Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Case Report
Case Series
Editorial
Editorial I
Editorial II
Original Article
Review
Review Article
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Case Report
Case Series
Editorial
Editorial I
Editorial II
Original Article
Review
Review Article
View/Download PDF

Translate this page into:

Editorial
18 (
3
); 1-3

Superoxide dismutase: Challenges, opportunities, and promises for clinical translation

Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia

Address for correspondence: Dr. Zafar Rasheed, Department of Pathology, College of Medicine, Qassim University, Qassim, Saudi Arabia. E-mail: zafarrasheed@qu.edu.sa

Licence

This is an open-access article distributed under the terms of the Creative Commons Attribution-Noncommercial-Share Alike 3.0 Unported, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Disclaimer:
This article was originally published by Qassim Uninversity and was migrated to Scientific Scholar after the change of Publisher.

Superoxide dismutase (SOD) is a crucial enzyme in the antioxidant defense system, playing a pivotal role in scavenging superoxide radicals and maintaining cellular redox balance. Its therapeutic potential has garnered significant attention in various fields, including medicine, biotechnology, and cosmetology. However, despite promising preclinical evidence, the clinical translation of SOD-based therapies faces numerous challenges, including issues related to formulation, delivery, pharmacokinetics, and efficacy. This editorial article critically examines the current status of SOD-based therapeutics, identifies key challenges hindering their clinical translation, and explores potential strategies to overcome these obstacles. By addressing these challenges and capitalizing on emerging opportunities and promises, the full therapeutic potential of SOD may cover the ways for the development of effective antioxidant therapies for a wide range of human diseases.

SOD is an essential enzyme that catalyzes the dismutation of superoxide radicals into oxygen and hydrogen peroxide, thus playing a crucial role in the cellular antioxidant defense system.[1,2] Clinical studies investigating the efficacy of SOD-based therapies in neurodegenerative disorders are underway, with promising preliminary results indicating the potential benefits of enhancing antioxidant defense mechanisms in slowing disease progression and improving patient outcomes.[3] SOD has also been shown to exert protective effects on the cardiovascular system by scavenging ROS, reducing endothelial dysfunction, and preserving vascular integrity.[4] In preclinical studies, overexpression of SOD has been demonstrated to attenuate oxidative stress and vascular inflammation, thereby preventing the development of atherosclerotic lesions and reducing the risk of cardiovascular events.[4] Moreover, clinical trials evaluating the therapeutic efficacy of SOD-based therapies, such as gene therapy and SOD mimetics, in patients with cardiovascular diseases have shown promising results, including improvements in endothelial function, myocardial perfusion, and overall cardiac function.[4] These findings underscore the potential of SOD as a novel therapeutic strategy for mitigating cardiovascular risk and improving outcomes in patients with heart disease. Furthermore, SOD has also emerged as a potential target for cancer therapy, given its role in modulating oxidative stress and regulating signaling pathways implicated in cell proliferation, apoptosis, and angiogenesis.[5] Several studies have demonstrated that SOD overexpression or supplementation can inhibit tumor growth and sensitize cancer cells to chemotherapy and radiation therapy.[6] In addition, SOD-based therapies have been explored as adjuvant treatments for cancer patients undergoing conventional anticancer therapies, with the aim of reducing treatment-related toxicity and enhancing therapeutic efficacy.[7-10] Preclinical trials investigating the combination of SOD mimetics with chemotherapy or radiotherapy have shown promising results, including improvements in treatment response rates and cancer survival outcomes.[3,7-10] However, further research is needed to optimize the dosing regimens and treatment strategies for maximizing the therapeutic benefits of SOD in cancer patients.[7] Aging is associated with a progressive decline in antioxidant defenses and an accumulation of oxidative damage, leading to age-related degenerative disorders, such as osteoporosis, sarcopenia, and frailty.[11] SOD has been proposed as a potential therapeutic target for attenuating age-related decline and promoting healthy aging by counteracting oxidative stress and preserving cellular function.[3,12,13] Preclinical studies have demonstrated that CuZn-SOD and catalase significantly increase the maximum life span, suggesting SOD with catalase potential as an anti-aging intervention.[13] Furthermore, clinical trials investigating the effects of SOD-based therapies on age-related disorders, such as osteoarthritis and frailty, are ongoing, with preliminary data suggesting beneficial effects on disease progression and functional outcomes.[14] These findings highlight the potential of SOD as a promising therapeutic strategy for promoting health-related benefits and preventing disease-related morbidities.

Despite the promising preclinical evidence supporting the therapeutic potential of SOD, the clinical translation of SOD-based therapies faces numerous challenges that have hindered their widespread adoption in clinical practice. One of the major challenges in the clinical development of SOD-based therapies is the formulation and stability of SOD formulations. SOD is a large, complex protein molecule that is susceptible to degradation and denaturation under physiological conditions.[2,15] Moreover, SOD has poor oral bioavailability and limited tissue penetration, further complicating its formulation and delivery.[4,15] Various strategies have been explored to improve the formulation and stability of SOD, including encapsulation in liposomes, nanoparticles, and other delivery vehicles.[16-18] These formulations can protect SOD from enzymatic degradation and enhance its pharmacokinetic profile, thereby improving its therapeutic efficacy and tissue distribution. However, further research is needed to optimize the formulation and delivery of SOD-based therapies for clinical use. Another major challenge in the clinical translation of SOD-based therapies is the pharmacokinetics and tissue distribution of SOD formulations.[19] SOD has a short half-life in circulation and tends to accumulate in the liver and other organs, limiting its availability at target sites.[20] Moreover, the biodistribution of SOD can be influenced by factors such as route of administration, formulation, and patient-specific factors, further complicating its pharmacokinetic profile.[15] To overcome these challenges, efforts have been made to optimize the pharmacokinetics and tissue distribution of SOD formulations through the use of targeted delivery strategies, such as receptor-mediated targeting and tissue-specific accumulation.[21,22] Moreover, the development of novel SOD mimetics and analogs with improved pharmacokinetic properties holds promise for enhancing the therapeutic efficacy and tissue distribution of SOD-based therapies.[23] Another major challenge is to ensure the efficacy and safety of SOD-based therapies for their clinical translation. While preclinical studies have demonstrated the therapeutic potential of SOD in various disease models, the translation of these findings to the clinic has been challenging due to issues related to dosing, treatment duration, and patient selection.[15] Moreover, concerns have been raised about the potential side effects of SOD, including immunogenicity, off-target effects, and long-term toxicity.[24,25] To address these concerns, rigorous preclinical and clinical studies are needed to evaluate the efficacy and safety of SOD-based therapies in well-defined patient populations with appropriate outcome measures. Moreover, the development of predictive biomarkers and surrogate endpoints can facilitate the assessment of treatment response and optimize dosing regimens for maximizing therapeutic benefits while minimizing adverse effects.

Despite the challenges associated with the clinical translation of SOD-based therapies, several opportunities and promises exist for advancing their development and enhancing their therapeutic efficacy. One promising approach for enhancing the clinical translation of SOD-based therapies is the development of targeted delivery strategies that enable site-specific accumulation and controlled release of SOD formulations.[15-20] Targeted delivery systems, such as liposomes, nanoparticles, and hydrogels, can improve the pharmacokinetics and tissue distribution of SOD, thereby enhancing its therapeutic efficacy while minimizing off-target effects.[15-20] Moreover, the use of ligand-targeted nanoparticles and antibody-conjugated SOD formulations can further enhance the specificity and selectivity of SOD delivery to diseased tissues, such as tumor microenvironments or inflamed tissues.[22,26] By capitalizing on targeted delivery strategies, we can maximize the therapeutic benefits of SOD-based therapies and overcome the limitations associated with non-specific distribution and off-target effects. Another promising approach for enhancing the clinical translation of SOD-based therapies is the development of novel SOD analogs and mimetics with improved pharmacokinetic properties and enhanced therapeutic efficacy.[22,23] By optimizing the structure and stability of SOD molecules, we can design analogs that exhibit prolonged circulation time, enhanced tissue penetration, and increased resistance to degradation, thereby improving their bioavailability and therapeutic potential. Moreover, the use of rational drug design and computational modeling techniques including artificial intelligence can facilitate the identification of novel SOD mimetics with superior enzymatic activity and antioxidant properties. By harnessing the power of structure-based drug design and high-throughput screening approaches, we can accelerate the discovery and development of next-generation SOD analogs for clinical use. Moreover, finally, combinatorial approaches that leverage the synergistic interactions between SOD and other antioxidant compounds or therapeutic modalities represent another promising strategy for enhancing the clinical translation of SOD-based therapies.[27,28] Preclinical studies have suggested that combining SOD with natural antioxidants, such as Vitamins C and E, or pharmacological agents targeting oxidative stress and inflammation pathways can enhance the therapeutic efficacy of SOD and improve treatment outcomes.[27-29] Moreover, the combination of SOD with conventional anticancer therapies, such as chemotherapy or radiotherapy, has shown promise in preclinical models and early-phase clinical trials, with synergistic effects on tumor growth inhibition and treatment response rates.[30,31] By exploring the potential synergies between SOD and other therapeutic agents, combinatorial treatment strategies surely develop that enhance the efficacy of SOD-based therapies and overcome resistance mechanisms associated with monotherapy approaches. In conclusion, SOD holds tremendous promise as a multifaceted therapeutic agent with diverse applications across various fields, including medicine, biotechnology, and cosmetology. However, the clinical translation of SOD-based therapies faces numerous challenges, including issues related to formulation, delivery, pharmacokinetics, and efficacy. By addressing these challenges and capitalizing on emerging opportunities, we can overcome the obstacles hindering the clinical development of SOD-based therapies and harness the full therapeutic potential of SOD for improving human health and well-being.

Competing Interests

The author declares no competing interests.

References

  1. , , , , , , . Superoxide dismutase:An updated review on its health benefits and industrial applications. Crit Rev Food Sci Nutr. 2022;62:7282-300.
    [Google Scholar]
  2. , . Superoxide radical and superoxide dismutases. Annu Rev Biochem. 1995;64:97-112.
    [Google Scholar]
  3. , . Therapeutic potentials of superoxide dismutase. Int J Health Sci (Qassim). 2018;12:88-93.
    [Google Scholar]
  4. , , . Superoxide dismutases:Role in redox signaling, vascular function, and diseases. Antioxid Redox Signal. 2011;15:1583-606.
    [Google Scholar]
  5. , , , , . Extracellular superoxide dismutase and its role in cancer. Free Radic Biol Med. 2017;112:464-79.
    [Google Scholar]
  6. , , . Regulation of superoxide dismutase genes:Implications in disease. Free Radic Biol Med. 2009;47:344-56.
    [Google Scholar]
  7. , , . Manganese superoxide dismutase in cancer prevention. Antioxid Redox Signal. 2014;20:1628-45.
    [Google Scholar]
  8. , , , , , , . The novel SOD mimetic GC4419 increases cancer cell killing with sensitization to ionizing radiation while protecting normal cells. Free Radic Biol Med. 2020;160:630-42.
    [Google Scholar]
  9. , , , , , . The effect of exogenous superoxide dismutase (SOD) on caspase-3 activation and apoptosis induction in Pc-3 prostate cancer cells. Res Rep Urol. 2020;12:503-8.
    [Google Scholar]
  10. , , , , , , . SOD1 promotes cell proliferation and metastasis in non-small cell lung cancer via an miR-409-3p/SOD1/SETDB1 epigenetic regulatory feedforward loop. Front Cell Dev Biol. 2020;8:213.
    [Google Scholar]
  11. , , . The redox stress hypothesis of aging. Free Radic Biol Med. 2012;52:539-55.
    [Google Scholar]
  12. , , , , , , . The role of manganese superoxide dismutase in skin aging. Dermatoendocrinol. 2012;4:232-5.
    [Google Scholar]
  13. , . Superoxide dismutase, aging, and degenerative disease. Free Radic Biol Med. 1994;17:249-58.
    [Google Scholar]
  14. , , , , , , . Clinical efficacy of melon GliSODin®for the treatment of aging-related dysfunction in motor organs-a double blind, randomized placebo-controlled study. J Clin Med. 2022;11:2747.
    [Google Scholar]
  15. , , , , , . Superoxide dismutase administration:A review of proposed human uses. Molecules. 2021;26:1844.
    [Google Scholar]
  16. , , , . Application and utility of liposomal neuroprotective agents and biomimetic nanoparticles for the treatment of Ischemic stroke. Pharmaceutics. 2022;14:361.
    [Google Scholar]
  17. , , , , , , . Nanoparticles for targeted delivery of antioxidant enzymes to the brain after cerebral ischemia and reperfusion injury. J Cereb Blood Flow Metab. 2013;33:583-92.
    [Google Scholar]
  18. , , , , . Cu, Zn- superoxide dismutase liposomal dry powder formulations production using supercritical CO2-assisted spray-drying:A proof-of-concept. J Supercrit Fluids. 2023;200:105991.
    [Google Scholar]
  19. , , , , , . Pharmacokinetics and distribution of superoxide dismutase liposomes in rats. Yao Xue Xue Bao. 2005;40:173-7.
    [Google Scholar]
  20. , , , , , , . Effect of nanoformulated copper/zinc superoxide dismutase on chronic ethanol-induced alterations in liver and adipose tissue. Alcohol. 2019;79:71-9.
    [Google Scholar]
  21. , , , , . Targeting strategies for tissue-specific drug delivery. Cell. 2020;181:151-67.
    [Google Scholar]
  22. , , , . On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proc Natl Acad Sci U S A. 2021;118:e2109256118.
    [Google Scholar]
  23. , , , , . SOD mimics:From the tool box of the chemists to cellular studies. Curr Opin Chem Biol. 2022;67:102109.
    [Google Scholar]
  24. , , . Immunological studies of oxidized superoxide dismutase in patients with systemic lupus erythematosus. Correlation with disease induction and progression. Saudi Med J. 2012;33:1177-84.
    [Google Scholar]
  25. , , , , , . Biochemical and immunological studies on erythrocytes superoxide dismutase modified by nitric oxide in patients with alopecia areata:Implications in alopecia patchy persistent and alopecia universalis. Immunol Lett. 2014;160:50-7.
    [Google Scholar]
  26. , , , . Mechanisms of ADC toxicity and strategies to increase ADC tolerability. Cancers (Basel). 2023;15:713.
    [Google Scholar]
  27. , , . Therapeutic potential of astaxanthin and superoxide dismutase in Alzheimer's disease. Open Biol. 2021;11:210013.
    [Google Scholar]
  28. , , , , , . Rationale of combination therapy with antioxidants in medical management of Peyronie's disease:Results of clinical application. Res Rep Urol. 2017;9:129-39.
    [Google Scholar]
  29. , , , , , , . Antioxidant and anti-tumor effects of dietary vitamins A, C, and E. Antioxidants (Basel). 2023;12:632.
    [Google Scholar]
  30. , , , , , . Therapeutic utilities of SOD mimetics:Cancer, radiotherapy and SOD mimetics. In:Madame Curie Bioscience Database. Austin, TX: Landes Bioscience;2000-2013; Available from: https://www.ncbi.nlm.nih.gov/books/NBK6099
    [Google Scholar]
  31. , , , , , , . Utilizing superoxide dismutase mimetics to enhance radiation therapy response while protecting normal tissues. Semin Radiat Oncol. 2019;29:72-80.
    [Google Scholar]

Fulltext Views
3

PDF downloads
3
View/Download PDF
Download Citations
BibTeX
RIS
Show Sections