Commentary - (2024)Volume 9, Issue 1
Targeted Treatment of Diseases of Immune Dysregulation
Smriti Mohan MD
*
*Correspondence:
Smriti Mohan, Department of Pediatric Rheumatology, University of Michigan,
Michigan,
USA,
Email:
Author info »
Description
Increasing awareness of immune system functioning has led to
discovery and understanding of diseases that affect both the
innate and adaptive immune systems. Historically, the immune
system was considered to have two arms, the innate and adaptive
systems. Innate immunity was considered the first and fastest,
though less specific line of immunological defense against
evolutionarily conserved sequences. Adaptive immunity was
considered a more specific, targeted line of immunological
defense, mediated by T and B lymphocytes through recognition
of foreign antigens with trained tolerance to self-antigens. It is
increasingly clear that there is crosstalk between the innate and
adaptive immune systems, and disorders of immune
dysregulation are caused by impaired regulation of both the
innate and/or adaptive arms of the immune system [1,2].
Successful treatment of these conditions relies on increasing our
knowledge of the molecular mechanisms leading to disease and
targeting these pathways. Currently available biologics are
categorized by the cytokines that they inhibit.
Currently available biologics and small molecule
inhibitors
Interleukin-1 inhibitors
Approved in treatment of SAIDs, RA and used in sJIA. Injection
site reactions, infections, and neutropenia are the main
side effects, as well as hepatotoxicity with anakinra, though
risks appear to be modest [3-8].
Anakinra: Daily injection, short-acting IL-1 receptor antagonist.
Rilonacept: Weekly injection, soluble IL-1 decoy receptor.
Canakinumab: Long-acting injection, monoclonal antibody.
Interleukin-18 inhibitors
Used for treatment of NLRC4 inflammasome mutations
(presents with early onset colitis and MAS). The main side effects
are injection site reaction, arthralgia,and upper respiratory
infections. There are 2 more monoclonal antibodies still in early
clinical trials.
Tadekinig alfa: Recombinant human IL-18 binding protein
[4]. Currently in clinical trials [9-13].
Novel inflammasome inhibitors
Currently in various stages of preclinical development to block
inflammation at the level of the inflammasome [14-17].
Interleukin-6 inhibitors
Approved in treatment of RA, JIA, sJIA, and is used to
treat refractory SAIDs [4]. Neutropenia, thrombocytopenia,
hyperlipidemia, and transaminitis are the major side effects
of these biologics [18-21].
Tocilizumab: Recombinant humanized IL-6 receptor
monoclonal antibody, available as injection or infusion [22].
Sarilumab: Fully human IL-6 receptor alpha monoclonal antibody
[12]. Appears to bind with greater affinity than tocilizumab and
may help in patients with AOSD who failed tocilizumab therapy
[22,23].
TH17 cytokine inhibitors
Block IL-17 and IL-22 which are produced by Th17 cells to
recruit neutrophils for defense against extracellular bacteria and
fungi. Also blocks IL-23, which is important in proliferation and
maintenance of Th17 cells. Approved in treatment of psoriasis
and arthritis, and in some cases, inflammatory bowel disease.
Main side effects include increased infection risk and antibody
development.
Ustekinumab: Fully human monoclonal antibody against IL-12
and IL-23. Approved in Crohn’s disease and ulcerative colitis in
addition to psoriasis and psoriatic arthritis. Induction dosing is
intravenous in IBD, otherwise administered as injections with
average maintenance dosing range from 8-12 weeks [24-26].
Secukinumab: Selective IL-17A monoclonal antibody, preventing
binding to IL-17 receptor. Administered by monthly injection and
approved for ankylosing spondylitis in addition to psoriasis and
psoriatic arthritis. It can trigger Crohn’s disease and is
contraindicated in treatment of IBD [27,28].
Ixekizumab: Humanized monoclonal antibody to IL-17A
similar to secukinumab.
Guselkumab: Monoclonal antibody against IL-23 administered
by injection. [29,30].
Interferon blockade
Anifrolumab
Fully humanized type I interferon monoclonal antibody targeting
IFN-α/β receptor. Administered as monthly intravenous infusion
[4]. Approved in treatment of systemic lupus erythematosus but
may have application in treatment of interferonopathies [31,32].
Emapalumab: Fully human anti-IFN-γ monoclonal antibody to
block signal transduction of IFN-γ. May be efficacious in
treatment of NRLC4-MAS inflammasomopathy and refractory
sJIA/AOSD with MAS and is administered by intravenous
infections every 2 weeks [12]. Appears to have mild adverse
effects, with increased risk of viral infections, though more data
needed [33,34].
Tumor necrosis factor alpha inhibitors
First biologic agents developed as targeted therapy against
proinflammatory cytokines, and approved in variety of
autoimmune conditions including RA, JIA, AS, IBD, psoriasis
and uveitis. Multiple biosimilars are now available in this
category [24]. Side effects include increased infection risk,
hypersensitivity, malignancy, and autoimmune conditions [4].
They are used in treatment of certain SAIDs, including TRAPS
and the vascular phenotype of DADA2 [35-40].
Etanercept
Binds soluble TNF-α administered as weekly injection.
Adalimumab: Fully humanized monoclonal antibody,
administered as injection every 2 weeks.
Infliximab: Chimeric monoclonal antibody administered as
intravenous infusion, with wider dosing and frequency to
optimize disease control than other agents.
Golimumab: Second generation, fully humanized monoclonal
antibody available in injection and intravenous forms.
Certolizumab pegol: Pegylated monoclonal antibody administered
as injection.
Janus kinase inhibitors
New immunosuppressive agents classified as targeted synthetics
DMARDs. Approved for wide indication of treatment of both
autoimmune and autoinflammatory diseases, including
interferonopathies, RA, JIA, AS, psoriatic arthritis, with
increasing use in dermatomyositis. They block a combination of
the 4 JAKs in the JAK/STAT pathway [4]. Major side effects
include increased infection risk, hyperlipidemia, and among
older adults, higher risk of cardiovascular events and malignancy.
There is also risk of acute cytokine storm syndrome from abrupt
cessation of JAK inhibitors [41-48].
Baricitinib and ruxolitinib: Block JAK1 and JAK2.
Tofacitinib: Blocks JAK1 and JAK3.
Upadacitinib and filgotinib: Selectively block JAK1.
T-cell inhibitors
Abatacept: Recombinant CTLA-4 fusion protein binding CD80
and CD86 on antigen-presenting cells, ultimately blocking T-cell
activation. Approved in JIA, RA, and PsA with trials in systemic
sclerosis and localized scleroderma. Available in injection and
intravenous forms. Has similar infection risk to other biologics
[49-54].
B-cell inhibitors
Rituximab: Chimeric monoclonal antibody against CD20,
approved for RA and other autoimmune conditions including
SLE, ANCA-vasculitis, and autoimmune encephalitis [39].
Administered as intravenous infusion, with maintenance doses
usually separated by several months. Side effects include
hypersensitivity reaction, hypogammaglobulinemia, and
increased malignancy risk [55-61].
Belimumab: Human monoclonal antibody that inhibits B
lymphocyte stimulator protein binding to receptors on
B lymphocytes. Currently approved in treatment of SLE,
and administered as monthly infusions or weekly injections
[62-64].
Bortezomib: Plasma cell inhibitor that blocks the 26S proteasome
involved in degrading ubiquitinated proteins. Administered by
injection or intravenous infusion. Has more side effects with
peripheral neuropathy, gastrointestinal issues, thrombocytopenia,
and fatigue. Approved in multiple myeloma, but recent studies
suggest there may be efficacy in autoimmune diseases [65-67].
Conclusion
Abnormalities in both the innate and adaptive immune pathways
are increasingly being implicated in pathogenesis of some diseases
previously characterized as either autoinflammatory or
autoimmune. Treatment of these conditions should be based on
the mechanisms of known pathogenesis. Side effects of biologics
include increased infection risk, hypersensitivity reaction, and
increased malignancy risk. Anakinra, tocilizumab, and JAK
inhibitors can also cause hepatotoxicity.
References
- Bindu S, Dandapat S, Manikandan R, Dinesh M, Subbaiyan A, Mani P et al. Prophylactic and therapeutic insights into trained immunity: A renewed concept of innate immune memory. Hum Vaccines Immunother. 2022;18(1):2040238.
[Google Scholar] [Pub Med] [Cross Ref]
- Zhang Y, Yang W, Li W, Zhao Y. NLRP3 Inflammasome: Checkpoint connecting innate and adaptive immunity in autoimmune diseases. Front Immunol. 2021;12:732933.
[Google Scholar] [Pub Med] [Cross Ref]
- Sfriso P, Bindoli S, Galozzi P. Adult-onset still’s disease: Molecular pathophysiology and therapeutic advances. Drugs. 2018;78(12):1187-1195.
[Google Scholar] [Pub Med] [Cross Ref]
- Du Y, Liu M, Nigrovic PA, Dedeoglu F, Lee PY. Biologics and JAK inhibitors for the treatment of monogenic systemic auto inflammatory diseases in children. J Allergy Clin Immunol. 2023;151(3):607-618.
[Google Scholar] [Pub Med] [Cross Ref]
- Arnold DD, Yalamanoglu A, Boyman O. Systematic review of safety and efficacy of IL-1-targeted biologics in treating immune-mediated disorders. Front Immunol. 2022;13:888392.
[Google Scholar] [Pub Med] [Cross Ref]
- Romano M, Arici ZS, Piskin D, Alehashemi S, Aletaha D, Barron KS, et al. The 2021 EULAR/American college of rheumatology points to consider for diagnosis, management and monitoring of the interleukin-1 mediated autoinflammatory diseases: Cryopyrin-associated periodic syndromes, tumour necrosis factor receptor-associated periodic syndrome, mevalonate kinase deficiency, and deficiency of the interleukin-1 receptor antagonist. Ann Rheum Dis. 2022;81(7):907-921.
[Google Scholar] [Pub Med] [Cross Ref]
- Murray GM, Ng SK, Beasley D, Johansen L, Ramanan AV. Severe hepatotoxicity as a rare side effect of anakinra in a patient with systemic JIA. Rheumatol Oxf Engl. 2021;60(9):e307-e308.
[Google Scholar] [Pub Med] [Cross Ref]
- Ahmed O, Brahmania M, Alsahafi M, Alkhowaiter S, Erb S. Anakinra hepatotoxicity in a patient with adult-onset still’s disease. ACG Case Rep J. 2015;2(3):173-174.
[Google Scholar] [Pub Med] [Cross Ref]
- Alehashemi S, Goldbach-Mansky R. Human autoinflammatory diseases mediated by NLRP3-, Pyrin-, NLRP1-, and NLRC4-inflammasome dysregulation updates on diagnosis, treatment, and the respective roles of IL-1 and IL-18. Front Immunol. 2020;11:1840.
[Google Scholar] [Pub Med] [Cross Ref]
- Kaplanski G. Interleukin-18: Biological properties and role in disease pathogenesis. Immunol Rev. 2018;281(1):138-153.
[Google Scholar] [Pub Med] [Cross Ref]
- Dinarello CA, Novick D, Kim S, Kaplanski G. Interleukin-18 and IL-18 binding protein. Front Immunol. 2013;8:4:289.
[Google Scholar] [Pub Med] [Cross Ref]
- Galozzi P, Bindoli S, Doria A, Sfriso P. Progress in biological therapies for adult-onset still’s disease. Biologics. 2022;16:21-34.
[Google Scholar] [Pub Med] [Cross Ref]
- Gabay C, Fautrel B, Rech J, Spertini F, Feist E, Kotter I, et al. Open-label, multicentre, dose-escalating phase II clinical trial on the safety and efficacy of tadekinig alfa (IL-18BP) in adult-onset Still’s disease. Ann Rheum Dis. 2018;77(6):840-847. 2017-212608.
[Google Scholar] [Pub Med] [Cross Ref]
- Oliviero F, Bindoli S, Scanu A, Feist E, Doria A, Galozzi P, et al. Autoinflammatory mechanisms in crystal-induced arthritis. Front Med. 2020;7:166.
[Google Scholar] [Pub Med] [Cross Ref]
- Moltrasio C, Romagnuolo M, Marzano AV. NLRP3 inflammasome and NLRP3-related autoinflammatory diseases: From cryopyrin function to targeted therapies. Front Immunol. 2022;13:1007705.
[Google Scholar] [Pub Med] [Cross Ref]
- Blevins HM, Xu Y, Biby S, Zhang S. The NLRP3 Inflammasome pathway: A review of mechanisms and inhibitors for the treatment of inflammatory diseases. Front Aging Neurosci. 2022;14:879021.
[Google Scholar] [Pub Med] [Cross Ref]
- Jiang H, He H, Chen Y, Huang W, Cheng J, Ye Jet, al. Identification of a selective and direct NLRP3 inhibitor to treat inflammatory disorders. J Exp Med. 2017;214(11):3219-3238.
[Google Scholar] [Pub Med] [Cross Ref]
- Barbulescu A, Delcoigne B, Askling J, Frisell T. Gastrointestinal perforations in patients with rheumatoid arthritis treated with biological disease-modifying antirheumatic drugs in Sweden: A nationwide cohort study. RMD Open. 2020;6(2):e001201.
[Google Scholar] [Pub Med] [Cross Ref]
- Curtis JR, Xie F, Chen L. The incidence of gastrointestinal perforations among rheumatoid arthritis patients. Arthritis Rheum. 2011;63(2):346-351.
[Google Scholar] [Pub Med] [Cross Ref]
- Xie F, Yun H, Bernatsky S, Curtis JR. Brief Report: Risk of gastrointestinal perforation among rheumatoid arthritis patients receiving Tofacitinib, Tocilizumab, or other biologic treatments. Arthritis Rheumatol. 2016;68(11):2612-2617.
[Google Scholar] [Pub Med] [Cross Ref]
- Soyer O, Demir S, Bilginer Y, Batu ED, Sonmez HE, Arici ZS, et al. Severe hypersensitivity reactions to biological drugs in children with rheumatic diseases. Atanaskovic‐Markovic M, ed. Pediatr Allergy Immunol. 2019;30(8):833-840.
[Google Scholar] [Pub Med] [Cross Ref]
- Ogata A, Kato Y, Higa S, Yoshizaki K. IL-6 inhibitor for the treatment of rheumatoid arthritis: A comprehensive review. Mod Rheumatol. 2019;29(2):258-267.
[Google Scholar] [Pub Med] [Cross Ref]
- Lamb YN, Deeks ED. Sarilumab: A review in moderate to severe rheumatoid arthritis. Drugs. 2018;78(9):929-940.
[Google Scholar] [Pub Med] [Cross Ref]
- Feuerstein JD, Ho EY, Shmidt E, Singh H, Falck-Ytter Y, Sultan S et al. Aga clinical practice guidelines on the medical management of moderate to severe luminal and perianal fistulizing crohn’s disease. Gastroenterology. 2021;160(7):2496-2508.
[Google Scholar] [Pub Med] [Cross Ref]
- Tan MG, Worley B, Kim WB, Ten Hove M, Beecker J. Drug-induced intracranial hypertension: A systematic review and critical assessment of drug-induced causes. Am J Clin Dermatol. 2020;21(2):163-172.
[Google Scholar] [Pub Med] [Cross Ref]
- Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, Yeiding N et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008;371(9625):1675-1684.
[Google Scholar] [Pub Med] [Cross Ref]
- Papp KA, Langley RG, Sigurgeirsson B, Abe M, Baker DR, Konno P, et al. Efficacy and safety of secukinumab in the treatment of moderate-to-severe plaque psoriasis: A randomized, double-blind, placebo-controlled phase II dose-ranging study. Br J Dermatol. 2013;168(2):412-421.
[Google Scholar] [Pub Med] [Cross Ref]
- Fauny M, Moulin D, D’Amico F, Netter P, Petitpain N, Arnone D et al. Paradoxical gastrointestinal effects of interleukin-17 blockers. Ann Rheum Dis. 2020;79(9):1132-1138.
[Google Scholar] [Pub Med] [Cross Ref]
- Blauvelt A, Papp KA, Griffiths CEM, Randazzo B, Wasfi Y, Shen YK et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: Results from the phase III, double-blinded, placebo-and active comparator-controlled VOYAGE 1 trial. J Am Acad Dermatol. 2017;76(3):405-417.
[Google Scholar] [Pub Med] [Cross Ref]
- Deodhar A, Helliwell PS, Boehncke WH, Kollimeier AP, Hsia EC, Subramanian RA et al. Guselkumab in patients with active psoriatic arthritis who were biologic-naive or had previously received TNFα inhibitor treatment (DISCOVER-1): A double-blind, randomised, placebo-controlled phase 3 trial. The Lancet. 2020;395(10230):1115-1125.
[Google Scholar] [Pub Med] [Cross Ref]
- Saulescu I, Ionescu R, Opris-Belinski D. Interferon in systemic lupus erythematosus-A halfway between monogenic autoinflammatory and autoimmune disease. Heliyon. 2022;8(11):e11741.
[Google Scholar] [Pub Med] [Cross Ref]
- Sim TM, Ong SJ, Mak A, Tay SH. Type I interferons in systemic lupus erythematosus: a journey from bench to bedside. Int J Mol Sci. 2022;23(5):2505.
[Google Scholar] [Pub Med] [Cross Ref]
- Locatelli F, Jordan MB, Allen C, Cesaro S, Rizzari C, Rao A, et al. Emapalumab in children with primary hemophagocytic lymphohistiocytosis. N Engl J Med. 2020;382(19):1811-1822.
[Google Scholar] [[Cross Ref]
- Garonzi C, Chinello M, Cesaro S. Emapalumab for adult and pediatric patients with hemophagocytic lymphohistiocytosis. Expert Rev Clin Pharmacol. 2021;14(5):527-534.
[Google Scholar] [Pub Med] [Cross Ref]
- Leone GM, Mangano K, Petralia MC, Nicoletti F, Fagone P. Past, present and (Foreseeable) future of biological anti-TNF alpha therapy. J Clin Med. 2023;12(4):1630.
[Google Scholar] [Pub Med] [Cross Ref]
- Matsuda T, Kambe N, Takimoto-Ito R, Ueki Y, Nakamizo S, Saito MK, et al. Potential benefits of TNF targeting therapy in blau syndrome, a NOD2-associated systemic autoinflammatory granulomatosis. Front Immunol. 2022;13:895765.
[Google Scholar] [Pub Med] [Cross Ref]
- Cudrici C, Deuitch N, Aksentijevich I. Revisiting TNF Receptor Associated Periodic Syndrome (TRAPS): Current perspectives. Int J Mol Sci. 2020;21(9):3263.
[Google Scholar] [Pub Med] [Cross Ref]
- Deuitch NT, Yang D, Lee PY, Yu X, Moura NS, Schnappauf O et al. TNF inhibition in vasculitis management in adenosine deaminase 2 deficiency (DADA2). J Allergy Clin Immunol. 2022;149(5):1812-1816.e6
[Google Scholar] [Pub Med] [Cross Ref]
- Shams S, Martinez JM, Dawson JRD, Flores J, Gabriel M, Garcia G, et al. The therapeutic landscape of rheumatoid arthritis: Current state and future directions. Front Pharmacol. 2021;12:680043
[Google Scholar] [Pub Med] [Cross Ref]
- Ramiro S, Nikiphorou E, Sepriano A, Ortolan A, Webers C, Baraliakos X et al. ASAS-EULAR recommendations for the management of axial spondyloarthritis: 2022 update. Ann Rheum Dis. 2023;82(1):19-34.
[Google Scholar] [Pub Med] [Cross Ref]
- Cetin Gedik K, Lamot L, Romano M, Demiraya E, Piskin D, Torreggiani S et al. The 2021 european alliance of associations for rheumatology/american college of rheumatology points to consider for diagnosis and management of autoinflammatory type I interferonopathies: CANDLE/PRAAS, SAVI and AGS. Ann Rheum Dis. 2022;81(5):601-613.
[Google Scholar] [Pub Med] [Cross Ref]
- Bertsias G. Therapeutic targeting of JAKs: From hematology to rheumatology and from the first to the second generation of JAK inhibitors. Mediterr J Rheumatol. 2020;31(Suppl 1):105.
[Google Scholar] [Pub Med] [Cross Ref]
- Boyadzhieva Z, Ruffer N, Burmester G, Pankow A, Krusche M. Effectiveness and safety of JAK inhibitors in autoinflammatory diseases: A systematic review. Front Med. 2022;9:930071.
[Google Scholar] [Pub Med] [Cross Ref]
- Hadjadj J, Castro CN, Tusseau M, Stolzenberg MC, Mazerolles F, Aladjidi N et al. Early-onset autoimmunity associated with SOCS1 haploinsufficiency. Nat Commun. 2020;11(1):5341
[Google Scholar] [Pub Med] [Cross Ref]
- Michniacki TF, Walkovich K, deMeyer L, deMeyer L, Basiaga ML, Horst KL, et al. SOCS1 haploinsufficiency presenting as severe enthesitis, bone marrow hypocellularity, and refractory thrombocytopenia in a pediatric patient with subsequent response to JAK inhibition. J Clin Immunol. 2022;42(8):1766-1777.
[Google Scholar] [Pub Med] [Cross Ref]
- Natour AEH, Kivity S. Biological therapies in inflammatory myopathies. Rambam Maimonides Med J. 2023;14(2):e0008.
[Google Scholar] [ PubMed] [ Cross Ref]
- Kim H, Gunter-Rahman F, McGrath JA, Lee E, Jesus AAD, Targoff IN et al. Expression of interferon-regulated genes in juvenile dermatomyositis versus Mendelian autoinflammatory interferonopathies. Arthritis Res Ther. 2020;22(1):69.
[Google Scholar] [PubMed] [Cross Ref]
- Le Voyer T, Gitiaux C, Authier FJ, Bodemer C, Melki I, Quartier P, et al. JAK inhibitors are effective in a subset of patients with juvenile dermatomyositis: A monocentric retrospective study. Rheumatology. 2021;60(12):5801-5808.
[Google Scholar] [PubMed] [Cross Ref]
- Jayatilleke A. Immunosuppression in rheumatologic and auto-immune disease. Handb Exp Pharmacol. 2022:272:181-208.
[Google Scholar] [PubMed] [Cross Ref]
- Pinto MV, Neves JF. Precision medicine: The use of tailored therapy in primary immunodeficiencies. Front Immunol. 2022;13:1029560.
[Google Scholar] [PubMed] [Cross Ref]
- Krausz M, Uhlmann A, Rump IC, Ihorst G, Goldacker S, Sogkas G et al. The ABACHAI clinical trial protocol: Safety and efficacy of abatacept (s.c.) in patients with CTLA-4 insufficiency or LRBA deficiency: a non-controlled phase 2 clinical trial. Contemp Clin Trials Commun. 2022;30:101008.
[Google Scholar] [PubMed] [Cross Ref]
- Kremer JM, Dougados M, Emery P, Durez P, Sibilia J, Shergy W et al. Treatment of rheumatoid arthritis with the selective costimulation modulator abatacept: twelve-month results of a phase iib, double-blind, randomized, placebo-controlled trial. Arthritis Rheum. 2005;52(8):2263-2271.
[Google Scholar] [PubMed] [Cross Ref]
- Watkins B, Qayed M, McCracken C, Bratrude B, Betz K, Suessmuth Y et al. Phase II Trial of costimulation blockade with abatacept for prevention of acute GVHD. J Clin Oncol. 2021;39(17):1865-1877.
[Google Scholar] [PubMed] [Cross Ref]
- Dominique A, Hetland ML, Finckh A, Gottenberg JE, Lannone F, Caporali R et al. Safety outcomes in patients with rheumatoid arthritis treated with abatacept: Results from a multinational surveillance study across seven European registries. Arthritis Res Ther. 2023;25(1):101.
[Google Scholar] [PubMed] [Cross Ref]
- Mariette X, Barone F, Baldini C, Bootsma H, Clark KL, Devita S et al. A randomized, phase II study of sequential belimumab and rituximab in primary sjögren’s syndrome. JCI Insight. 2022;7(23):e163030.
[Google Scholar] [ PubMed] [Cross Ref]
- Chung SA, Langford CA, Maz M, Abril A, Gorelik M, Guyatt G et al. 2021 American college of rheumatology/vasculitis foundation guideline for the management of antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheumatol. 2021;73(8):1366-1383.
[Google Scholar] [PubMed] [Cross Ref]
- Fraenkel L, Bathon JM, England BR, Clair EW St, Arayssi T, Carandang K et al. 2021 American college of rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Care Res. 2021;73(7):924-939.
[Google Scholar] [PubMed] [Cross Ref]
- Dinoto A, Ferrari S, Mariotto S. Treatment options in refractory autoimmune encephalitis. CNS Drugs. 2022;36(9):919-931.
[Google Scholar] [PubMed] [Cross Ref]
- Gottenberg JE, Guillevin L, Lambotte O, Combe B, Allanore Y, Cantagrel A et al. Tolerance and short term efficacy of rituximab in 43 patients with systemic autoimmune diseases. Ann Rheum Dis. 2005;64(6):913-920.
[Google Scholar] [PubMed] [Cross Ref]
- Higashida J, Wun T, Schmidt S, Naguwa SM, Tuscano JM. Safety and efficacy of rituximab in patients with rheumatoid arthritis refractory to disease modifying antirheumatic drugs and anti-tumor necrosis factor-alpha treatment. J Rheumatol. 2005;32(11):2109-2115.
[Google Scholar] [PubMed]
- Marinho A, Delgado Alves J, Fortuna J, Faria R, Almeida I, Alves G et al. Biological therapy in systemic lupus erythematosus, antiphospholipid syndrome, and Sjögren’s syndrome: Evidence and practice-based guidance. Front Immunol. 2023;14:1117699.
[Google Scholar] [PubMed] [Cross Ref]
- Krustev E, Clarke AE, Barber MRW. B cell depletion and inhibition in systemic lupus erythematosus. Expert Rev Clin Immunol. 2023;19(1):55-70.
[Google Scholar] [PubMed] [Cross Ref]
- Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzova D et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63(12):3918-3930.
[Google Scholar] [PubMed] [Cross Ref]
- Navarra SV, Guzmán RM, Gallacher AE, Hall S, Levy RA, Jimenz RE et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: A randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377(9767):721-731.
[Google Scholar] [PubMed] [Cross Ref]
- Sanz-Solas A, Labrador J, Alcaraz R, Cuevas B, Vinuesa R, Cuevas MV et al. Bortezomib pharmacogenetic biomarkers for the treatment of multiple myeloma: Review and future perspectives. J Pers Med. 2023;13(4):695.
[Google Scholar] [PubMed] [Cross Ref]
- Cengiz Seval G, Beksac M. The safety of bortezomib for the treatment of multiple myeloma. Expert Opin Drug Saf. 2018;17(9):953-962.
[Google Scholar] [PubMed] [Cross Ref]
- Modica RF, Thatayatikom A, Bell-Brunson DH, Elder ME. Bortezomib is efficacious in the treatment of severe childhood-onset neuropsychiatric systemic lupus erythematosus with psychosis: A case series and mini-review of B-cell immunomodulation in antibody-mediated diseases. Clin Rheumatol. 2023;42(7):1965-1979.
[Google Scholar] [PubMed] [Cross Ref]
Author Info
Smriti Mohan MD
*
Department of Pediatric Rheumatology, University of Michigan, Michigan, USA
Citation: Mohan S (2024) Targeted Treatment of Diseases of Immune Dysregulation. Lupus: Open Access. 9:276.
Received: 29-Dec-2023, Manuscript No. LOA-23-28833;
Editor assigned: 03-Jan-2024, Pre QC No. LOA-23-28833 (PQ);
Reviewed: 17-Jan-2024, QC No. LOA-23-28833;
Revised: 25-Jan-2024, Manuscript No. LOA-23-28833 (R);
Published:
02-Feb-2024
, DOI: 10.35248/2684-1630.24.9.276
Copyright: © 2024 Mohan S. 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.