Targeting Monocyte Abnormalities in Systemic Lupus Erythematosus through Omics-Based Drug Repurposing (2024)

Loading

  • Journals
  • Policies
  • Register
  • Contact Us
  • Special Collections

Journal of Cellular Immunology

ISSN: 2689-2812

Editors

Author GuidelinesSubmit Manuscript

Latest ContentCurrent IssuesArchives

Open Special IssuesPublished Special IssuesAbout Special Issues

Commentary Open Access
Volume 6 | Issue 3 | DOI: https://doi.org/10.33696/immunology.6.196

Targeting Monocyte Abnormalities in Systemic Lupus Erythematosus through Omics-Based Drug Repurposing

Panagiotis Garantziotis1,2,3.*, Dimitrios Nikolakis4,5,6, Eleni Frangou1,7,8, George Bertsias9,10, Dimitrios T. Boumpas1,11
  • 1Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
  • 2Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
  • 3Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg and Uniklinikum Erlangen
  • 4Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Department of Gastroenterology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
  • 5Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
  • 6Amsterdam Institute for Infection & Immunity, Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
  • 7Department of Nephrology, Limassol General Hospital, State Health Services Organization, Limassol, Cyprus
  • 8University of Nicosia Medical School, Cyprus
  • 9Rheumatology, Clinical Immunology and Allergy Department, Medical School University of Crete, Heraklion, Greece
  • 10Laboratory of Auoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
  • 114th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece

    + Affiliations - Affiliations

    *Corresponding Author

    Panagiotis Garantziotis, garantziotis.p@gmail.com

    Received Date: April 10, 2024

    Accepted Date: May 22, 2024

    Citation

    Garantziotis P, Nikolakis D, Frangou E, Bertsias G, Boumpas DT. Targeting Monocyte Abnormalities in Systemic Lupus Erythematosus through Omics-Based Drug Repurposing. J Cell Immunol. 2024;6(3):113-116.

    Copyright © 2024 Garantziotis P, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

    Commentary

    Systemic Lupus Erythematosus (SLE) is a complex disease marked by extensive immune system dysfunction, culminating in a diverse spectrum of clinical phenotypes of varying severity [1]. Despite the significant advancements in elucidating the pathogenesis of the disease, the management of SLE remains largely empirical with attainment of low disease activity and remission targets being an infrequent outcome among patients [1]. Strikingly, the intricate molecular landscape and the notorious heterogeneity of the disease pose substantial challenges in the design of successful clinical trials, resulting in the scarcity of approved therapeutic modalities, which further amplifies the burden on SLE patients.

    The systemic nature of the disease underscores the widely distributed immune alterations that drive autoimmunity targeting nucleic acids and their associated proteins, and inciting tissue-damaging inflammation [2]. Evidence highlights the essential role of innate immune system in the initiation, propagation of autoimmunity and development of organ damage in SLE [3] and a plethora of phenotypic and functional abnormalities of the macrophage/monocyte lineage cells has been reported [4,5]. The impaired phagocytic activity of macrophages resulting in the inefficient clearance of apoptotic material and immune-complexes is instrumental in the breakdown of self-tolerance and the subsequent promotion of autoantibody production, a unifying feature of SLE [6]. Additionally, in SLE, monocytes are acknowledged as significant contributors to the type I interferon production, a hallmark of the disease [7]. Monocytes serve as the primary source of IFN in the pristane-induced murine lupus model, defined by high IFN signature, and human monocyte-derived macrophages demonstrate elevated IFN-α and IFN-β gene expression when transfected with a small non-coding Y RNA or stimulated with immune complexes [7]. The aberrant activation of autoreactive B and T cells observed in SLE could be partially ascribed to the dysregulated cytokine production by monocytes. Monocytes from SLE patients exhibit heightened production of B-lymphocyte stimulator (BLyS), fostering the survival and proliferation of B cells [8]. Moreover, in the peripheral blood of SLE patients, monocytes are major producers of IL-10 and IL-6, amplifying antibody production and facilitating plasma cell differentiation, respectively [8]. Thus, directing therapeutic interventions towards monocytes could potentially target fundamental abnormalities in SLE, offering significant therapeutic benefits to the patients.

    Considering the paucity of approved medications in SLE and the high attrition rates, repurposing "old" drugs for the disease is increasingly appealing, providing the advantage of utilizing de-risked compounds for new indications. Advanced computational tools have streamlined both de novo drug development and drug repurposing processes, enabling the reduction of overall costs and shortening development timelines (Table 1). The Connectivity Map (CMap) project serves as a pioneering drug repurposing platform, incorporating gene expression responses from four human cell lines exposed to various doses of a large collection of FDA-approved compounds [9]. Building upon this groundwork, the NIH-supported Library of Integrated Network-Based Cellular Signatures (LINCS) expanded the transcriptomic databases of the CMap project by integrating gene expression profiles from over 60 cell lines before and after exposure to more than 20,000 perturbagens [10]. Using the Lincscloud, the successor to CMap, drug candidates capable of reversing the SLE-related transcriptional signatures were determined, highlighting the therapeutic potential of phosphoinositide 3-kinase (PI3K) and mammalian target of rapamycin (mTOR) inhibitors in SLE therapeutics [11]. Utilizing CMap for computational drug repurposing analysis and integrating significant gene targets derived from transcriptome-wide association studies, several clinically relevant drug classes potentially suitable for SLE treatment were detected [12]. Among these are glucocorticoid receptor agonists, histone deacetylase (HDAC) inhibitors, mTOR inhibitors, and topoisomerase inhibitors, all identified as promising candidates [12]. In the same context, patient stratification based on the drug responsiveness suggested that mTOR and TNFa inhibitors could potentially reverse the transcriptional signatures of the lymphocyte- and neutrophil-driven subgroups, respectively [13]. Achieving low disease activity state or remission is crucial for SLE patients, offering significant benefits in terms of minimizing organ damage accrual and preventing flares [14]. Interestingly, the drug-target interaction analysis highlights the modulation of toll-like receptor (TLR) cascades, Bruton tyrosine kinase (BTK) activity, cytotoxic T lymphocyte antigen 4 (CTLA-4)-related inhibitory signaling, and the nucleotide-binding oligomerization domain leucine-rich repeat-containing protein 3 (NLRP3) inflammasome pathways as promising strategies for attaining lupus low disease activity state or remission in SLE [15].

    Table 1. Selected studies of computational drug repurposing in SLE.

    Study (year of publication)

    Computational approach

    Key drug candidates

    Toro-Domínguez et al. (2017) [11]

    Lincscloud

    PI3K and mTOR inhibitors

    Toro-Domínguez et al. (2019) [13]

    CLUE

    mTOR and TNFa inhibitors

    Owen et al. (2020) [25]

    LINCS, STITCH (v.5.0), and IPA

    African ancestry: bortezomib, PF-06650833, IRAK4-specific inhibitor; European ancestry: TYK2 inhibitor

    Noor et al. (2021) [26]

    Semantic Web (SW) technologies

    Aspirin, azathioprine, cyclophosphamide, indomethacin, methotrexate, leflunomide, warfarin, clopidogrel, peginterferon alfa-2a, and peginterferon alfa-2b

    Frangou et al. (2022) [16]

    L1000 Characteristic Direction Signature Search Engine (L1000CDS2)

    R(+)−6-BROMO-APB, HEMADO, norketamine hydrochloride and trichostatin A

    Garantziotis et al. (2022) [17]

    CMap, iLINCS

    PI3K/mTOR pathway inhibitors, JAK2 inhibitor

    Nikolakis et al. (2023) [19]

    iLINCS, CLUE, L1000CDS2, DGIDb

    NF-κB inhibitor, Pim-1/NFATc1/NLRP3 pathway inhibitor, HSP90 inhibitors

    Khunsriraksakul et al. (2023) [12]

    CMap

    Glucocorticoid receptor agonist, HDAC inhibitor, mTOR inhibitor, and topoisomerase inhibitor

    Parodis et al. (2024) [15]

    Reactome Pathways

    Bruton tyrosine kinase inhibitors, TLR7 and TLR9 inhibitors

    Our group has also previously employed computational signature mapping, comparing transcriptional profiles of drugs against diseases or clinical phenotypes, to identify potential novel therapeutic agents in autoimmune diseases [16-19]. Applying an unbiased, whole blood transcriptome driven molecular taxonomy approach and leveraging two robust, high throughput platforms (iLINCS and CLUE), we devised a personalized drug repurposing pipeline to propose novel compounds capable of counteracting the endotype-specific transcriptional abberations observed in SLE patients [17]. Notably, agents such as dactolisib and fedratinib, targeting the PI3K/mTOR and the JAK/STAT pathways respectively, emerged as promising candidates to ameliorate the SLE related transcriptional disturbances in a personalized manner [17]. In light of the limitations of the currently available therapeutic interventions, failing to induce remission in over 50% of patients with lupus nephritis (LN), we next conducted a comparative cross-tissue and cross-species gene expression analysis to identify nephritis-specific genes for subsequent drug repurposing analysis [16]. Using the L1000 Characteristic Direction Signature Search Engine, we detected agents predicted to reverse the transcriptional signatures associated with active LN and the transition from preclinical to overt disease [16]. Among these, R(+)−6-BROMO-APB was predicted to reverse the former signature, while HEMADO, norketamine hydrochloride, and trichostatin A were anticipated to counteract the latter signature in the HA1E kidney cell line, indicating the potential for further evaluation in LN therapeutics [16].

    Given the reasonable assumption that drugs reversing the transcriptional profile linked to a disease state hold therapeutic promise and acknowledging the pivotal involvement of macrophage/monocyte lineage cells in SLE pathogenesis, the identification of compounds capable of modifying SLE-specific transcriptional abnormalities in monocytes warrants significant consideration. Employing the iLINCS, CLUE, and L1000CDS2 platforms, we identified small molecules predicted to most effectively counteract the SLE monocyte-specific gene signature [19]. Compounds disrupting the Pim-1/NFATc1/NLRP3 signaling axis or inhibiting the heat shock protein 90 (HSP90) were forecasted to efficiently mitigate the abnormal monocyte signature in SLE, consistent with experimental evidence suggesting that inhibition of the Pim-1/NFATc1/NLRP3 pathway improves nephritis in lupus mouse models [20], and HSP90 facilitates TLR7/9-mediated nucleic acid recognition in SLE [21].

    Focusing solely on transcriptome reversal may overlook opportunities to modulate key regulatory hub genes within the biological system, potentially constraining the effectiveness of drug repurposing endeavors. Therefore, adopting a holistic approach that integrates information on hub genes and their interactions alongside transcriptomic data is essential to ensure comprehensive target identification and therapeutic efficacy. Applying a network-based drug repurposing strategy incorporating upstream regulators of the SLE monocyte-signature, the IL-12/IL-23 inhibitor ustekinumab was identified as a potential candidate for efficiently disrupting the molecular interaction network of monocytes. While proteasome inhibitors effectively deplete autoreactive plasma cells and demonstrate therapeutic efficacy in preclinical mouse models of LN, evidence suggests that immunoproteasome inhibition may selectively induce apoptosis in CD14+ monocytes, supporting the validity of our network-based drug repurposing strategy, which implied the therapeutic targeting of SLE monocytes by bortezomib [22].

    Despite the significant successes achieved in drug repurposing, several limitations must be carefully considered [23,24]. While repurposed drugs may bypass phase I clinical trials, which primarily focus on safety evaluation, concerns regarding drug safety remain a significant challenge. The safety profile established for a drug in one patient population may not automatically translate to another population, prompting a need for the re-evaluation of safety parameters. Furthermore, the dosing regimen validated for the original indication may not be optimal for new therapeutic uses, necessitating adjustments and further investigation. Lack of specificity for the new indication poses a substantial hurdle, particularly when utilizing SLE blood specimens that do not entirely mirror the molecular alterations in target tissues. Lastly, intellectual property barriers can impose additional limitations on the drug repurposing process.

    In conclusion, omics-based drug repurposing emerges as a promising alternative to de novo drug development, offering the potential to expedite the delivery of compounds to patients. Leveraging machine-learning and artificial intelligence methodologies to integrate the vast array of publicly available SLE omics data holds significant promise for advancing drug repurposing efforts in the treatment of SLE.

    1. Fanouriakis A, Tziolos N, Bertsias G, Boumpas DT. Update on the diagnosis and management of systemic lupus erythematosus. Annals of the rheumatic diseases. 2021 Jan 1;80(1):14-25.

    2. Crow MK. Pathogenesis of systemic lupus erythematosus: risks, mechanisms and therapeutic targets. Annals Rheumatic Dis. 2023 Aug 1;82(8):999-1014.

    3. Gupta S, Kaplan MJ. Bite of the wolf: innate immune responses propagate autoimmunity in lupus. The J clin invest. 2021 Feb 1;131(3):e144918.

    4. Katsiari CG, Liossis SN, Sfikakis PP. The pathophysiologic role of monocytes and macrophages in systemic lupus erythematosus: a reappraisal. Seminars in Arthritis and Rheumatism. 2010 Jun;39(6):491-503.

    5. Stergioti EM, Manolakou T, Sentis G, Samiotaki M, Kapsala N, Fanouriakis A, et al. Transcriptomic and proteomic profiling reveals distinct pathogenic features of peripheral non-classical monocytes in systemic lupus erythematosus. Clin Immunol. 2023 Oct;255:109765.

    6. Mahajan A, Herrmann M, Muñoz LE. Clearance Deficiency and Cell Death Pathways: A Model for the Pathogenesis of SLE. Front Immunol. 2016 Feb 8;7:35.

    7. Rönnblom L, Leonard D. Interferon pathway in SLE: one key to unlocking the mystery of the disease. Lupus Sci Med. 2019 Aug 13;6(1):e000270.

    8. Li Y, Lee PY, Reeves WH. Monocyte and macrophage abnormalities in systemic lupus erythematosus. Arch Immunol Ther Exp (Warsz). 2010 Oct;58(5):355-64.

    9. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006 Sep 29;313(5795):1929-35.

    10. Keenan AB, Jenkins SL, Jagodnik KM, Koplev S, He E, Torre D, et al. The Library of Integrated Network-Based Cellular Signatures NIH Program: System-Level Cataloging of Human Cells Response to Perturbations. Cell Syst. 2018 Jan 24;6(1):13-24.

    11. Toro-Domínguez D, Carmona-Sáez P, Alarcón-Riquelme ME. Support for phosphoinositol 3 kinase and mTOR inhibitors as treatment for lupus using in-silico drug-repurposing analysis. Arthritis Res Ther. 2017 Mar 11;19(1):54.

    12. Khunsriraksakul C, Li Q, Markus H, Patrick MT, Sauteraud R, McGuire D, et al. Multi-ancestry and multi-trait genome-wide association meta-analyses inform clinical risk prediction for systemic lupus erythematosus. Nat Commun. 2023 Feb 7;14(1):668.

    13. Toro-Domínguez D, Lopez-Domínguez R, García Moreno A, Villatoro-García JA, Martorell-Marugán J, Goldman D, et al. Differential Treatments Based on Drug-induced Gene Expression Signatures and Longitudinal Systemic Lupus Erythematosus Stratification. Sci Rep. 2019 Oct 29;9(1):15502.

    14. Pitsigavdaki S, Nikoloudaki M, Garantziotis P, Silvagni E, Repa A, Marangoni A, et al. Pragmatic targets for moderate/severe SLE and their implications for clinical care and trial design: sustained DORIS or LLDAS for at least 6 months is sufficient while their attainment for at least 24 months ensures high specificity for damage-free progression. Ann Rheum Dis. 2024 Mar 12;83(4):464-74.

    15. Parodis I, Lindblom J, Barturen G, Ortega-Castro R, Cervera R, Pers JO, et al. PRECISESADS Clinical Consortium; Alarcón-Riquelme ME, Beretta L. Molecular characterisation of lupus low disease activity state (LLDAS) and DORIS remission by whole-blood transcriptome-based pathways in a pan-European systemic lupus erythematosus cohort. Ann Rheum Dis. 2024 Feb 19:ard-2023-224795.

    16. Frangou E, Garantziotis P, Grigoriou M, Banos A, Nikolopoulos D, Pieta A, et al. Cross-species transcriptome analysis for early detection and specific therapeutic targeting of human lupus nephritis. Ann Rheum Dis. 2022 Oct;81(10):1409-19.

    17. Garantziotis P, Nikolakis D, Doumas S, Frangou E, Sentis G, Filia A, et al. Molecular Taxonomy of Systemic Lupus Erythematosus Through Data-Driven Patient Stratification: Molecular Endotypes and Cluster-Tailored Drugs. Front Immunol. 2022 May 9;13:860726.

    18. Garantziotis P, Doumas SAP, Boletis I, Frangou E. Gene Expression as a Guide to the Development of Novel Therapies in Primary Glomerular Diseases. J Clin Med. 2021 May 24;10(11):2262.

    19. Nikolakis D, Garantziotis P, Sentis G, Fanouriakis A, Bertsias G, Frangou E, et al. Restoration of aberrant gene expression of monocytes in systemic lupus erythematosus via a combined transcriptome-reversal and network-based drug repurposing strategy. BMC Genomics. 2023 Apr 18;24(1):207.

    20. Fu R, Xia Y, Li M, Mao R, Guo C, Zhou M, et al. Pim-1 as a Therapeutic Target in Lupus Nephritis. Arthritis Rheumatol. 2019 Aug;71(8):1308-18.

    21. Saito K, Kukita K, Kutomi G, Okuya K, Asanuma H, Tabeya T, et al. Heat shock protein 90 associates with Toll-like receptors 7/9 and mediates self-nucleic acid recognition in SLE. Eur J Immunol. 2015 Jul;45(7):2028-41.

    22. Basler M, Claus M, Klawitter M, Goebel H, Groettrup M. Immunoproteasome Inhibition Selectively Kills Human CD14+ Monocytes and as a Result Dampens IL-23 Secretion. J Immunol. 2019 Oct 1;203(7):1776-85.

    23. Li X, Peng T. Strategy, Progress, and Challenges of Drug Repurposing for Efficient Antiviral Discovery. Front Pharmacol. 2021 May 4;12:660710.

    24. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019 Jan;18(1):41-58.

    25. Owen KA, Price A, Ainsworth H, Aidukaitis BN, Bachali P, Catalina MD, et al. Analysis of Trans-Ancestral SLE Risk Loci Identifies Unique Biologic Networks and Drug Targets in African and European Ancestries. Am J Hum Genet. 2020 Nov 5;107(5):864-81.

    26. Noor A, Assiri A. A novel computational drug repurposing approach for Systemic Lupus Erythematosus (SLE) treatment using Semantic Web technologies. Saudi J Biol Sci. 2021 Jul;28(7):3886-92.

    • Download PDF
    • Join as Reviewer
    • Propose a Special Issue

    Recommended Articles

    The Pathogenesis of Continuous Spike and Waves during Slow Sleep Syndrome: Short Communication

    Continuous spikes during slow wave sleep (CSWS) syndrome is an age-related and self-limited severe epileptic encephalopathy characterized by electrical status epilepticus in sleep (ESES) on electroencephalogram, seizures, and developmental regression

    Sleep Disturbances are a Significant Predictor of Chikungunya Arthritis Flare Severity

    Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitoes that causes debilitating arthritis. In a cohort of 500 Colombian patients, one quarter reported joint pain 20 months after infection, and further, 1 out of 8 had joint pain three years after infection.

    Impact of Sleep on Autophagy and Neurodegenerative Disease: Sleeping Your Mind Clear

    Sleep is an evolutionarily conserved phenomenon which has survived tremendous evolutionary pressures. Its disruption has deleterious implications for human health. The importance of sleep is illustrated by the fact that sleep deprivation in many animals leads to death. While sleep is tightly regulated by a combination of intrinsic and extrinsic factors it becomes progressively disrupted in old age and in neurodegenerative diseases including Alzheimer’s disease (AD), frontotemporal dementia (FTD), Parkinson’s disease (PD) and Huntington’s disease (HD).

    The Issue of Monocyte Activation in ASD: Troubles withTranslation

    Autism spectrum disorder (ASD) prevalence has increased year on year for the past two decades and currently affects 1 in 44 individuals in the US. An increasing number of studies have pointed to increased immune activation as both an etiological agent and also involved in the ongoing pathological process of ASD. Both adaptive and innate immune responses have been implicated. Evidence of innate dysregulation has so far included increased production of innate inflammatory cytokines, increased cell numbers, and altered activation in monocytes in the blood and microglia in the brain.

    Treatment of Sleep Disorders in Children with Atopic Dermatitis: What's New?

    Atopic dermatitis (AD), also known as atopic eczema, is a chronic and inflammatory disease caused by an interaction of genetic, immunological, and environmental factors. It is characterized by skin lesions and itching and progresses with periods of improvement and worsening.

    Author Information X

    References Information

    About Scientific Archives

    Scientific Archives is a global publisher initiated with the mission of ensuring equal opportunity for accessing science to research community all over the world. Spreading research findings with great relevance to all channels without any barrier is our goal. We want to overcome the challenges of Open Access with ensured quality and transparency.

    Read More

    Targeting Monocyte Abnormalities in Systemic Lupus Erythematosus through Omics-Based Drug Repurposing (2024)

    FAQs

    What are the therapeutic targets for systemic lupus erythematosus? ›

    Both belimumab and anifrolumab target molecules produced by the innate immune system that play a key role in the activation of adaptive immune response in SLE. Therefore, the role of innate immune system, centered on dendritic cells, has been the focus of new therapeutic targets for SLE.

    What are immunosuppressive drugs in systemic lupus erythematosus? ›

    Treating Lupus with Immunosuppressive Medications
    • Azathioprine (Imuran)
    • Mycophenolate mofetil (Cellcept)
    • Cyclosporine (Neoral, Sandimmune, Gengraf)
    • Methotrexate (Rheumatrex)
    • Leflunomide (Arava)
    • Cyclophosphamide (Cytoxan)
    • Chlorambucil (Leukeran)
    • Nitrogen mustard (Mustargen)

    What is the adherence to medications in systemic lupus erythematosus? ›

    Conclusion: this study revealed that more than half of the studied patient with SLE were low level adherence to their medications, and more than half of them had poor knowledge related to SLE disease. In addition to it was found that patient related factors are most factors affect medications adherence for SLE.

    How do you treat systemic lupus erythematosus Pubmed? ›

    Hydroxychloroquine is recommended for all patients with SLE to prevent flare-ups, organ damage, and thrombosis and increase long-term survival.

    What is the treatment of choice for systemic lupus erythematosus? ›

    Specific medication recommendations include the following: Hydroxychloroquine is recommended for all patients with SLE. Glucocorticoids can provide rapid symptom relief, but the medium- to long-term aim should be to minimize the daily dose to ≤7.5 mg/day prednisone equivalent or to discontinue them.

    What is the primary target of systemic lupus erythematosus (SLE)? ›

    The treatment target of SLE should be remission of systemic symptoms and organ manifestations or, where remission cannot be reached, the lowest possible disease activity, measured by a validated lupus activity index and/or by organ-specific markers.

    What happens if you leave lupus untreated? ›

    One of the most serious complications of SLE is lupus nephritis, which describes an immune system attack on the kidneys, leading to inflammation and the possibility of organ damage. Left untreated, symptoms can worsen and lead to kidney failure.

    Can you get disability for lupus? ›

    If your doctor provides you with a lupus diagnosis, you may qualify for Social Security Disability Insurance (SSDI) once you can no longer work due to impaired physical function. To get approved, you'll need to meet the Social Security Administration's (SSA) criteria under the Lupus listing in their Blue Book.

    Is coffee bad for lupus? ›

    Researchers of one study found that those with lupus who had a daily caffeine intake between 154 mg – 377 mg/day experienced a lower prevalence of “lupus nephritis, neuropsychiatric involvement, hematological manifestations, hypocomplementemia, and anti-DNA positivity.” These individuals also had lower blood levels of ...

    What are anti inflammatory drugs for systemic lupus erythematosus? ›

    Common NSAIDs include:
    • Ibuprofen (like Advil® or Motrin®)
    • Naproxen (like Aleve® or Naprosyn®)
    • Indomethacin (Indocin®)
    • Nabumetone (Relafen®)
    • Celecoxib (Celebrex®)
    • Aspirin.
    Aug 4, 2021

    What are the 46 drugs that cause drug-induced lupus? ›

    Causes
    • Isoniazid.
    • Hydralazine.
    • Procainamide.
    • Tumor-necrosis factor (TNF) alpha inhibitors (such as etanercept, infliximab and adalimumab)
    • Minocycline.
    • Quinidine.
    Apr 30, 2023

    How do you stop drug-induced lupus? ›

    While lupus may damage your kidneys or lungs, drug-induced lupus rarely affects your body's major organs. It's also temporary. Once you stop the medicine that causes it, symptoms usually clear up within a few weeks or months.

    What is the latest drug for lupus? ›

    In 2021, the FDA approved Saphnelo (anifrolumab-fnia), a targeted therapy to treat adults with systemic lupus. Advances in the understanding and treatment of lupus over the last several decades have resulted in people with the disease living longer and better lives.

    What medications should you avoid if you have lupus? ›

    Sulfa drugs, which make a person more sensitive to the sun, such as: Bactrim® and Septra® (trimethoprim-sulfamethoxazole); sulfisoxazole (Gantrisin®); tolbutamide (Orinase®); sulfasalazine (Azulfidine®); diuretics. Sun-sensitizing tetracycline drugs such as minocycline (Minocin®)

    What is the new treatment for systemic lupus erythematosus? ›

    Saphnelo (anifrolumab)—approved in August 2021. This monoclonal antibody (a protein that finds and attaches to one type of substance, called a cytokine, in the body) is designed to treat an excess of interferon activation, which plays an essential role in lupus inflammation.

    What is the treat to target approach in SLE? ›

    Roger: “Treat-to-target” is a medical strategy that sets a treatment objective/goal. For SLE, the objective can be either remission – defined as the absence of signs and symptoms of significant inflammatory disease activity – or low disease activity.

    What is the therapeutic management of the patient with systemic lupus erythematosus? ›

    • Lifestyle changes and preventive interventions. Sun protection. Diet and nutrition. Exercise. Avoiding smoking. Vaccines. ...
    • Medication therapy. Hydroxychloroquine. Nonsteroidal antiinflammatory drugs (NSAIDs) Immunosuppressive medications. Other medications.
    • Reproductive considerations. Pregnancy. Family planning and contraception.
    Feb 7, 2024

    What is the treatment goal for lupus? ›

    In addition to controlling your systems, the overall goals of your treatment plan will be: Reduce your inflammation. Prevent flares and treat them as they occur. Minimize damage to your organs.

    What does SLE target? ›

    Systemic lupus erythematosus (SLE) is a multisystemic autoimmune disease characterised by the presence of autoantibodies towards nuclear antigens, immune complex deposition, and chronic inflammation at classic target organs such as skin, joints, and kidneys.

    References

    Top Articles
    Brook Park Resident Wins $10,000 on Ohio Lottery Scratch-Off
    Scratch-offs | Hoosier Lottery
    7 C's of Communication | The Effective Communication Checklist
    Woodward Avenue (M-1) - Automotive Heritage Trail - National Scenic Byway Foundation
    Cash4Life Maryland Winning Numbers
    Chalupp's Pizza Taos Menu
    Videos De Mexicanas Calientes
    Free VIN Decoder Online | Decode any VIN
    Roblox Developers’ Journal
    Gameplay Clarkston
    Https Www E Access Att Com Myworklife
    Wmlink/Sspr
    Snowflake Activity Congruent Triangles Answers
    Danielle Longet
    Hope Swinimer Net Worth
    What to do if your rotary tiller won't start – Oleomac
    Charmeck Arrest Inquiry
    Top tips for getting around Buenos Aires
    A rough Sunday for some of the NFL's best teams in 2023 led to the three biggest upsets: Analysis - NFL
    House Of Budz Michigan
    Nissan Rogue Tire Size
    Rachel Griffin Bikini
    Icommerce Agent
    Craigslist Toy Hauler For Sale By Owner
    Aaa Saugus Ma Appointment
    ELT Concourse Delta: preparing for Module Two
    Geometry Review Quiz 5 Answer Key
    Pokemon Unbound Shiny Stone Location
    Sef2 Lewis Structure
    Craigs List Tallahassee
    8005607994
    3 2Nd Ave
    Cable Cove Whale Watching
    Viduthalai Movie Download
    Miles City Montana Craigslist
    Free Tiktok Likes Compara Smm
    R/Sandiego
    Bursar.okstate.edu
    Wake County Court Records | NorthCarolinaCourtRecords.us
    Craigslist Dallastx
    Of An Age Showtimes Near Alamo Drafthouse Sloans Lake
    Blackstone Launchpad Ucf
    Avance Primary Care Morrisville
    Kb Home The Overlook At Medio Creek
    Sarahbustani Boobs
    Pulaski County Ky Mugshots Busted Newspaper
    Levi Ackerman Tattoo Ideas
    Enjoy Piggie Pie Crossword Clue
    Horseneck Beach State Reservation Water Temperature
    Smoke From Street Outlaws Net Worth
    Electric Toothbrush Feature Crossword
    Karen Kripas Obituary
    Latest Posts
    Article information

    Author: Twana Towne Ret

    Last Updated:

    Views: 5513

    Rating: 4.3 / 5 (64 voted)

    Reviews: 95% of readers found this page helpful

    Author information

    Name: Twana Towne Ret

    Birthday: 1994-03-19

    Address: Apt. 990 97439 Corwin Motorway, Port Eliseoburgh, NM 99144-2618

    Phone: +5958753152963

    Job: National Specialist

    Hobby: Kayaking, Photography, Skydiving, Embroidery, Leather crafting, Orienteering, Cooking

    Introduction: My name is Twana Towne Ret, I am a famous, talented, joyous, perfect, powerful, inquisitive, lovely person who loves writing and wants to share my knowledge and understanding with you.