In a similar context, the application of liquid chromatographic methods to analyze cyclosporine and gingerol may support the advancement of innovative drug delivery systems for RA, ensuring precision and reliability in pharmaceutical development.
Targeted Delivery of Cyclosporine (CsA)
Targeted delivery of Cyclosporine (CsA) offers a viable approach to overcoming the difficulties in administering systems, including toxicity, high therapeutic doses, and elevated treatment costs. CsA’s immunosuppressive properties is attributed to its selective inhibition of T lymphocytes, which predominantly reside within the lymphatic system (Park et al., 2016). CsA-containing lipid microspheres have been utilized to selectively transfer the medication to the lymphatic system in the thorax, achieving higher lymphatic concentrations while reducing systemic blood levels, thus improving its therapeutic index. Additionally, lipid-surfactant micelles and polylactic acid microspheres have shown potential for controlled release and targeted lymphatic delivery of CsA, enhancing the drug’s therapeutic efficacy and stability (Shah et al., 2006).
These advancements align with the creation of Innovative Medication Delivery Methods aimed at improving the pharmacokinetics and pharmacodynamics of CsA(Table 1). Combined with liquid chromatographic techniques, the qualitative and quantitative analysis of CsA formulations ensures precision, reproducibility, and optimized delivery for managing Rheumatoid Arthritis (RA) effectively (Guada, M., et al., 2016). This targeted approach minimizes the high dosages typically required for systemic administration, which, in turn, lowers treatment costs and reduces the risk of adverse effects like nephrotoxicity (Nation et al., 2019).
Category | Description |
---|---|
Challenges in CsA Delivery | Toxicity, high therapeutic doses, and elevated treatment costs. |
Mechanism of Action | Selective inhibition of T lymphocytes within the lymphatic system. |
Targeted Drug Delivery | Lipid microspheres selectively transfer CsA to the lymphatic system, reducing systemic blood levels and improving the therapeutic index. |
Innovative Drug Carriers | Lipid-surfactant micelles and polylactic acid microspheres enable controlled release and targeted lymphatic delivery of CsA. |
Pharmacokinetic and Pharmacodynamic Benefits | Enhanced therapeutic efficacy, stability, and reduced nephrotoxicity through controlled and sustained release. |
Role of Liquid Chromatographic Techniques | Ensures precision in qualitative and quantitative analysis of CsA formulations, optimizing drug delivery for Rheumatoid Arthritis (RA) management. |
Clinical Implications | Lower required dosages, reduced treatment costs, and minimized adverse effects. |
HPLC in Drug Analysis | Enables accurate measurement of drug concentrations, stability, and release profiles, ensuring effective formulation development. |
Lipid-surfactant micelles and polylactic acid microspheres enhance the stability, bioavailability, and sustained release of Cyclosporine (CsA), increasing its retention in lymphatic tissues and improving immunosuppressive effects. These systems support more effective RA management by targeting inflammation and immune dysregulation. High-Performance Liquid Chromatography (HPLC) plays a key role in accurately measuring drug concentration, stability, and release, essential for validating advanced CsA formulations (Wolska and Szymańska, 2023).
Cyclosporine Analysis
Cyclosporine has been effectively analyzed using advanced techniques such as LC-MS/MS and EMIT in human blood. Though EMIT shows a slight positive bias compared to LC-MS/MS, both methods are reliable and suitable for clinical monitoring, especially in transplant patients.
In Rheumatoid Arthritis (RA), Cyclosporine A (CyA) was successfully encapsulated into PSA-PCL micelles, achieving a loading efficiency of 29.3% and a drug loading capacity of 0.09 mg/mg. Encapsulation increased micelle size at both 25ºC and 37ºC, making them suitable for targeting inflamed tissues with leaky vasculature.
The micelles were synthesized using colominic acid Sodium Salt (PSA) and Polycaprolactone (PCL), with amide bonds formed through controlled polymerization. PSA improved the pharmacokinetics of CyA, enhancing its stability and bioavailability. These micelles offer a promising strategy for RA treatment by enabling targeted delivery and minimizing systemic side effects. Integration with HPLC techniques ensures accurate drug quantification and formulation validation (Ghazanfari and Sepehrnia, 2024).
Pathogenesis of Rheumatoid Arthritis (RA)
Rheumatoid Arthritis (RA) is a chronic autoimmune disease that primarily targets synovial joints, leading to inflammation, joint destruction, and reduced function. Its complex pathophysiology involves both genetic and environmental factors. The disease is marked by an autoimmune response in which the immune system attacks the synovium, causing synovitis and eventually cartilage and bone damage. Autoantibodies such as Rheumatoid Factor (RF) and Anti-Citrullinated Protein Antibodies (ACPA) contribute to this process by forming immune complexes that activate inflammatory cells (Prasad et al., 2023).
Infiltration of T cells, B cells, and macrophages into the synovial fluid promotes the release of pro-inflammatory cytokines like interleukins and TNF-α, driving further tissue damage. Hormonal factors, especially estrogen fluctuations in women, also influence RA onset and severity. If untreated, RA can result in joint deformities, disability, and systemic complications. Early diagnosis and advanced therapies, including biologics and DMARDs, are essential for controlling inflammation and preserving joint function (Perumal et al., 2024; Panichi et al., 2024; Pisetsky, 2023).
Current Treatment Strategy for RA Therapy
Rheumatoid Arthritis (RA) treatment is increasingly guided by personalized medicine due to the disease’s complexity and variable responses. Biomarkers such as CRP, ACPA, and RF help tailor therapies, while genetic markers like HLA-DR4 predict disease severity and drug response (Dedmon, 2020). Combining pharmacologic and non-pharmacologic strategies improves symptom control, function, and quality of life (Reimold and Chandran, 2014).
These systems allow for the co-delivery of drugs like cyclosporine and gingerol, providing a dual-action strategy that reduces inflammation and modulates immune activity. Simultaneously, advances in regenerative medicine such as mesenchymal stem cell therapies are being investigated to repair joint damage and potentially reverse disease progression. As research evolves, wearable technologies offer new opportunities for real-time monitoring of disease activity (Guk et al., 2019). These tools enable patients and clinicians to better track progression, tailor treatment, and support adherence, fostering a more personalized and outcomes-driven approach to RA care.
Patents Related to NDDS for RA Therapy
Patents pertaining to Rheumatoid Arthritis (RA) treatment using NDDS highlight the growing interest in innovative therapeutic approaches that minimise adverse effects while increasing medication efficacy. Nano-sized carrier systems, such as Solid Lipid Nanoparticles (SLNs), gold Nanoparticles (AuNPs), chitosan nanoparticles, polymeric nanoparticles, Liposomes (LPs), Nanoemulsions (NEs), nanomicelles, and nanocapsules, are being explored for their ability to enhance the targeted administration and bioavailability of anti-inflammatory and immunosuppressive drugs (Syed and Devi, 2019).
These systems can encapsulate drugs like tacrolimus, Dexamethasone (DEX), Curcumin (CUR), Methotrexate (MTX), and Celecoxib (CEL), enabling sustained release and targeted action at inflamed sites. Studies show that NDDS reduce systemic toxicity, improve therapeutic efficacy, and offer controlled release-making them strong candidates for RA treatment. Recent patents also aim to address challenges like poor solubility and rapid metabolism, supporting better patient adherence and long-term disease control (Mavlyanov and Bekenova, 2024).
Challenges and Opportunities in RA Treatment
Technological advances have significantly improved the detection and treatment of Rheumatoid Arthritis (RA). Imaging tools like ultrasound and MRI enable early and accurate assessment of joint damage, while targeted therapies such as small molecule inhibitors and biologics offer more personalized care. Although a permanent cure remains unavailable (Mueller et al., 2021), emerging approaches like precision medicine, advanced drug delivery systems, and regenerative therapies hold promise for more effective and lasting RA management (Alzoubi et al., 2023).
The focus on precision medicine, innovative drug delivery systems, and regenerative therapies is opening new avenues for managing Rheumatoid Arthritis (RA) more effectively. Utilising genetic, genomic, and clinical data, precision medicine customises therapy for each patient, improving therapeutic outcomes while minimizing side effects (Figure 2). Advances in biomarkers and genetic profiling enable clinicians to identify patients who may respond best to specific therapies, ensuring a more targeted approach to disease management (Kalia, 2015). Together, these approaches reflect a shift toward a more holistic and patient-centric model of care. While significant challenges remain, these advancements provide a hopeful outlook for improving the quality of life and long-term outcomes for RA patients (Taylor et al., 2016).

Figure 2:
Gingerol and Cyclosporine-Loaded Nanostructured Lipid Carrier (NLC) for Targeted Rheumatoid Arthritis Therapy.
Developments in Cyclosporine Quantification Analytical Techniques for Therapeutic Drug Monitoring
Several studies have focused on developing and validating analytical methods for quantifying Cyclosporine (CSA) and other immunosuppressants using chromatographic techniques such as LC-MS/MS, HPLC, and UPLC. These methods aim to ensure accurate monitoring and effective therapeutic drug management. One such approach involves a simplified LC-MS/MS technique for detecting CSA in whole blood, using CSA-d12 as an internal standard. This method employs a modified one-step protein precipitation process and a C18 column for chromatographic separation, achieving a rapid total run time of 4.3 min (Stoll et al., 2006).
Studies have shown that hematocrit levels do not affect Cyclosporine (CSA) measurements, confirming the reliability of certain LC-MS/MS methods for therapeutic monitoring (Gavala and Myrianthefs, 2017). The use of isotope-labeled internal standards enhances the accuracy of immunosuppressant quantification, including CSA, tacrolimus, sirolimus, and everolimus (Shipkova and Svinarov, 2016). Validated UPLC-MS/MS methods enable simultaneous monitoring of these drugs with high precision, while HPLC techniques effectively assess CSA release from nanoparticles, showing strong accuracy and linearity.
An isocratic HPLC method was developed and validated for analyzing Cyclosporine (CSA) dissolution samples, offering accurate and reliable results. Immunoassays revealed inconsistencies in CSA concentrations, underscoring the value of HPLC in therapeutic monitoring (Fang et al., 2024). These advancements support improved drug quantification, guiding clinical decisions and drug development efforts (Taddeo et al., 2020).
CONCLUSION
The integration of advanced liquid chromatographic techniques in the analysis of cyclosporine and gingerol is pivotal for the creation of innovative rheumatoid arthritis medication delivery methods management. These methods not only ensure accurate qualitative and quantitative assessments but also facilitate the optimization of therapeutic strategies that address the limitations of traditional treatments. The synergistic effects of combining cyclosporine and gingerol, supported by robust analytical methodologies, hold significant potential for improving patient outcomes in RA. Future studies should keep concentrating on honing these analytical methods and investigating their applications in clinical settings to improve the therapeutic effectiveness and security of RA therapies.
ACKNOWLEDGEMENT
I would like to express my sincere gratitude to the research community whose invaluable contributions for this review article.
CONFLICT OF INTEREST
The authors declare that there is no conflict of interest.
ABBREVIATIONS
NDDS: Novel Drug Delivery Systems; RA: Rheumatoid Arthritis; DMARDs: Disease-Modifying Anti-Rheumatic Medications; NSAIDs: Non-Steroidal Anti-Inflammatory Drugs; CsA: Cyclosporine; ACPA: Anti-Citrullinated Protein Antibodies; TNF: Tumour Necrosis Factor; CRP: C-Reactive Protein; SLNs: Solid Lipid Nanoparticles; DEX: Dexamethasone; CUR: Curcumin; MTX: Methotrexate; CEL: Celecoxib; Ht: Hematocrit.
Cite this article:
Priyanka K, Rashmi NG. Liquid Chromatographic Techniques for the Qualitative and Quantitative Analysis of Cyclosporine and Gingerol: Advances in Rheumatoid Arthritis Management. J Young Pharm. 2025;17(3):504-510.
REFERENCES
- Alsahli, M. A., Almatroodi, S. A., Almatroudi, A., Khan, A. A., Anwar, S., Almutary, A. G., Alrumaihi, F., & Rahmani, A. H. (2021). 6‐gingerol, a major ingredient of ginger attenuates diethylnitrosamine‐induced liver injury in rats through the modulation of oxidative stress and anti‐inflammatory activity. Mediators of Inflammation, 2021(1), Article 6661937. https://doi.org/10.1155/2021/6661937
- Al-Ziyadi, R. K. M., Hayati, N., Rezaei, M. R., & Es-haghi, A. (2024). Preparation and characterization of chitosan-coated nanostructured lipid carriers (CS-NLC) containing (6)-gingerol and investigating their toxicity against MCF-7 breast cancer cell line. BioNanoScience, 14(1), 153-163. https://doi.org/10.1007/s12668-023-01261-4
- Alzoubi, L., Aljabali, A. A. A., & Tambuwala, M. M. (2023). Empowering precision medicine: The impact of 3D printing on personalized therapeutic. AAPS PharmSciTech, 24(8), 228. https://doi.org/10.1208/s12249-023-02682-w
- Ayshah Rosli, N., Hasham, R., Abdul Aziz, A., Ubaidah Noh, T., & Jemon, K. (2024). Nanostructured lipid carrier loaded with Zingiber officinale oil to enhance transdermal bioactive delivery for topical formulation. Microchemical Journal, 200, Article 110470. https://doi.org/10.1016/j.microc.2024.110470
- Dedmon, L. E. (2020). The genetics of rheumatoid arthritis. Rheumatology, 59(10), 2661-2670. https://doi.org/10.1093/rheumatology/keaa232
- Dhule, K. D., & Nandgude, T. D. (2023). Lipid nano-system based topical drug delivery for management of rheumatoid arthritis: An overview. Advanced Pharmaceutical Bulletin, 13(4), 663-677. https://doi.org/10.34172/apb.2023.075
- Fang, Z., Zhang, H., Guo, J., & Guo, J. (2024). Overview of therapeutic drug monitoring and clinical practice. Talanta, 266(1), Article 124996. https://doi.org/10.1016/j.talanta.2023.124996
- Garcês, A., Amaral, M. H., Sousa Lobo, J. M., & Silva, A. C. (2018). Formulations based on solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for cutaneous use: A review. European Journal of Pharmaceutical Sciences, 112, 159-167. https://doi.org/10.1016/j.ejps.2017.11.023
- Gavala, A., & Myrianthefs, P. (2017). Comparison of point-of-care versus central laboratory measurement of hematocrit, hemoglobin, and electrolyte concentrations. Heart and Lung, 46(4), 246-250. https://doi.org/10.1016/j.hrtlng.2017.04.003
- Ghazanfari, M. R., & Sepehrnia, S. (2024). Reviewing advances in rheumatoid arthritis treatment: From disease-modifying antirheumatic drugs to innovative drug delivery systems. Immunoregulation, 6(2), 105-118. https://doi.org/10.32598/Immunoregulation.6.2.4
- Guada, M., Beloqui, A., Kumar, M. N. V. R., Préat, V., Dios-Viéitez, M., & Blanco-Prieto, M. J. (2016). Reformulating cyclosporine A (CsA): More than just a life cycle management strategy. Journal of Controlled Release, 225, 269-282. https://doi.org/10.1016/j.jconrel.2016.01.056
- Guk, K., Han, G., Lim, J., Jeong, K., Kang, T., Lim, E.-K., & Jung, J. (2019). Evolution of wearable devices with real-time disease monitoring for personalized healthcare. Nanomaterials, 9(6), 813. https://doi.org/10.3390/nano9060813
- He, F., Wen, N., Xiao, D., Yan, J., Xiong, H., Cai, S., Liu, Z., & Liu, Y. (2020). Aptamer-based targeted drug delivery systems: Current potential and challenges. Current Medicinal Chemistry, 27(13), 2189-2219. https://doi.org/10.2174/0929867325666181008142831
- Kalia, M. (2015). Biomarkers for personalized oncology: Recent advances and future challenges. Metabolism: Clinical and Experimental, 64(3) (Suppl. 1), S16-S21. https://doi.org/10.1016/j.metabol.2014.10.027
- Lehár, J., Krueger, A. S., Avery, W., Heilbut, A. M., Johansen, L. M., Price, E. R., Rickles, R. J., Short, G. F., Staunton, J. E., Jin, X., Lee, M. S., Zimmermann, G. R., & Borisy, A. A. (2009). Synergistic drug combinations tend to improve therapeutically relevant selectivity. Nature Biotechnology, 27(7), 659-666. https://doi.org/10.1038/nbt.1549
- Mavlyanov, I. R., & Bekenova, G. T. (2024). Mavlyanov SI modern approaches to the treatment of rheumatoid arthritis: Issues of low efficacy of pharmacotherapy from the point of view of treatment adherence. International Journal of Integrative and Modern Medicine, 2(6), 120-129. https://medicaljournals.eu/index.php/IJIMM/article/view/521/468
- Meyer, O., de Bandt, M., Berthelot, J.-M., Cantagrel, A., Combe, B., Fautrel, B., Flipo, R.-M., Lioté, F., Maillefert, J.-F., Saraux, A., Wendling, D., Guillemin, F., Le Loët, X., & Working Group for Therapeutic Strategies for Rheumatoid Arthritis. (2007). Clinical practice format for choosing a second-line disease modifying anti-rheumatic drug in early rheumatoid arthritis after failure of 6 months’ first-line DMARD therapy. Joint Bone Spine, 74(1), 73-78. https://doi.org/10.1016/j.jbspin.2006.05.008
- Mucke, J., Krusche, M., & Burmester, G. R. (2022). A broad look into the future of rheumatoid arthritis. Therapeutic Advances in Musculoskeletal Disease, 14, Article 1759720X221076211. https://doi.org/10.1177/1759720X221076211
- Mueller, A.-L., Payandeh, Z., Mohammadkhani, N., Mubarak, S. M. H., Zakeri, A., Alagheband Bahrami, A., Brockmueller, A., & Shakibaei, M. (2021). Recent advances in understanding the pathogenesis of rheumatoid arthritis: New treatment strategies. Cells, 10(11), 3017. https://doi.org/10.3390/cells10113017
- Nation, R. L., Rigatto, M. H. P., Falci, D. R., & Zavascki, A. P. (2019). Polymyxin acute kidney injury: Dosing and other strategies to reduce toxicity. Antibiotics, 8(1), 24. https://doi.org/10.3390/antibiotics8010024
- Panichi, V., Costantini, S., Grasso, M., Arciola, C. R., & Dolzani, P. (2024). Innate immunity and synovitis: Key players in osteoarthritis progression. International Journal of Molecular Sciences, 25(22), Article 12082. https://doi.org/10.3390/ijms252212082
- Park, Y.-H., Min, K. A., Song, Y.-K., Ham, S., & Kim, C.-K. (2016). Chemically conjugated novel liposomal formulation for intravenous delivery of cyclosporin A. Colloids and Surfaces. Part A, Physicochemical and Engineering Aspects, 495, 229-237. https://doi.org/10.1016/j.colsurfa.2016.02.008
- Peng, C., Cheng, J., & Cheng, Q. (2017). A supervised learning model for high-dimensional and large-scale data. ACM Transactions on Intelligent Systems and Technology, 8(2), 1-23. https://doi.org/10.1145/2972957
- Perumal, S., Mayilsamy, S., & Thangaraj, S. (2024). Rheumatological perspective of osteoarthritis and their common clinical presentations from patients who are attending teaching hospital. Naturalista Campano, 28(1), 1999-2025. https://www.museonaturalistico.it/index.php/journal/article/view/379/300
- Pisetsky, D. S. (2023). Pathogenesis of autoimmune disease. Nature Reviews. Nephrology, 19(8), 509-524. https://doi.org/10.1038/s41581-023-00720-1
- Prasad, P., Verma, S., Surbhi, Ganguly, N. K., Chaturvedi, V., & Mittal, S. A. (2023). Rheumatoid arthritis: Advances in treatment strategies. Molecular and Cellular Biochemistry, 478(1), 69-88. https://doi.org/10.1007/s11010-022-04492-3
- Raclariu, A. C., Heinrich, M., Ichim, M. C., & de Boer, H. (2018). Benefits and limitations of DNA barcoding and metabarcoding in herbal product authentication. Phytochemical Analysis, 29(2), 123-128. https://doi.org/10.1002/pca.2732
- Rai, V., Patel, N., Mammen, S. R., Chaudhary, S. M., Arshad, S., Munazzam, S. W., & Munazzam, S. W. (2023). Futuristic novel therapeutic approaches in the treatment of rheumatoid arthritis. Cureus, 15(11), Article e49738. https://doi.org/10.7759/cureus.49738
- Reimold, A. M., & Chandran, V. (2014). Nonpharmacologic therapies in spondyloarthritis. Best Practice and Research. Clinical Rheumatology, 28(5), 779–792. https://doi.org/10.1016/j.berh.2014.10.003
- Shah, N. M., Parikh, J., Namdeo, A., Subramanian, N., & Bhowmick, S. (2006). Preparation, characterization and in vivo studies of proliposomes containing cyclosporine A. Journal of Nanoscience and Nanotechnology, 6(9-10), 2967-2973. https://doi.org/10.1166/jnn.2006.403
- Shinde, C., Venkatesh, M. P., Pramod Kumar, T., & Pai, D. R. (2021). Nanostructured lipid carrier-based smart gel: A delivery platform for intra-articular therapeutics. Autoimmunity, 54(1), 35-44. https://doi.org/10.1080/08916934.2020.1846184
- Shipkova, M., & Svinarov, D. (2016). LC-MS/MS as a tool for TDM services: Where are we? Clinical Biochemistry, 49(13-14), 1009-1023. https://doi.org/10.1016/j.clinbiochem.2016.05.001
- Siddique, I. (2023). High-performance liquid chromatography: Comprehensive techniques and cutting-edge innovations. European Journal of Advances in Engineering and Technology, 10(9), 66-70. https://doi.org/10.2139/ssrn.4885931
- Stoll, D. R., Cohen, J. D., & Carr, P. W. (2006). Fast, comprehensive online two-dimensional high performance liquid chromatography through the use of high temperature ultra-fast gradient elution reversed-phase liquid chromatography. Journal of Chromatography. A, 1122(1-2), 123-137. https://doi.org/10.1016/j.chroma.2006.04.058
- Syed, A., & Devi, V. K. (2019). Potential of targeted drug delivery systems in treatment of rheumatoid arthritis. Journal of Drug Delivery Science and Technology, 53, Article 101217. https://doi.org/10.1016/j.jddst.2019.101217
- Szentesi, M., Nagy, Z., Mangel, Z. K., & Géher, P. (2019). AB0447 biological therapy and radiosynoviorthesis in patients with rheumatoid arthritis and psoriatic arthritis. Annals of the Rheumatic Diseases, 78, 1686-1692. https://doi.org/10.1136/annrheumdis-2019-eular.3071
- Taddeo, A., Prim, D., Bojescu, E.-D., Segura, J.-M., & Pfeifer, M. E. (2020). Point-of-care therapeutic drug monitoring for precision dosing of immunosuppressive drugs. The Journal of Applied Laboratory Medicine, 5(4), 738-761. https://doi.org/10.1093/jalm/jfaa067
- Taylor, P. C., Moore, A., Vasilescu, R., Alvir, J., & Tarallo, M. (2016). A structured literature review of the burden of illness and unmet needs in patients with rheumatoid arthritis: A current perspective. Rheumatology International, 36(5), 685-695. https://doi.org/10.1007/s00296-015-3415-x
- Tenti, S., Bruyère, O., Cheleschi, S., Reginster, J.-Y., Veronese, N., & Fioravanti, A. (2023). An update on the use of conventional and biological disease-modifying anti-rheumatic drugs in hand osteoarthritis. Therapeutic Advances in Musculoskeletal Disease, 15, Article 1759720X231158618. https://doi.org/10.1177/1759720X231158618
- Thakur, S., Riyaz, B., Patil, A., Kaur, A., Kapoor, B., & Mishra, V. (2018). Novel drug delivery systems for NSAIDs in management of rheumatoid arthritis: An overview. Biomedicine and Pharmacotherapy, 106, 1011-1023. https://doi.org/10.1016/j.biopha.2018.07.027
- Wolska, E., & Szymańska, M. (2023). Comparison of the in vitro drug release methods for the selection of test conditions to characterize solid lipid microparticles. Pharmaceutics, 15(2), 511. https://doi.org/10.3390/pharmaceutics15020511
- Zhang, C., Wang, N., Xu, Y., Tan, H.-Y., & Feng, Y. (2021). Identification of key contributive compounds in a herbal medicine: A novel mathematic biological evaluation approach. Advanced Theory and Simulations, 4(6), Article 2000279. https://doi.org/10.1002/adts.202000279
- Zhu, B., & Chen, Y. Y. (2017). Development and application of liquid chromatography in life sciences. Journal of Chromatography and Separation Techniques, 8(2), 1-4. https://doi.org/10.4172/2157-7064.1000358.