Open Access Open Access  Restricted Access Subscription or Fee Access

Novel Nano Polymeric Lipid-Based Carriers for Emerging Pain Management of Rheumatoid Arthritis

Pankaj Kumar, Dhananjay kumar

Abstract


An inflammatory condition known as rheumatoid arthritis (RA) results in chronic joint inflammation, severe disability, and early death. RA is 3–5 times more common in women than in men, with a global frequency of roughly 0.3 percent–1%. The ultimate goal of RA treatment is to give symptomatic alleviation; there is no recognized cure for RA. NSAIDs are frequently used to treat RA, including indomethacin, diclofenac, ibuprofen, celecoxib, and specifies the characteristics. off-target effects are common with these powerful medications, and patient compliance suffers as a result. Additionally, non-steroidal anti-inflammatory medicines have a variety of formulation issues, including as poor bioavailability, gastrointestinal enzyme breakdown, food interactions, toxicity, and low solubility and permeability. To get over these roadblocks, researchers have turned to the topical route of drug delivery, which has higher patient compliance and avoids the first side effect that comes with traditional oral administration. Additionally, nanosized carriers such as exosomes, micro emulsifiers, nanocapsules, niosomes, solid lipid nanoparticles, and transferosomes have been built to assist drugs penetrate the skin's layers and reach the site of inflammation. These drug delivery methods are non-toxic, have a high drug encapsulation efficiency, and provide a long-term drug release. This review, which presents a landscape of topically applied Nano formulations for the symptomatic treatment of RA, discusses the impact of plan execution on the physicochemical characteristics of these niosomes in terms of particulate matter, charge density, drug - loading, and release of drug.


Keywords


Rheumatoid Arthritis, Novel Nano Polymeric Lipid-Based Carriers, Drug Delivery System.

Full Text:

PDF

References


Guo Q, Wang Y, Xu D, Nossent J, Pavlos NJ, Xu J. Rheumatoid arthritis: pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018; 6 (1), 1–14.

Global RA Network. About Arthritis and RA.[online] https://globalranetwork.org/project/disease-info/

Arthritis India. [online]. Available from https://www.arthritis-india.com/rheumatoid-arthritis.html.

Kraus VB. Editorial [Hot Topic: Waiting for Action on the Osteoarthritis Front (Guest Editors: Virginia Byers Kraus and Thomas Aigner)]. Current drug targets. 2010; 11 (5): 518–20.

McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. New England Journal of Medicine. 2011; 365 (23): 2205–19.

Schnitzer TJ, Truitt K, Fleischmann R, Dalgin P, Block J, Zeng Q, Bolognese J, Seidenberg B, et al. Phase II Rofecoxib Rheumatoid Arthritis Study Group. The safety profile, tolerability, and effective dose range of rofecoxib in the treatment of rheumatoid arthritis. Clinical therapeutics. 1999; 21 (10): 1688–702.

Krug H, Broadwell LK, Berry M, et al. Tolerability and efficacy of nabumetone and naproxen in the treatment of rheumatoid arthritis. Clinical therapeutics. 2000; 22 (1): 40–52.

Hoes JN, Jacobs JW, Buttgereit Fet al. Current view of glucocorticoid co-therapy with DMARDs in rheumatoid arthritis. Nature Reviews Rheumatology. 2010; 6 (12): 693–702.

Buttgereit F, Doering G, Schaeffler A, et al. Targeting pathophysiological rhythms: prednisone chronotherapy shows sustained efficacy in rheumatoid arthritis. Annals of the rheumatic diseases. 2010; 69 (7): 1275–80.

Smolen JS, Landewé R, Breedveld FC, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs. Annals of the rheumatic diseases. 2010; 69 (6): 964–75.

Verstappen SM, van Albada-Kuipers GA, Bijlsma JW, Blaauw AA, Utrecht Rheumatoid Arthritis Cohort Study Group (SRU). A good response to early DMARD treatment of patients with rheumatoid arthritis in the first year predicts remission during follow up. Ann Rheum Dis. 2005; 64 (1): 38–43.

Crofford LJ. Use of NSAIDs in treating patients with arthritis. Arthritis Res Ther. 2013; 15 Suppl 3(Suppl 3): S2.

Strehl C, van der Goes MC, Bijlsma JW, Jacobs JW, Buttgereit F. Glucocorticoid-targeted therapies for the treatment of rheumatoid arthritis. Expert Opin Investig Drugs. 2017; 26 (2):

–195.

Brown PM, Pratt AG, Isaacs JD. Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Nat Rev Rheumatol. 2016; 12 (12): 731–742.

Rein P, Mueller RB. Treatment with Biologicals in Rheumatoid Arthritis: An Overview. Rheumatol Ther. 2017; 4 (2): 247–261.

Baji P, Péntek M, Czirják L, Szekanecz Z, et al. Efficacy and safety of infliximab-biosimilar compared to other biological drugs in rheumatoid arthritis: a mixed treatment comparison. Eur J Health Econ. 2014; 15 Suppl 1(Suppl 1): S53–64.

Singh JA, Hossain A, Tanjong Ghogomu E, Mudano AS, et al. Biologics or tofacitinib for people with rheumatoid arthritis unsuccessfully treated with biologics: a systematic review and network meta-analysis. Cochrane Database Syst Rev. 2017; 3 (3): CD012591.

Soeken KL, Miller SA, Ernst E. Herbal medicines for the treatment of rheumatoid arthritis: a systematic review. Rheumatology. 2003; 42 (5): 652–9.

Ulbrich W, Lamprecht A. Targeted drug-delivery approaches by nanoparticulate carriers in the therapy of inflammatory diseases. Journal of The Royal Society Interface. 2010; 7(suppl_1): S55–66.

Prosperi, D.; Colombo, M.; Zanoni, I.; Granucci, F. Drug nanocarriers to treat autoimmunity and chronic inflammatory diseases. Semin. Immunol. 2017, 34, 61–67.

Serra P, Santamaria P. Nanoparticle-based autoimmune disease therapy. Clinical Immunology. 2015; 160 (1): 3–13.

Serra P, Santamaria P. Nanoparticle-based autoimmune disease therapy. Clinical Immunology. 2015; 160 (1): 3–13.

Kapoor B, Singh SK, Gulati M, Gupta R, Vaidya Y. Application of liposomes in treatment of rheumatoid arthritis: quo vadis. The scientific world Journal. 2014; 2014.

Yang M, Feng X, Ding J, et al. Nanotherapeutics relieve rheumatoid arthritis. Journal of Controlled Release. 2017; 252: 108–24.

Jain S, Doshi AS, Iyer AK, Amiji MM. Multifunctional nanoparticles for targeting cancer and inflammatory diseases. Journal of drug targeting. 2013; 21 (10): 888–903.

Metselaar JV, Van den Berg WB, Holthuysen AE, et al. Liposomal targeting of glucocorticoids to synovial lining cells strongly increases therapeutic benefit in collagen type II arthritis. Annals of the rheumatic diseases. 2004; 63 (4): 348–53.

Hofkens W, Storm G, Van Den Berg WB, Van Lent PL. Liposomal targeting of glucocorticoids to the inflamed synovium inhibits cartilage matrix destruction during murine antigen-induced arthritis. International journal of pharmaceutics. 2011; 416 (2): 486–92.

Moghimi SM, Hunter AC, Murray JC. Long-circulating and target-specific nanoparticles: theory to practice. Pharmacological reviews. 2001; 53 (2): 283–318.

Duncan R. The dawning era of polymer therapeutics. Nature reviews Drug discovery. 2003; 2 (5): 347–60.

Jung YS, Park W, Na K. Temperature-modulated noncovalent interaction controllable complex for the long-term delivery of etanercept to treat rheumatoid arthritis. Journal of Controlled Release. 2013; 171 (2): 143–51.

Ganta S, Devalapally H, Shahiwala A, Amiji M. A review of stimuli-responsive nanocarriers for drug and gene delivery. Journal of controlled release. 2008; 126 (3): 187–204.

Chandrasekar D, Sistla R, Ahmad FJ, et al. Folate coupled poly (ethyleneglycol) conjugates of anionic poly (amidoamine) dendrimer for inflammatory tissue specific drug delivery. Journal of Biomedical Materials Research Part A. 2007; 82 (1): 92–103.

Fernandes JC, Wang H, Jreyssaty C, et al. Bone-protective effects of nonviral gene therapy with Folate–Chitosan DNA nanoparticle containing Interleukin-1 receptor antagonist gene in rats with adjuvant-induced arthritis. Molecular Therapy. 2008 Jul 1; 16 (7): 1243–51.

Lee H, Lee MY, Bhang SH, et al., Hyaluronate–gold nanoparticle/tocilizumab complex for the treatment of rheumatoid arthritis. Acs Nano. 2014; 8 (5): 4790–8.

Zhou HF, Chan HW, Wickline SA, Lanza GM, Pham CT. αvβ3–Targeted nanotherapy suppresses inflammatory arthritis in mice. The FASEB Journal. 2009; 23 (9): 2978–85.

Barrera P, Blom A, Van Lent PL, et al. Synovial macrophage depletion with clodronate‐containing liposomes in rheumatoid arthritis. Arthritis & Rheumatism: Official Journal of the American College of Rheumatology. 2000; 4 3(9): 1951–9.

Chellat F, Merhi Y, Moreau A. Therapeutic potential of nanoparticulate systems for macrophage targeting. Biomaterials. 2005; 26 (35): 7260–75.

Ogale S, Hitraya E, Henk HJ. Patterns of biologic agent utilization among patients with rheumatoid arthritis: a retrospective cohort study. BMC musculoskeletal disorders. 2011; 12 (1): 1–1.

Goyal A, Kumar S, Nagpal M, Singh I, Arora S. Potential of novel drug delivery systems for herbal drugs. Indian journal of pharmaceutical education and research. 2011; 45 (3): 225–35.

Salim N, García-Celma MJ, Escribano E, et al. Formation of nanoemulsion containing ibuprofen by PIC method for topical delivery. Materials Today: Proceedings. 2018; 5: S172–9.

Mandawgade SD, Patravale VB. Development of SLNs from natural lipids: application to topical delivery of tretinoin. International journal of pharmaceutics. 2008; 363 (1–2): 132–8.

Liu W, Hu M, Liu W, et al., Investigation of the carbopol gel of solid lipid nanoparticles for the transdermal iontophoretic delivery of triamcinolone acetonide acetate. International journal of pharmaceutics. 2008; 364 (1): 135–41.

Naseri N, Valizadeh H, Zakeri-Milani P. Solid lipid nanoparticles and nanostructured lipid carriers: structure, preparation and application. Advanced pharmaceutical bulletin. 2015; 5 (3): 305.

Mishra V, Bansal KK, Verma A, et al. Solid lipid nanoparticles: Emerging colloidal nano drug delivery systems. Pharmaceutics. 2018; 10 (4): 191.

Maestrelli F, Bragagni M, Mura P. Advanced formulations for improving therapies with anti-inflammatory or anaesthetic drugs: A review. Journal of Drug Delivery Science and Technology. 2016; 32: 192–205.

Desai P, Patlolla RR, Singh M. Interaction of nanoparticles and cell-penetrating peptides with skin for transdermal drug delivery. Molecular membrane biology. 2010; 27 (7): 247–59.

Mukherjee S, Ray S, Thakur RS. Solid lipid nanoparticles: a modern formulation approach in drug delivery system. Indian journal of pharmaceutical sciences. 2009; 71 (4): 349.

Teeranachaideekul V, Boonme P, Souto EB, et al. Influence of oil content on physicochemical properties and skin distribution of Nile red-loaded NLC. Journal of controlled release. 2008; 128 (2): 134–41.

Jain SK, Chourasia MK, Masuriha R, et al. Solid lipid nanoparticles bearing flurbiprofen for transdermal delivery. Drug delivery. 2005; 12 (4): 207–15.

Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al.Liposome: classification, preparation, and applications. Nanoscale research letters. 2013; 8 (1): 1–9.

Gregoriadis G. Engineering liposomes for drug delivery: progress and problems. Trends in biotechnology. 1995; 13 (12): 527–37.

Vemuri S, Rhodes CT. Preparation and characterization of liposomes as therapeutic delivery systems: a review. Pharmaceutica Acta Helvetiae. 1995; 70 (2): 95–111.

Beroström K, Österberg E, Holmberg K, et al. Effects of branching and molecular weight of surface-bound poly (ethylene oxide) on protein rejection. Journal of Biomaterials Science, Polymer Edition. 1995; 6 (2): 123–32.

Wang M, Thanou M. Targeting nanoparticles to cancer. Pharmacological research. 2010; 62 (2): 90–9.

Garg A, Tisdale AW, Haidari E, Kokkoli E. Targeting colon cancer cells using PEGylated liposomes modified with a fibronectin-mimetic peptide. International journal of pharmaceutics. 2009; 366 (1–2): 201–10.

Meel RV, Vehmeijer LJ, Kok RJ, et al. Ligand-targeted particulate nanomedicines undergoing clinical evaluation: current status. Intracellular delivery III. 2016: 163–200.

Iwaszkiewicz KS, Hua S. Development of an effective topical liposomal formulation for localized analgesia and anti-inflammatory actions in the Complete Freund’s Adjuvant rodent model of acute inflammatory pain. Pain Physician. 2014;17(6):E719.

Trif M, Guillen C, Vaughan DM, et al. Liposomes as possible carriers for lactoferrin in the local treatment of inflammatory diseases. Experimental Biology and Medicine. 2001; 226 (6): 559–64.

Rajera R, Nagpal K, Singh SK, Mishra DN. Niosomes: a controlled and novel drug delivery system. Biological and Pharmaceutical Bulletin. 2011; 34 (7): 945–53.

Jamal M, Imam SS, Aqil M, et al. Transdermal potential and anti-arthritic efficacy of ursolic acid from niosomal gel systems. International immunopharmacology. 2015; 29 (2): 361–9.

Abidin L, Mujeeb M, Imam SS, et al. Enhanced transdermal delivery of luteolin via non-ionic surfactant-based vesicle: quality evaluation and anti-arthritic assessment. Drug delivery. 2016; 23 (3): 1069–74.

Garg NK, Tyagi RK, Singh B, et al. Nanostructured lipid carrier mediates effective delivery of methotrexate to induce apoptosis of rheumatoid arthritis via NF-κB and FOXO1. International journal of pharmaceutics. 2016; 499 (1–2): 301–20.

Gupta A, Eral HB, Hatton TA, et al. Nanoemulsions: formation, properties and applications. Soft matter. 2016; 12 (11): 2826–41.

Tang SY, Sivakumar M, Ng AM, Shridharan P. Anti-inflammatory and analgesic activity of novel oral aspirin-loaded nanoemulsion and nano multiple emulsion formulations generated using ultrasound cavitation. International journal of pharmaceutics. 2012; 430 (1–2): 299–306.

Salim N, Basri M, Rahman MB, et al. Modification of palm kernel oil esters nanoemulsions with hydrocolloid gum for enhanced topical delivery of ibuprofen. International journal of nanomedicine. 2012; 7: 4739.

Saberi AH, Fang Y, McClements DJ. Fabrication of vitamin E-enriched nanoemulsions: factors affecting particle size using spontaneous emulsification. Journal of colloid and interface science. 2013; 391: 95–102.

Abdulbaqi IM, Darwis Y, Khan NA, et al. Ethosomal nanocarriers: the impact of constituents and formulation techniques on ethosomal properties, in vivo studies, and clinical trials. International journal of nanomedicine. 2016; 11: 2279.

Kumar Sarwa K, Rudrapal M, Mazumder B. Topical ethosomal capsaicin attenuates edema and nociception in arthritic rats. Drug Delivery. 2015; 22 (8): 1043–52.

Fan C, Li X, Zhou Y. Enhanced topical delivery of tetrandrine by ethosomes for treatment of arthritis. BioMed research international. 2013; 2013.

Moura CC, Segundo MA, das Neves J. Co-association of methotrexate and SPIONs into anti-CD64 antibody-conjugated PLGA nanoparticles for theranostic application. International journal of nanomedicine. 2014; 9: 4911.

Mashaghi S, Jadidi T, Koenderink G, Mashaghi A. Lipid nanotechnology. International journal of molecular sciences. 2013; 14 (2): 4242–82.

Kesharwani D, Paliwal R, Satapathy T, Paul SD. Rheumatiod arthritis: An updated overview of latest therapy and drug delivery. Journal of pharmacopuncture. 2019; 22 (4): 210.

Coco R, Plapied L, Pourcelle V, Drug delivery to inflamed colon by nanoparticles: comparison of different strategies. International journal of pharmaceutics. 2013; 440 (1): 3–12. (1): 3–12.

Ortíz R, Quiñonero F, García-Pinel B, et al. Nanomedicine to overcome multidrug resistance mechanisms in colon and pancreatic cancer: recent progress. Cancers. 2021; 13 (9): 2058.

Chuang SY, Lin CH, Huang TH, Fang JY. Lipid-based nanoparticles as a potential delivery approach in the treatment of rheumatoid arthritis. Nanomaterials. 2018; 8 (1): 42.

Gulati M, Grover M, Singh S, Singh M. Lipophilic drug derivatives in liposomes. International journal of pharmaceutics. 1998; 165 (2): 129–68.

S. Venkataram, W.M. Awni, K. Jordan, and Y.E. Rahman, “Pharmacokinetics of two alternative dosage forms for cyclosporine: liposomes and intralipid,” Journal of Pharmaceutical Sciences. 1990; 79 (3): 216–219.


Refbacks

  • There are currently no refbacks.


Copyright (c) 2022 Research & Reviews: A Journal of Immunology