1
|
Arribas-Rodríguez E, De Prado Á, de Andrés B, Velayos B, Barrio J, Romero A, García-Alonso FJ, Martín-Muñoz Á, Garrote JA, Arranz E, Fernández-Salazar L, Bernardo D. Tofacitinib downregulates JAK1 and JAK3 on human intestinal monocytes and macrophages without affecting dendritic cells phenotype or function. J Transl Autoimmun 2025; 10:100271. [PMID: 39925953 PMCID: PMC11802370 DOI: 10.1016/j.jtauto.2025.100271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/11/2025] Open
Abstract
Background Ulcerative colitis (UC) is an inflammatory disorder of the gastrointestinal tract. Although Tofacitinib, which inhibits the JAK1 and JAK3 signalling pathway, is approved to treat patients with UC, its specific mechanism of action remain elusive. Given the central role that conventional dendritic cells (cDC) elicit in gut homeostasis, we hypothesised that Tofacitinib acts modulating cDC function in UC. Methods Human biopsies were obtained from colon of controls, and patients with UC (active and quiescent). Lamina propria mononuclear cells (LPMC) were ex-vivo cultured in the presence/absence of Tofacitinib. The specific effect elicited over human intestinal cDC, monocytes and macrophages was assessed by flow cytometry. cDC were also enriched following Tofacitinib conditioning in order to assess its effect over naïve T-cells. Results Several human intestinal cDC, monocyte and macrophage subsets can be found in the human colon, with these cells being more similar between controls and patients with qUC referred to patients with aUC. Following ex-vivo culture, Tofacitinib downregulated JAK1 expression on intestinal monocytes from patients with both active and quiescent UC. As for macrophages, JAK1 was decreased on patients with active UC while JAK was downregulated on macrophages from patients with quiescent disease. Tofacitinib did not modulate the phenotype or function of human intestinal cDC. Conclussion Tofacitinib does not modulate the phenotype and function of human intestinal cDC in UC. On the contrary, it displays a differential capacity to modulate intestinal monocyte and macrophage phenotype. Future studies should address whether it also translates into a differential function of these cells.
Collapse
Affiliation(s)
- Elisa Arribas-Rodríguez
- Mucosal Immunology Lab, Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - Ángel De Prado
- Mucosal Immunology Lab, Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Beatriz de Andrés
- Servicio de Cirugía General, Hospital Clínico Universitario, Valladolid, Spain
| | - Benito Velayos
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
| | - Jesús Barrio
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Alejandro Romero
- Servicio de Cirugía General, Hospital Clínico Universitario, Valladolid, Spain
| | | | - Álvaro Martín-Muñoz
- Cytometry Facility. Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - José A. Garrote
- Mucosal Immunology Lab, Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - Eduardo Arranz
- Mucosal Immunology Lab, Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | | | - David Bernardo
- Mucosal Immunology Lab, Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
2
|
Yargi-Ozkocak B, Cilingir-Kaya OT, Peker Eyuboglu İ, Erzik C, Direskeneli H, Celiker H. Therapeutic efficacy of tofacitinib in PDSAg-induced chronic experimental autoimmune uveitis in Wistar rat. Immunopharmacol Immunotoxicol 2025:1-9. [PMID: 40433864 DOI: 10.1080/08923973.2025.2508278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE Tofacitinib, a small molecule pan-JAK inhibitor, targets key inflammatory pathways that play a pivotal role in the pathophysiology of uveitis. Its ability to inhibit multiple cytokine signaling pathways makes it a promising candidate for the treatment of ocular inflammation. This study aimed to investigate the effect of oral tofacitinib on peptide-derived S-antigen (PDSAg)-induced chronic experimental autoimmune uveitis (EAU) in rats. MATERIALS AND METHODS Nineteen albino Wistar rats were divided into five groups. Groups 1-3 (5 rats each) were immunized with 15 micrograms PDSAg to induce EAU; Group 2 received tofacitinib (5 mg/kg) by gavage twice daily. Group 3 received saline in the same manner as Group 2. Group 4 (2 rats) was a healthy control group. Group 5 (2 rats) received only tofacitinib. Uveitis development and treatment efficacy were evaluated using clinical scoring based on the the signs of anterior segment inflammation and histological scoring based on the deterioration of the retinal architectural structure. RESULTS Uveitis confirmed based on histological evidence was observed in all EAU groups (Groups 1-3) compared to the healthy control group (Group 4) (p < 0.001, Mann-Whitney U test). Tofacitinib significantly delayed the onset of uveitis in Group 2 when compared with Groups 1 and 3, which did not receive tofacitinib (p < 0.001, Mann-Whitney U test). Histological scores also showed a trend toward reduction (p = 0.393, Mann-Whitney U test). CONCLUSIONS Oral tofacitinib delayed uveitis onset and reduced clinical and histological findings, suggesting its potential as an alternative treatment for uveitis.
Collapse
Affiliation(s)
- Berru Yargi-Ozkocak
- Beyoglu Eye Training and Research Hospital, University of Health Sciences Turkey, Istanbul, Turkey
| | | | - İrem Peker Eyuboglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Marmara University, İstanbul, Turkey
| | - Can Erzik
- Department of Medical Biology and Genetics, Faculty of Medicine, Marmara University, İstanbul, Turkey
| | - Haner Direskeneli
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Marmara University, Istanbul, Turkey
| | - Hande Celiker
- Division of Uveal Disease, Department of Ophthalmology, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
3
|
Heidari N, Ghannadzadeh Kermani Pour R, Farshbafnadi M, Heidari A, Ghane Y. A systematic review of tumor necrosis factor-α blockers, anti-interleukins, and small molecule inhibitors for dissecting cellulitis of the scalp treatment. Orphanet J Rare Dis 2025; 20:236. [PMID: 40383754 PMCID: PMC12085841 DOI: 10.1186/s13023-025-03720-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Dissecting cellulitis of the scalp (DCS) is a type of neutrophilic scarring alopecia identified by the development of folliculitis with clusters of perifollicular pustules and then progresses to abscesses and intercommunicating sinus formation. In the absence of evidence-based guidelines, the treatment of DCS remains a therapeutic challenge. Our study aimed to assess the safety and efficacy of biologics, including tumor necrosis factor-α (TNF-α) blockers, anti-interleukins (ILs), and small molecule inhibitors, including Janus kinase (JAK) inhibitors and phosphodiesterase inhibitors in treating DCS. METHODS PubMed/Medline, Scopus, and Ovid Embase databases were systematically searched until February 4th, 2024. Study selection was restricted to case reports, case series, cohort studies, and clinical trials published in English-language. NIH and Murad et al.'s quality assessment tools were utilized for critical appraisal. RESULTS A total of 34 articles involving 81 patients met the inclusion criteria. The immunomodulators studied for the treatment of DCS include adalimumab, infliximab, certolizumab pegol, ustekinumab, secukinumab, guselkumab, risankizumab, tildrakizumab, apremilast, upadacitinib, and baricitinib. Our findings implied that TNF-α blockers and IL inhibitors were associated with clinical improvement in most individuals with moderate-to-severe DCS, especially in those who had failed earlier treatments. Moreover, certolizumab pegol could be a safe option for DCS in pregnancy. In addition, the prescription of small molecule inhibitors, including JAK inhibitors and apremilast in DCS patients, demonstrated a significant amelioration in DCS symptoms with a desirable safety profile. Nevertheless, the available data was limited, warranting further investigation. Besides, all aforementioned immunomodulators are still debated for their effectiveness on hair regrowth and reversing the scarring process. CONCLUSIONS The application of immunomodulators in treating DCS was associated with satisfactory outcomes, although there is still a need to assess the long-term safety and effectiveness of these therapeutic agents in preventing disease progression and new flare-ups.
Collapse
Affiliation(s)
- Nazila Heidari
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Amirhossein Heidari
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Yekta Ghane
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Fowler EA, Sacramento LA, Bowman BA, Lee B, Lio CWJ, Dong YD, Spicer JA, Trapani JA, Novais FO. Hypoxia and IL-15 cooperate to induce perforin expression by CD8 T cells and promote damage to the skin in murine cutaneous leishmaniasis. J Invest Dermatol 2025:S0022-202X(25)00479-8. [PMID: 40373956 DOI: 10.1016/j.jid.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/17/2025]
Abstract
Cutaneous leishmaniasis is a disease caused by protozoan parasites of the genus Leishmania, and although parasites influence disease severity, cytotoxic CD8 T cell responses mediate damage to the infected skin. We found that the cytotoxic protein perforin was expressed in CD8 T cells only upon recruitment to Leishmania-infected skin, suggesting that lesional inflammatory cues induced perforin. Here, using a mouse model of Leishmania major infection, we demonstrated that the expression of perforin was driven by a combination of hypoxia and IL-15, both of which are microenvironmental signals present within Leishmania-infected skin. We also demonstrated that the major sources of Il15 mRNA in cutaneous leishmaniasis lesions are neutrophils and macrophages and that macrophages exposed to hypoxia in vitro produce more Il15. Since perforin is only present in lesions, we reformulated a small molecule perforin inhibitor for topical application and found that local inhibition of perforin is sufficient to ameliorate disease in established cutaneous leishmaniasis. Thus, topical perforin inhibition may be considered a therapeutic strategy for patients with cutaneous leishmaniasis and other inflammatory skin diseases where cytotoxic CD8 T cells contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Erin A Fowler
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University; Columbus, USA
| | - Laís Amorim Sacramento
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania; Philadelphia, USA
| | - Bridget A Bowman
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University; Columbus, USA
| | - Bella Lee
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University; Columbus, USA
| | - Chan-Wang J Lio
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University; Columbus, USA; Pelotonia Institute for Immuno-Oncology; The Ohio State Comprehensive Cancer Center, Columbus, OH, USA
| | - Yao-Da Dong
- Medicine Manufacturing Innovation Centre, Monash Institute for Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Julie A Spicer
- Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| | | | - Fernanda O Novais
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University; Columbus, USA.
| |
Collapse
|
5
|
Alabbasi S, Tacconelli S, de Vries M, Gunnarsson I, Oke V, Kvarnström M, De Michele A, Gregorio PD, Patrignani P, Idborg H, Jakobsson PJ. Janus kinase inhibitors enhance prostanoid biosynthesis in human whole blood in vitro: implications for cardiovascular side effects and prevention strategies. Ann Rheum Dis 2025:S0003-4967(25)00882-9. [PMID: 40328613 DOI: 10.1016/j.ard.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/27/2025] [Accepted: 03/27/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES Janus kinase inhibitors (JAKis) effectively treat chronic inflammatory diseases but are associated with cardiovascular side effects through unknown mechanisms. This study aimed to investigate the impact of JAKis on prothrombotic thromboxane (TX)A2 production in human whole blood (WB) as a possible mechanism. METHODS We evaluated the effects of 4 JAKis- tofacitinib, baricitinib, filgotinib, and upadacitinib (0.04-20.0 μM)-on TXB2 biosynthesis in clotting WB from healthy subjects, serving as a marker for platelet TXA2 generation. Additionally, we assessed the impact of these JAKis on TXB2 production in WB from healthy subjects, patients with systemic lupus erythematosus (SLE), and treatment-naïve patients with axial spondyloarthritis (axSpA) after 24-hour lipopolysaccharide (LPS) stimulation, as a marker of platelet and leukocyte prostanoid biosynthesis. RESULTS All JAKis increased serum TXB2 production in clotting WB, although not in a concentration-dependent manner. In LPS-stimulated WB, tofacitinib (1 μM) significantly increased TXB2 production in healthy subjects (HSs) (42% ± 33%, n = 17), patients with SLE (57% ± 39%, n = 12), and patients with axSpA (31% ± 23%, n = 15). Baricitinib (1 μM) also increased TXB2 in HSs (30% ± 22%, n = 10). Upadacitinib showed a trend towards increased TXB2 (46% ± 40%, n = 7), while filgotinib did not (21% ± 19%, n = 7). Aspirin (100 μM) almost completely reduced serum TXB2 in the presence of all JAKis. CONCLUSIONS The enhanced biosynthesis of TXA2 in platelets, with a minor contribution from leukocytes, may contribute to the increased cardiovascular risk associated with JAKis. Low-dose aspirin may offer a protective effect, warranting further investigations.
Collapse
Affiliation(s)
- Sabreen Alabbasi
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, G. d'Annunzio University Medical School, Chieti, Italy
| | - Mirjam de Vries
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Vilija Oke
- Center for Rheumatology (CFR), Academic Specialist Center, Stockholm Region and Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Marika Kvarnström
- Center for Rheumatology (CFR), Academic Specialist Center, Stockholm Region and Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Alessandra De Michele
- Systems Pharmacology and Translational Therapeutics Laboratory, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, G. d'Annunzio University Medical School, Chieti, Italy
| | - Patrizia Di Gregorio
- Transfusion Medicine Service of the Azienda Sanitaria Locale (ASL) Lanciano-Vasto-Chieti and G. d'Annunzio University, Chieti, Italy
| | - Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, G. d'Annunzio University Medical School, Chieti, Italy
| | - Helena Idborg
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden.
| | - Per-Johan Jakobsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
6
|
Sieiro Santos C, Herrero JG, Ordas Martínez J, Álvarez Castro C, López Robles A, Colindres R, Martín ER, Sahagun AM, Ruiz de Morales JG. Immunogenicity to herpes zoster recombinant subunit vaccine in immune-mediated rheumatic patients under treatment with JAK inhibitors. Rheumatology (Oxford) 2025; 64:2442-2450. [PMID: 39447032 DOI: 10.1093/rheumatology/keae584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/25/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVES Patients with immune-mediated rheumatic diseases (IMRDs) face an elevated risk of varicella-zoster virus infection (VZV) and herpes zoster (HZ). Treatment with immunosuppressors further increases the risk. A new recently approved adjuvant recombinant inactive vaccine offers safe protection against HZ. However, limited data exist on the efficacy of this new vaccine in patients with IMRDs treated with JAK inhibitors (JAK-i). We aimed to characterize B- and T-cell immune responses elicited by the HZ recombinant subunit vaccine in patients with IMRDs under treatment with JAK-i, and to identify factors that might be associated with reduced immunogenicity, and therefore reduced protection. METHODS We investigated humoral and cellular CD4 and CD8 immune responses following a two-dose regimen of the recombinant inactive vaccine in 43 patients with rheumatic diseases treated with different JAK-i. The responses were compared with age, gender and disease-matched healthy controls. RESULTS Patients with IMRDs treated with JAK-i showed reduced seroconversion rate (63% vs. 100% and lower VZV IgG titres (1222 ± 411 vs. 3048 ± 556, P < 0.0001) as compared with healthy controls. Functional T CD4 (IL-2 plus IFN-γ secretion) and T CD8 (granzyme A and/or granzyme B secretion) immune responses were also significantly diminished in IMRD patients. Negative correlation was found between VZV antibody titres and age, specific treatments (baricitinib, tofacitinib, upadacitinib), cumulative MTX and glucocorticoid doses, history of multiple DMARDs and treatment duration with JAK-i. Functional T-CD4 responses but not functional T-CD8 responses also showed similar negative correlations. Positive associations were observed between functional T-CD4 and T-CD8 responses. CONCLUSIONS Our study provides valuable insights into the immune responses elicited by the recombinant inactive vaccine in patients with IMRDs treated with JAK-i. In these patients we have observed a broad impact on the adaptive humoral and cellular immune responses, suggesting a potential reduction in protection against HZ infection and VZV reactivation.
Collapse
Affiliation(s)
- Cristiana Sieiro Santos
- Rheumatology Department, Complejo Asistencial Universitario de León, León, Spain
- Biomedical Sciences Department, Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - Juan Garcia Herrero
- Immunology Department, Complejo Asistencial Universitario de León, León, Spain
| | - Jose Ordas Martínez
- Rheumatology Department, Complejo Asistencial Universitario de León, León, Spain
| | | | | | - Ronald Colindres
- Immunology Department, Complejo Asistencial Universitario de León, León, Spain
| | | | - Ana M Sahagun
- Biomedical Sciences Department, Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - Jose G Ruiz de Morales
- Immunology Department, Complejo Asistencial Universitario de León, León, Spain
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| |
Collapse
|
7
|
Liu R, Shi X, Zeng W, Wang Y, Yan Z, Deng W, Hui J, Xia R, Mo L, Xu J, Liao T, Miao Y. Novel janus kinase 3 inhibitor ritlecitinib suppresses T and B cell responses to prevent acute cardiac allograft rejection in mice. Clin Immunol 2025; 273:110445. [PMID: 39954912 DOI: 10.1016/j.clim.2025.110445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/30/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Acute rejection is the main contributor to early allograft failure. Current immunosuppressive regimens have shortcomings, such as toxicity and minimal inhibitory effects on B cells. Hence, developing novel, effective, and selective anti-acute rejection drugs is crucial. Therefore, this study aimed to investigate the inhibitory effect of ritlecitinib (PF-06651600), a new janus kinase 3 inhibitor, on T and B cells, as well its preventive effects on acute allograft rejection. A murine cardiac transplantation model coupled with cell culture- and immunohistochemistry-based techniques was used. In vitro assays demonstrated that ritlecitinib inhibits naïve CD4+ T cell differentiation into T helper (Th)1 and Th17 cells, reduces relative inflammatory cytokines, and suppresses CD4+ and CD8+ T cell proliferation. Furthermore, ritlecitinib inhibited B cell activation and differentiation and antibody production. In vivo experiments revealed that ritlecitinib significantly prolongs allograft survival, decreases serum donor-specific antibody immunoglobulin G levels, alleviats allograft damage, and reduces C4d deposition and T, B, and plasma cell infiltration in allografts. Moreover, B and plasma cell percentages and counts were significantly decreased in recipient spleen, lymph nodes, bone marrow, and blood. Overall, ritlecitinib prevented acute rejection in cardiac transplantation by inhibiting T and B cells, suggesting its potential as a novel clinical immunosuppressant.
Collapse
Affiliation(s)
- Rumin Liu
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyi Shi
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenli Zeng
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuchen Wang
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziyan Yan
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenfeng Deng
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jialiang Hui
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Renfei Xia
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liqian Mo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Xu
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tao Liao
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yun Miao
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
8
|
Jarneborn A, Kopparapu PK, Jin T. The Dual-Edged Sword: Risks and Benefits of JAK Inhibitors in Infections. Pathogens 2025; 14:324. [PMID: 40333091 PMCID: PMC12030494 DOI: 10.3390/pathogens14040324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025] Open
Abstract
Janus kinase inhibitors (JAKis) represent a relatively new class of immunomodulatory drugs with potent effects on various cytokine signalling pathways. They have revolutionized the treatment landscape for autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, and ulcerative colitis. However, their ability to modulate immune responses presents a dual-edged nature, influencing both protective immunity and pathological inflammation. This review explores the complex role of JAKis in infectious settings, highlighting both beneficial and detrimental effects. On the one hand, experimental models suggest that JAK inhibition can impair host defence mechanisms, increasing susceptibility to certain bacterial and viral infections. For example, tofacitinib-treated mice exhibited more severe joint erosions in Staphylococcus aureus (S. aureus) septic arthritis and showed impaired viral clearance in herpes simplex encephalitis. Additionally, clinical data confirm an increased risk of herpes zoster in patients receiving JAKis, underscoring the need for rigorous monitoring. On the other hand, JAK inhibition has demonstrated protective effects in certain infectious and hyperinflammatory conditions. In sepsis models, including cecal ligation and puncture (CLP) and S. aureus bacteraemia, tofacitinib improved survival by attenuating excessive inflammation. Furthermore, JAKis, particularly baricitinib, have shown substantial efficacy in mitigating cytokine storms during severe COVID-19 infections, leading to improved clinical outcomes and reduced mortality. These observations suggest that JAKis have a role in modulating hyperinflammatory responses in select infectious contexts. In conclusion, JAKis present a complex interplay between immunosuppression and immunomodulation. While they increase the risk of certain infections, they also show potential in managing hyperinflammatory conditions such as cytokine storms. The key challenge is determining which patients and situations benefit most from JAKis while minimizing risks, requiring a careful and personalized treatment approach.
Collapse
Affiliation(s)
- Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (P.K.K.); (T.J.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (P.K.K.); (T.J.)
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (P.K.K.); (T.J.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| |
Collapse
|
9
|
Bi Z, Zhang Q, Gao H, Ge H, Zhan J, Yang M, Bu B. The JAK1/3 Inhibitor Tofacitinib Regulates Th Cell Profiles and Humoral Immune Responses in Myasthenia Gravis. Muscle Nerve 2025; 71:474-486. [PMID: 39821232 DOI: 10.1002/mus.28348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
INTRODUCTION/AIMS Tofacitinib, a first-generation Janus kinase (JAK) 1/3 inhibitor, is commonly used for treating ulcerative colitis and rheumatoid arthritis. However, its role in myasthenia gravis (MG) remains unclear. This study aimed to evaluate the immunomodulatory effects of tofacitinib on experimental autoimmune myasthenia gravis (EAMG) and peripheral blood mononuclear cells (PBMCs) from patients with MG. METHODS Flow cytometry, enzyme-linked immunosorbent assay (ELISA), quantitative reverse transcription polymerase chain reaction (qRT-PCR), and Western blot were used to evaluate the effects of tofacitinib on T helper (Th) cell profiles, humoral immune responses, and the JAK-signal transducer and activator of transcription (STAT) pathway proteins. RESULTS In vivo, tofacitinib significantly ameliorated EAMG severity in rats, reducing the proportions of Th1, Th17 and memory B cells, and anti-acetylcholine receptor (AChR) antibodies levels, while increasing the proportions of regulatory T (Treg) cells. In vitro, tofacitinib administration resulted in a significant decrease in the proportions of Th1 and IgG-secreting B cell, and a significant upregulation of Treg cells in mononuclear cells (MNCs) from EAMG rats, which was consistent with findings in PBMCs from MG patients. Further analysis revealed that tofacitinib inhibited CD4+ T cell differentiation into Th1 by decreasing phosphorylated STAT1 levels, while promoting Treg differentiation via increased phosphorylated STAT5 levels in MNCs from EAMG rats. DISCUSSION Tofacitinib modulates Th cell profiles and humoral immune responses by targeting the JAK-STAT pathway, suggesting its potential as a therapeutic candidate for MG. Further clinical studies are warranted to evaluate the efficacy and safety of tofacitinib in MG patients.
Collapse
Affiliation(s)
- Zhuajin Bi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Huajie Gao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Huizhen Ge
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayang Zhan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Mengge Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Yadav P, Wairkar S. Tofacitinib in focus: Fascinating voyage from conventional formulations to novel delivery systems. Int J Pharm 2025; 671:125253. [PMID: 39842741 DOI: 10.1016/j.ijpharm.2025.125253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/11/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Tofacitinib, a Janus kinase (JAK) inhibitor, has emerged as a primary therapeutic agent for managing autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, dermatitis and ulcerative colitis. By inhibiting the phosphorylation of JAK enzymes, tofacitinib prevents their activation within the JAK-STAT signaling pathway, which is vital for inflammatory responses. However, the tofacitinib delivery presents significant challenges, including pH-dependent solubility, poor permeability and susceptibility to oral degradation. This review provides an in-depth analysis of current and emerging formulations to enhance the delivery and efficiency of tofacitinib. This review highlights the physicochemical, pharmacodynamic and pharmacokinetic properties of tofacitinib. Additionally, it discusses various strategies, including oral modified release formulations, topical/transdermal delivery utilizing lipid-based and polymeric systems, and parenteral delivery systems. Recent advancements in nanotechnology, such as liposomes, micelles, keratinocyte exosomes, proposomes, proglycosomes, transethosomes, squalenyl nanoparticles and lyotropic liquid crystalline nanoparticles, are explored as potential nanocarriers to existing delivery constraints. The development of advanced tofacitinib delivery systems can address the challenges in its delivery and improve therapeutic outcomes and patient compliance, paving the way for enhanced treatment strategies in autoimmune and inflammatory conditions.
Collapse
Affiliation(s)
- Priti Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India.
| |
Collapse
|
11
|
Ito Y, Watanabe D, Okamoto N, Miyazaki H, Tokunaga E, Ku Y, Ooi M, Hoshi N, Kohashi M, Kanzawa M, Kodama Y. Activated type 17 helper T cells affect tofacitinib treatment outcomes. Sci Rep 2025; 15:6112. [PMID: 39971758 PMCID: PMC11840122 DOI: 10.1038/s41598-025-87076-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025] Open
Abstract
The incidence of ulcerative colitis (UC) is on the rise also in Japan. Simultaneously, therapeutic options, including biologics and Janus kinase (JAK) inhibitors, have significantly expanded over the past decade. Although tofacitinib (TOF), one of JAK inhibitors, is a viable option for patients with moderate to severe UC, there is insufficient data to predict responsiveness of TOF treatment. The present study aimed to determine whether the infiltration of IL-17 A-positive mononuclear cells into the colonic mucosa can predict responsiveness to TOF treatment. Patients with UC who underwent TOF treatment were divided into responder and failure groups. Subsequently, we conducted a comparative analysis to identify differences in the infiltration of IL-17 A-positive cells into the colonic mucosa through immunohistochemical examination of colon biopsy samples. The proportion of IL-17 A positive mononuclear cells in colon biopsy samples was significantly higher in the failure group than among responders (38.2% vs. 21.2%). Consistent with this finding, our re-analysis of RNA sequence datasets available in the Gene Expression Omnibus (GEO) database suggested that TOF exerts a more pronounced influence on Th1 cells compared with IL-17-producing Th17 cells. In summary, an abundance of IL-17 A-positive mononuclear cells in the colonic mucosa has the potential to predict the responsiveness to TOF treatment.
Collapse
Affiliation(s)
- Yuki Ito
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Daisuke Watanabe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan.
| | - Norihiro Okamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Haruka Miyazaki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Eri Tokunaga
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Yuna Ku
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Makoto Ooi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Michitaka Kohashi
- Department of Gastroenterology, Kakogawa Central City Hospital, Kakogawa, Hyogo, Japan
| | - Maki Kanzawa
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| |
Collapse
|
12
|
Iman K, Akter L, Rahman MM, Laila K, Islam MI, Rahman SA. Treatment of refractory poly articular course juvenile idiopathic arthritis with tofacitinib: Extended experience from Bangladesh. PLoS One 2025; 20:e0312174. [PMID: 39854407 PMCID: PMC11759992 DOI: 10.1371/journal.pone.0312174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/02/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Juvenile Idiopathic arthritis (JIA) is one of the most common chronic diseases in children. It still remains a challenge to treat refractory poly-articular course JIA patients, especially in Bangladesh, where patients from low socio-economic backgrounds are unable to manage biological agents. Tofacitinib is one of the alternative options to biological agents, which can be taken orally and is cost effective. The purpose of this prospective observational study was to evaluate the efficacy of tofacitinib in the treatment of refractory poly-articular course JIA cases. MATERIALS AND METHODS This study was carried out in the Department of Pediatrics, Bangabandhu Sheikh Mujib Medical University (BSMMU). A total number of 50 refractory polyarticular course JIA patients received JAK-2 inhibitor, tofacitinib along with other drugs according to the recommended doses. The disease activity level was measured by Juvenile Arthritis Disease Activity Score-27 (JADAS-27). All the cases were assessed at baseline, 6th, 12th and52nd week of tofacitinib therapy. The relevant statistical tests were applied for data analysis. RESULTS After treating the refractory cases with tofacitinib, arthritis subsided, and laboratory parameters improved in all the cases. Overall JADAS-27 score improvement was 40.67%, 56.38% and 96% at 6th, 12th and 52nd week of follow-up respectively. It was also possible to taper the dose of steroid gradually and stopped it by 24 weeks. Tofacitinib was well tolerated with minimum side effects. CONCLUSIONS Tofacitinib was effective to all the children with poly-articular course JIA. It was well tolerated and had very few tolerable adverse effects.
Collapse
Affiliation(s)
- Kazi Iman
- Department of Pediatrics, Dr. M R Khan Shishu Hospital & ICH, Mirpur, Dhaka, Bangladesh
| | - Laboni Akter
- Department of Pediatrics, Dhaka Medical College Hospital, Dhaka, Bangladesh
| | | | - Kamrul Laila
- Department of Pediatrics, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Mohammad Imnul Islam
- Department of Pediatrics, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Shahana A. Rahman
- Department of Pediatrics, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| |
Collapse
|
13
|
Li N, Xu T, Wu Z, Zhao Y, Ruan M, Xu H, Chen W, Wang H, Wang S, Wang Y, Liang Q. Arabinogalactan from Cynanchum atratum induces tolerogenic dendritic cells in gut to restrain autoimmune response and alleviate collagen-induced arthritis in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156269. [PMID: 39586124 DOI: 10.1016/j.phymed.2024.156269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by multiple joints lesions. Tolerogenic dendritic cells (tolDCs) play crucial roles in maintaining immune homeostasis. The immunomodulatory activity of plant-derived arabinogalactan (AGs) has been well investigated, however, whether AGs could suppress autoimmune responses by inducing tolDCs is remain unclear. DESIGN Collagen-induced arthritis (CIA, a mouse model of RA) mice were utilized to ascertain the role of AGs (obtained from Cynanchum atratum) in autoimmune responses. An antibiotic cocktail was administered to eliminate gut microbiota. Germ-free (GF) and Toll-like receptor 2 (TLR2) knockout mice were used to determine the function of AGs in intestinal immune cells. RESULTS The oral administration of dietary AGs substantially reduced the severity of CIA and rebalanced the ratio of regulatory T cells (Tregs) to T helper 17 (Th17) cells. Although the antibiotic cocktail depleted the mice's gut microbiota, AGs had a therapeutic effect on their CIA. AGs restored Treg/Th17 homeostasis by inducing CD103+ tolDCs, regardless of the gut microbiota of the GF mice. Coculture experiments confirmed that AGs induced tolDCs and transforming growth factor β (TGF-β) secretion, leading to Treg amplification. RNA sequencing and TLR2 knockout experiments revealed that AGs induced tolDCs through a TLR2-mediated mechanism. Preventive interventions with AGs established a tolerogenic intestinal immune microenvironment, which delayed the onset and progression of CIA. AGs functioned synergistically with tofacitinib, a JAK inhibitor, to effectively restore Treg/Th17 balance and alleviate CIA. CONCLUSION This study introduces a novel microbiota-independent mechanism through which soluble dietary AGs inhibit systemic autoimmune responses. Our findings provide insights into the supplementation of dietary AGs in patients with preclinical or progressive RA.
Collapse
Affiliation(s)
- Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Zhaoshun Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Yuchen Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Ming Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Weihao Chen
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Huijun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Shunchun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China.
| |
Collapse
|
14
|
Jaguan D, Nguyen KT, Goldfarb N. Selective small molecule inhibitors for hidradenitis suppurativa: Today and tomorrow. J Am Acad Dermatol 2024; 91:S31-S36. [PMID: 39626997 DOI: 10.1016/j.jaad.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 12/12/2024]
Abstract
Hidradenitis suppurativa (HS) is an autoinflammatory condition characterized by abscesses, inflammatory nodules, and tunnels in intertriginous sites of the body. The pathogenesis of HS involves follicular occlusion in combination with environmental, genetic, hormonal, and metabolic factors. HS lesions are characterized by an influx of neutrophils, histiocytes, B and T cells, and upregulation of proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1, interleukin-17, and interferons. Selective small molecule inhibitors (SMIs) are organic compounds that bind to active sites on target proteins involved in inflammatory signaling pathways, most commonly blocking enzymes, ion channels and receptors. SMIs are divided into conventional and selective SMIs. Selective SMIs are further subdivided into kinase and nonkinase SMIs. Currently there are five selective SMIs available in the United States with demonstrated efficacy for HS in clinical studies including apremilast, topical ruxolitinib, upadacitinib, fostamatinib, and sirolimus. These selective SMIs target four pathways hypothesized to be important to HS pathogenesis including phosphodiestase 4, Janus kinases, spleen tyrosine kinase, and mammalian target of rapamycin. Several new SMIs are currently in the clinical trial pipeline targeting Bruton's tyrosine kinase, aryl hydrocarbon receptors, heat shock protein 90 as well as interleukin-1 and -17 signaling pathways.
Collapse
Affiliation(s)
- Daniella Jaguan
- Georgetown University School of Medicine, Washington, District of Columbia; Department of Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Kim T Nguyen
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota; University of Nebraska Medical Center, College of Medicine, Omaha, Nebraska
| | - Noah Goldfarb
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota; Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Departments of Medicine and Dermatology, Minneapolis VA Health Care System, Minneapolis, Minnesota.
| |
Collapse
|
15
|
Mammoliti O, Menet C, Cottereaux C, Blanc J, De Blieck A, Coti G, Geney R, Oste L, Ostyn K, Palisse A, Quinton E, Schmitt B, Borgonovi M, Parent I, Jagerschmidt C, De Vos S, Vayssiere B, López-Ramos M, Shoji K, Brys R, Amantini D, Galien R, Joannesse C. Design of a potent and selective dual JAK1/TYK2 inhibitor. Bioorg Med Chem 2024; 114:117932. [PMID: 39447537 DOI: 10.1016/j.bmc.2024.117932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024]
Abstract
Janus kinase (JAK) inhibitors have gathered interest as treatments for several inflammatory and autoimmune diseases. The four first marketed inhibitors target JAK1, with varying selectivity towards other JAK family members, but none inhibit tyrosine kinase-2 (TYK2) at clinically relevant doses. TYK2 is required for the signaling of the interleukin (IL)-12 and IL-23 cytokines, which are key to the polarization of TH1 and TH17 cells, respectively; two cell subtypes that play major roles in inflammatory diseases. Herein, we report our effort towards the optimization of a potent and selective dual JAK1/TYK2 inhibitor series starting from a HTS hit. Structural information revealed vectors required to improve both JAK1 and TYK2 potency as well as selectivity towards JAK2. The potent inhibition of both JAK1 (3.5 nM) and TYK2 (5.7 nM) in biochemical assays by our optimized lead compound, as well as its notable selectivity against JAK2, were confirmed in cellular and whole blood assays. Inhibition of TYK2 by the lead compound was demonstrated by dose-dependent efficacy in an IL-23-induced psoriasis-like inflammation mouse model.
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Christel Menet
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Céline Cottereaux
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Javier Blanc
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Ann De Blieck
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Ghjuvanni Coti
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Raphaël Geney
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Line Oste
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Koen Ostyn
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Adeline Palisse
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Evelyne Quinton
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Benoit Schmitt
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Monica Borgonovi
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Isabelle Parent
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Steve De Vos
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | | | | | - Kenji Shoji
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - David Amantini
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - René Galien
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | |
Collapse
|
16
|
Talasila S, Lee E, Teichner EM, Siegfried EC, Jackson Cullison SR. Analysis of publicly available adverse events reported for pediatric patients treated with Janus kinase inhibitors. Pediatr Dermatol 2024; 41:1040-1046. [PMID: 39235110 DOI: 10.1111/pde.15721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/20/2024] [Indexed: 09/06/2024]
Abstract
Janus kinase inhibitors (JAKi) are drugs that block tyrosine kinases responsible for transducing cytokine signals. The first JAKi was approved by the US Food and Drug Administration (FDA) in 2011 to treat rheumatoid arthritis in adults. A pediatric indication was not approved until 8 years later, for acute graft-versus-host disease. Since then, topical and oral formulations have gained FDA approval for pediatric patients with dermatologic diseases. While increasing evidence supports the safety of these medications in adults, data are limited in children. We sought to determine whether JAKi adverse events (AEs) as reported in clinical trials and via postapproval pharmacovigilance services are comparable in adult and pediatric patients. Pharmacovigilance data were extracted from the FDA's Adverse Event Reporting System and the Canada Vigilance Adverse Reaction Online Database for baricitinib, upadacitinib, abrocitinib, ruxolitinib, and tofacitinib. The pooled data were analyzed to detect the most common AEs for specific JAKi and for the drug class. We assessed 399,649 AEs from 133,216 adults and 2883 AEs from 955 patients under 18 years old and identified slightly different AE profiles for the two age groups. Both populations had increased risk for infections and gastrointestinal AEs. However, pediatric patients reported a higher proportion of blood and lymphatic disorders, while reports of nervous system and musculoskeletal/connective tissue disorders were more common in adults. The spectrum of AEs extracted from pharmacovigilance reports was similar to clinical trials. The JAKi AE profiles we observed may prove helpful in counseling patients and their parents before starting therapy and for monitoring once patients are on therapy.
Collapse
Affiliation(s)
- Sahithi Talasila
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Emily Lee
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Eric M Teichner
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Elaine C Siegfried
- Saint Louis University and SSM Cardinal Glennon Children's Hospital, St. Louis, Missouri, USA
| | | |
Collapse
|
17
|
Suresh B, Reshmi PR, Jaipal J, Pise GA, Prasad SS, Manohar N. Tofacitinib in Leprosy: A Novel Therapeutic Approach in Chronic Recalcitrant Type II Reactions. Cureus 2024; 16:e74694. [PMID: 39735044 PMCID: PMC11681993 DOI: 10.7759/cureus.74694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Leprosy is a chronic, infectious, and debilitating disorder that primarily affects the skin and peripheral nerves. The disease course may be complicated by immune-mediated reactions during or after therapy, which may further worsen nerve damage. Type II lepra reaction (T2LR) is a painful inflammatory condition with systemic features, such as fever, tender erythematous nodules, arthritis, neuritis, orchitis, lymphadenitis, and iritis. Erythema nodosum leprosum (ENL), the hallmark of type II lepra reactions, results in hospitalization and consequent impairment in quality of life. The treatment options include long-term high-dose systemic corticosteroids, thalidomide, and/or clofazimine. However, the prognosis is often complicated by the adverse effects of the drugs; therefore, there is a need for alternative and safer therapies. Herein, we present the case of a 31-year-old male with recurrent lepra reactions who did not respond adequately to steroids. Therefore, we initiated therapy with tofacitinib, a non-selective inhibitor of the Janus kinase/signal transduction and transcription activation (JAK/STAT) pathway. The results included complete resolution of abnormalities on blood laboratory investigations and symptomatic resolution of symptoms. In this article, we delve into the possible role of tofacitinib in T2LR and other inflammatory conditions.
Collapse
Affiliation(s)
- Benaka Suresh
- Dermatology, Belagavi Institute of Medical Sciences, Belagavi, IND
| | - Pooja R Reshmi
- Dermatology, Belagavi Institute of Medical Sciences, Belagavi, IND
| | | | - Gajanan A Pise
- Dermatology, Belagavi Institute of Medical Sciences, Belagavi, IND
| | | | - Naveen Manohar
- Dermatology, The Oxford Medical College, Hospital, and Research Centre, Bangalore, IND
| |
Collapse
|
18
|
Kang ZP, Xiao QP, Huang JQ, Wang MX, Huang J, Wei SY, Cheng N, Wang HY, Liu DY, Zhong YB, Zhao HM. Curcumin Attenuates Dextran Sodium Sulfate Induced Colitis in Obese Mice. Mol Nutr Food Res 2024; 68:e2300598. [PMID: 39380356 DOI: 10.1002/mnfr.202300598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 06/01/2024] [Indexed: 10/10/2024]
Abstract
SCOPE Curcumin (Cur), with diverse pharmacological properties, shows anti-obesity, immunomodulatory, and anti-inflammatory effects. Its role in ulcerative colitis complicated by obesity remains unclear. METHODS AND RESULTS Here, colitis is induced in obese mice using dextran sulfate sodium (DSS), followed by administration of Cur at a dosage of 100 mg kg-1 for 14 days. Cur effectively alleviates DSS-induced colitis in obese mice, accompanied by an increase in body weight and survival rate, reduction in disease activity index, elongation of the colon, decrease in colonic weight, and improvements in ulcer formation and inflammatory cell infiltration in colonic tissues. Additionally, Cur effectively improves lipid metabolism and the composition of the gut microbiota, and enhances mucosal integrity and boosts anti-oxidative stress capacity in obese mice with colitis. Importantly, Cur is effective in improving the homeostasis of memory T cells in obese mice with colitis. Furthermore, Cur regulates inflammatory cytokines expression and inhibits activation of the JAK2/STAT signaling pathway in colonic tissues of obese mice with colitis. CONCLUSIONS Cur alleviates colitis in obese mice through a comprehensive mechanism that improves lipid metabolism, modulates gut microbiota composition, enhances mucosal integrity and anti-oxidative stress, balances memory T cell populations, regulates inflammatory cytokines, and suppresses the JAK2/STAT signaling pathway.
Collapse
Affiliation(s)
- Zeng-Ping Kang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Qiu-Ping Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Jia-Qi Huang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Meng-Xue Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Jie Huang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Si-Yi Wei
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Nian Cheng
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Hai-Yan Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - You-Bao Zhong
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| | - Hai-Mei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
- Formula-Pattern Research Center of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi Province, 330004, China
| |
Collapse
|
19
|
Virtanen A, Spinelli FR, Telliez JB, O'Shea JJ, Silvennoinen O, Gadina M. JAK inhibitor selectivity: new opportunities, better drugs? Nat Rev Rheumatol 2024; 20:649-665. [PMID: 39251770 DOI: 10.1038/s41584-024-01153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Cytokines function as communication tools of the immune system, serving critical functions in many biological responses and shaping the immune response. When cytokine production or their biological activity goes awry, the homeostatic balance of the immune response is altered, leading to the development of several pathologies such as autoimmune and inflammatory disorders. Cytokines bind to specific receptors on cells, triggering the activation of intracellular enzymes known as Janus kinases (JAKs). The JAK family comprises four members, JAK1, JAK2, JAK3 and tyrosine kinase 2, which are critical for intracellular cytokine signalling. Since the mid-2010s multiple JAK inhibitors have been approved for inflammatory and haematological indications. Currently, approved JAK inhibitors have demonstrated clinical efficacy; however, improved selectivity for specific JAKs is likely to enhance safety profiles, and different strategies have been used to accomplish enhanced JAK selectivity. In this update, we discuss the background of JAK inhibitors, current approved indications and adverse effects, along with new developments in this field. We address the issue of JAK selectivity and its relevance in terms of efficacy, and describe new modalities of JAK targeting, as well as new aspects of JAK inhibitor action.
Collapse
Affiliation(s)
- Anniina Virtanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Francesca Romana Spinelli
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza Universitá di Roma, Rome, Italy
| | | | - John J O'Shea
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olli Silvennoinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Fimlab laboratories, Tampere, Finland
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
20
|
Solimani F, Ghoreschi K. [Janus kinase inhibitors for skin disorders]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:781-790. [PMID: 39212722 DOI: 10.1007/s00105-024-05406-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Immune factors such as interferon‑ɣ and interleukin 4 belong to the group of cytokines that are dependent on type I/II receptors for their signal transmission. Upon activation, these receptors transmit their signal to the cell nucleus and, thus, modulate gene transcription via a signaling cascade consisting of Janus kinases (JAK). This family of four kinases (JAK 1, JAK 2, JAK 3, and tyrosine kinase 2 (TYK2)) subsequently activate members of the signal transducer and activator of transcription (STAT). This finding turned the JAK/STAT signaling pathway into a pharmacological target for the treatment of inflammatory diseases in which cytokines using type I/II receptors play a pathogenic role. In 2018, the European Medicines Agency (EMA) approved tofacitinib for the treatment of psoriatic arthritis. This was the first approval of a JAK/STAT pathway inhibitor for patients treated by dermatologists and rheumatologists. Since then, several new JAK inhibitors have been approved for dermatologic diseases such as atopic dermatitis, alopecia areata, vitiligo, and plaque-type psoriasis. In addition, JAK inhibitors are being investigated for the treatment of many other skin diseases. Thus, systemic JAK inhibitors complete the spectrum of immunotherapeutics with a broader immunological approach compared to monoclonal antibodies. The low molecular weight of JAK inhibitors enables the preparation of these drugs for both systemic and topical administration. Their utilization could represent a valuable alternative to topical steroids. The safety profile of JAK inhibitors must be taken into account. Possible long-term effects may become apparent in the next few years. This article describes both approved JAK inhibitors and relevant new JAK inhibitors that are promising candidates for approval as therapeutics in dermatology.
Collapse
Affiliation(s)
- Farzan Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Luisenstr. 2, 10117, Berlin, Deutschland.
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Deutschland.
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Luisenstr. 2, 10117, Berlin, Deutschland
| |
Collapse
|
21
|
Abdelnabi MN, Hassan GS, Shoukry NH. Role of the type 3 cytokines IL-17 and IL-22 in modulating metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1437046. [PMID: 39156888 PMCID: PMC11327067 DOI: 10.3389/fimmu.2024.1437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.
Collapse
Affiliation(s)
- Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
22
|
Kiełbowski K, Plewa P, Bratborska AW, Bakinowska E, Pawlik A. JAK Inhibitors in Rheumatoid Arthritis: Immunomodulatory Properties and Clinical Efficacy. Int J Mol Sci 2024; 25:8327. [PMID: 39125897 PMCID: PMC11311960 DOI: 10.3390/ijms25158327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent autoimmune disorder. The pathogenesis of the disease is complex and involves various cellular populations, including fibroblast-like synoviocytes, macrophages, and T cells, among others. Identification of signalling pathways and molecules that actively contribute to the development of the disease is crucial to understanding the mechanisms involved in the chronic inflammatory environment present in affected joints. Recent studies have demonstrated that the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway regulates the behaviour of immune cells and contributes to the progression of RA. Several JAK inhibitors, such as tofacitinib, baricitinib, upadacitinib, and filgocitinib, have been developed, and their efficacy and safety in patients with RA have been comprehensively investigated in a number of clinical trials. Consequently, JAK inhibitors have been approved and registered as a treatment for patients with RA. In this review, we discuss the involvement of JAK/STAT signalling in the pathogenesis of RA and summarise the potential beneficial effects of JAK inhibitors in cells implicated in the pathogenesis of the disease. Moreover, we present the most important phase 3 clinical trials that evaluated the use of these agents in patients.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| |
Collapse
|
23
|
Jarneborn A, Hu Z, Deshmukh M, Kopparapu PK, Jin T. Tofacitinib Treatment Suppresses CD4+ T-Cell Activation and Th1 Response, Contributing to Protection against Staphylococcal Toxic Shock. Int J Mol Sci 2024; 25:7456. [PMID: 39000566 PMCID: PMC11242597 DOI: 10.3390/ijms25137456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Staphylococcal toxic shock syndrome (STSS) is a rare, yet potentially fatal disease caused by Staphylococcus aureus (S. aureus) enterotoxins, known as superantigens, which trigger an intense immune response. Our previous study demonstrated the protective effect of tofacitinib against murine toxin-induced shock and a beneficial effect against S. aureus sepsis. In the current study, we examined the effects of tofacitinib on T-cell response in peripheral blood using a mouse model of enterotoxin-induced shock. Our data revealed that tofacitinib suppresses the activation of both CD4+ and CD8+ T cells in peripheral blood. Furthermore, both gene and protein levels of Th1 cytokines were downregulated by tofacitinib treatment in mice with enterotoxin-induced shock. Importantly, we demonstrated that CD4+ cells, but not CD8+ cells, are pathogenic in mice with enterotoxin-induced shock. In conclusion, our findings suggest that tofacitinib treatment suppresses CD4+ T-cell activation and Th1 response, thereby aiding in protection against staphylococcal toxic shock in mice. This insight may guide the future development of novel therapies for STSS.
Collapse
Affiliation(s)
- Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
| | - Meghshree Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| |
Collapse
|
24
|
Michels A, Heiland R, Hammerschmidt S, Farcas A, Voigt TP, Braun SA, Metze D, Tsianakas A. Erfolgreiche Behandlung eines therapieresistenten generalisierten Granuloma anulare mit dem JAK‐Inhibitor Abrocitinib. J Dtsch Dermatol Ges 2024; 22:841-843. [PMID: 38857107 DOI: 10.1111/ddg.15426_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 01/11/2024] [Indexed: 06/11/2024]
Affiliation(s)
| | - Rebecca Heiland
- Klinik für Dermatologie und Allergologie, Fachklinik Bad Bentheim
| | | | - Alexandra Farcas
- Klinik für Dermatologie und Allergologie, Fachklinik Bad Bentheim
| | | | | | - Dieter Metze
- Klinik für Hautkrankheiten, Universitätsklinikum Münster
| | | |
Collapse
|
25
|
Michels A, Heiland R, Hammerschmidt S, Farcas A, Voigt TP, Braun SA, Metze D, Tsianakas A. Successful treatment of recalcitrant generalized granuloma annulare with the JAK inhibitor abrocitinib. J Dtsch Dermatol Ges 2024; 22:841-843. [PMID: 38491389 DOI: 10.1111/ddg.15426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 01/11/2024] [Indexed: 03/18/2024]
Affiliation(s)
- Alexander Michels
- Department of Dermatology and Allergology, Specialist Hospital Bad Bentheim, Bad Bentheim, Germany
| | - Rebecca Heiland
- Department of Dermatology and Allergology, Specialist Hospital Bad Bentheim, Bad Bentheim, Germany
| | - Stephanie Hammerschmidt
- Department of Dermatology and Allergology, Specialist Hospital Bad Bentheim, Bad Bentheim, Germany
| | - Alexandra Farcas
- Department of Dermatology and Allergology, Specialist Hospital Bad Bentheim, Bad Bentheim, Germany
| | - Tilman Peter Voigt
- Department of Dermatology and Allergology, Specialist Hospital Bad Bentheim, Bad Bentheim, Germany
| | | | - Dieter Metze
- Department of Dermatology, University Hospital Muenster, Muenster, Germany
| | - Athanasios Tsianakas
- Department of Dermatology and Allergology, Specialist Hospital Bad Bentheim, Bad Bentheim, Germany
| |
Collapse
|
26
|
Sohrabi S, Masoumi J, Naseri B, Ghorbaninezhad F, Alipour S, Kazemi T, Ahmadian Heris J, Aghebati Maleki L, Basirjafar P, Zandvakili R, Doustvandi MA, Baradaran B. STATs signaling pathways in dendritic cells: As potential therapeutic targets? Int Rev Immunol 2024; 43:138-159. [PMID: 37886903 DOI: 10.1080/08830185.2023.2274576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/17/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APCs), including heterogenous populations with phenotypic and functional diversity that coordinate bridging innate and adaptive immunity. Signal transducer and activator of transcriptions (STAT) factors as key proteins in cytokine signaling were shown to play distinct roles in the maturation and antigen presentation of DCs and play a pivotal role in modulating immune responses mediated by DCs such as differentiation of T cells to T helper (Th) 1, Th2 or regulatory T (Treg) cells. This review sheds light on the importance of STAT transcription factors' signaling pathways in different subtypes of DCs and highlights their targeting potential usages for improving DC-based immunotherapies for patients who suffer from cancer or diverse autoimmune conditions according to the type of the STAT transcription factor and its specific activating or inhibitory agent.
Collapse
Affiliation(s)
- Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Pedram Basirjafar
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Raziyeh Zandvakili
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Furtunescu AR, Georgescu SR, Tampa M, Matei C. Inhibition of the JAK-STAT Pathway in the Treatment of Psoriasis: A Review of the Literature. Int J Mol Sci 2024; 25:4681. [PMID: 38731900 PMCID: PMC11083046 DOI: 10.3390/ijms25094681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Psoriasis is a highly prevalent dermatological disease associated with an increased systemic inflammatory response. In addition, joint involvement is also present in around 20% of patients. Therefore, treatment modalities used in this condition should be simultaneously effective at improving skin manifestations, reducing inflammation, and addressing psoriatic arthritis when present. Twenty years ago, the introduction of biologic treatments for psoriasis was a turning point in the management of this condition, offering an effective and reasonably safe option for patients whose disease could not be adequately controlled with conventional therapies. At the moment, Janus Kinase inhibitors (JAKis) are a new class of promising molecules in the management of psoriasis. They are orally administered and can show benefits in patients who failed biologic therapy. We conducted a scoping review in order to identify randomized-controlled trials that investigated different JAKis in patients with plaque psoriasis and psoriatic arthritis, with an emphasis on molecules that have been approved by the European Medicines Agency and the Food and Drug Administration. The added value of this study is that it collected information about JAKis approved for two different indications, plaque psoriasis and psoriatic arthritis, in order to provide an integrated understanding of the range of effects that JAKis have on the whole spectrum of psoriasis manifestations.
Collapse
Affiliation(s)
- Andreea Roxana Furtunescu
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| | - Simona Roxana Georgescu
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| | - Mircea Tampa
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| | - Clara Matei
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
28
|
Skorupan N, Peer CJ, Zhang X, Choo-Wosoba H, Ahmad MI, Lee MJ, Rastogi S, Sato N, Yu Y, Pegna GJ, Steinberg SM, Kalsi SS, Cao L, Figg WD, Trepel JB, Pastan I, FitzGerald D, Alewine C. Tofacitinib to prevent anti-drug antibody formation against LMB-100 immunotoxin in patients with advanced mesothelin-expressing cancers. Front Oncol 2024; 14:1386190. [PMID: 38706610 PMCID: PMC11066227 DOI: 10.3389/fonc.2024.1386190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024] Open
Abstract
Background LMB-100 is a mesothelin (MSLN)-targeting recombinant immunotoxin (iTox) carrying a Pseudomonas exotoxin A payload that has shown promise against solid tumors, however, efficacy is limited by the development of neutralizing anti-drug antibodies (ADAs). Tofacitinib is an oral Janus Kinase (JAK) inhibitor that prevented ADA formation against iTox in preclinical studies. Methods A phase 1 trial testing LMB-100 and tofacitinib in patients with MSLN-expressing cancers (pancreatic adenocarcinoma, n=13; cholangiocarcinoma, n=1; appendiceal carcinoma, n=1; cystadenocarcinoma, n=1) was performed to assess safety and to determine if tofacitinib impacted ADA formation. Participants were treated for up to 3 cycles with LMB-100 as a 30-minute infusion on days 4, 6, and 8 at two dose levels (100 and 140 µg/kg) while oral tofacitinib was administered for the first 10 days of the cycle (10 mg BID). Peripheral blood was collected for analysis of ADA levels, serum cytokines and circulating immune subsets. Results The study was closed early due to occurrence of drug-induced pericarditis in 2 patients. Pericarditis with the combination was not reproducible in a transgenic murine model containing human MSLN. Two of 4 patients receiving all 3 cycles of treatment maintained effective LMB-100 levels, an unusual occurrence. Sustained increases in systemic IL-10 and TNF-α were seen, a phenomenon not observed in prior LMB-100 studies. A decrease in activated T cell subsets and an increase in circulating immunosuppressive myeloid populations occurred. No radiologic decreases in tumor volume were observed. Discussion Further testing of tofacitinib to prevent ADA formation is recommended in applicable non-malignant disease settings. Clinical trial registration https://www.clinicaltrials.gov/study/NCT04034238.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xianyu Zhang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mehwish I. Ahmad
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shraddha Rastogi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nahoko Sato
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Guillaume Joe Pegna
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shelley S. Kalsi
- Hematology Consult and Graduate Medical Section, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David FitzGerald
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christine Alewine
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
29
|
Liu C, Liu X, Xin H, Li X. A Mendelian randomization study on the causal effects of circulating cytokines on the risk of vitiligo. Front Med (Lausanne) 2024; 11:1375339. [PMID: 38695020 PMCID: PMC11061512 DOI: 10.3389/fmed.2024.1375339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/12/2024] [Indexed: 05/04/2024] Open
Abstract
Background Accumulating evidence reveals an association between circulating cytokine levels and vitiligo. However, the causal association between circulating cytokine levels and vitiligo remains unrevealed. Methods We performed a two-sample Mendelian randomization (MR) analysis using a genome-wide association study of the 41 cytokines dataset, which was conducted with 3 Finnish cohorts (n = 8,293). Vitiligo data were acquired from strictly defined vitiligo data collected by FinnGenbiobank analysis, which included 207,613 European ancestors (131 vitiligo patients, 207,482 controls). The inverse-variance weighted (IVW) method, weighted median (WME), simple model, weighted model, and MR-Egger were used to determine the changes in vitiligo pathogenic cytokine taxa, followed by sensitivity analysis, including horizontal pleiotropy analysis. The MR Steiger test evaluated the strength of a causal association, and the leave-one-out method was used to assess the reliability of the results. The possibility of reverse causality was also investigated using a reverse MR study. Results We observed that rising IL-4 levels generated an enhanced probability of vitiligo in IVW (OR 2.72, 95%CI 1.19-6.22, p = 0.018). According to the results of the MR analysis, there were causal links between IL-4 and vitiligo. Results were steady after sensitivity and heterogeneity analyses. Conclusion Our research reveals that a genetically determined increased level of circulating IL-4 may be linked to a higher risk of developing vitiligo. The development of innovative treatment approaches (such as tofacitinib or dupilumab) that focus on blocking IL-4 as a novel way of preventing and treating vitiligo is significantly impacted by our findings.
Collapse
Affiliation(s)
- Chengling Liu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the PLA, Guilin, China
| | - Xingchen Liu
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Haiming Xin
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the PLA, Guilin, China
| | - Xin Li
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the PLA, Guilin, China
| |
Collapse
|
30
|
Luo F, Zhang Y, Wang P. Tofacitinib for the treatment of severe rare skin diseases: a narrative review. Eur J Clin Pharmacol 2024; 80:481-492. [PMID: 38231227 DOI: 10.1007/s00228-024-03621-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024]
Abstract
PURPOSE Autoimmune bullous diseases, connective tissue diseases, and vasculitis represent a group of severe rare skin diseases. While glucocorticoids and immunosuppressive agents serve as standard treatments for these diseases, their efficacy is limited due to adverse side effects, indicating the need for alternative approaches. Biologics have been used in the management of some rare skin diseases. However, the use of biologics is associated with concerns, such as infection risk and high costs, prompting the quest for efficacious and cost-effective alternatives. This study discusses the safety issues associated with tofacitinib and its potential in treating rare skin diseases. METHODS This narrative review focuses on the pharmacodynamic properties of tofacitinib and its impact on the JAK/STAT pathway. In addition, we present a comprehensive discussion of the effects and mechanism of action of tofacitinib for each severe rare skin disease. RESULTS This role of tofacitinib in treating severe rare skin diseases has been discussed, shedding light on its promising prospects as a treatment modality. Few reports of serious adverse events are available in patients treated with tofacitinib. CONCLUSION We explored the mechanism of action, efficacy, and safety considerations of tofacitinib and found that it can be used as a treatment option for rare skin diseases. However, multicenter clinical studies are needed to confirm the efficacy and safety of JAK inhibitors.
Collapse
Affiliation(s)
- Fenglin Luo
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310000, China
| | - Yuanyuan Zhang
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310000, China
| | - Ping Wang
- Department of Dermatology, Hangzhou Third People's Hospital, Hangzhou, 310000, China.
| |
Collapse
|
31
|
Eichinger JM, Shan DM, Greenzaid JD, Anakwenze L, Feldman SR. Clinical pharmacokinetics and pharmacodynamics of oral systemic nonbiologic therapies for psoriasis patients. Expert Opin Drug Metab Toxicol 2024; 20:249-262. [PMID: 38529623 DOI: 10.1080/17425255.2024.2335310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
INTRODUCTION Psoriasis is a chronic inflammatory immune condition. Treatments for psoriasis vary with disease severity, ranging from topicals to systemic biologic agents. The pharmacokinetic (PK) and pharmacodynamic (PD) properties of these therapies establish drug efficacy, toxicity, and optimal dosing to ensure therapeutic drug levels are sustained and adverse effects are minimized. AREAS COVERED A literature search was performed on PubMed, Google Scholar, and Ovid MEDLINE for PK and PD, efficacy, and safety data regarding oral systemic nonbiologic therapies utilized for moderate-to-severe plaque psoriasis. The findings were organized into sections for each drug: oral acitretin, methotrexate, cyclosporine, apremilast, tofacitinib, and deucravacitinib. EXPERT OPINION Some psoriasis patients may not respond to initial therapy. Ongoing research is evaluating genetic polymorphisms that may predict an improved response to specific medications. However, financial and insurance barriers, as well as limited genetic polymorphisms correlated with treatment response, may restrict the implementation of genetic testing necessary to personalize treatments. How well psoriasis patients adhere to treatment may contribute greatly to variation in response. Therapeutic drug monitoring may help patients adhere to treatment, improve clinical response, and sustain disease control.
Collapse
Affiliation(s)
| | - Divya M Shan
- Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Jonathan D Greenzaid
- Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lisa Anakwenze
- University of Louisville School of Medicine, Louisville, KY, USA
| | - Steven R Feldman
- Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Dermatology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
32
|
Arzivian A, Zhang E, Laube R, Leong R. First-trimester exposure to tofacitinib in ulcerative colitis: A case report of a healthy newborn and literature review. Clin Case Rep 2024; 12:e8764. [PMID: 38617066 PMCID: PMC11014800 DOI: 10.1002/ccr3.8764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024] Open
Abstract
Tofacitinib is contraindicated in pregnancy. We present a patient with ulcerative colitis on tofacitinib who had an unplanned pregnancy. Tofacitinib was ceased, switched to vedolizumab, and she gave birth to a healthy newborn at term. Case reports of reassuring outcomes provide real-world data that assists decision-making for future patients.
Collapse
Affiliation(s)
- Arteen Arzivian
- Department of Gastroenterology and HepatologyMacquarie University HospitalSydneyNew South WalesAustralia
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical SchoolMacquarie UniversitySydneyNew South WalesAustralia
| | - Eva Zhang
- Department of Gastroenterology and HepatologyMacquarie University HospitalSydneyNew South WalesAustralia
| | - Robyn Laube
- Department of Gastroenterology and HepatologyMacquarie University HospitalSydneyNew South WalesAustralia
| | - Rupert Leong
- Department of Gastroenterology and HepatologyMacquarie University HospitalSydneyNew South WalesAustralia
| |
Collapse
|
33
|
Dehghani P, Varshosaz J, Mirian M, Minaiyan M, Kazemi M, Bodaghi M. Keratinocyte Exosomes for Topical Delivery of Tofacitinib in Treatment of Psoriasis: an In Vitro/ In Vivo Study in Animal Model of Psoriasis. Pharm Res 2024; 41:263-279. [PMID: 38263341 PMCID: PMC10879239 DOI: 10.1007/s11095-023-03648-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024]
Abstract
INTRODUCTION Exosomes are extracellular vesicles in the range of 40-150 nm released from the cell membrane. Exosomes secreted by keratinocytes can communicate with other keratinocytes and immune cells with specific biomarkers at their surface, which may be effective on inflammation of psoriasis and its pathogenesis. OBJECTIVE The present study aimed to formulate and study effectiveness of an exosomal delivery system of tofacitinib (TFC). METHODS TFC was loaded by different methods in exosomes and then characterized for particle size, zeta potential, drug loading efficiency, and release efficiency. By comparing these parameters, the probe sonication method was chosen to load TFC into exosomes. The MTT assay was used to compare the cytotoxicity of the free drug with the TFC-loaded exosomes (TFC-Exo), and Real-time PCR was used to determine the expression levels of several genes involved in psoriasis expressed in the A-431 keratinocyte and their suppression after treatment. Animal model of psoriasis was induced in BALB/c mice by imiquimod and the efficacy of free TFC, and TFC-Exo were studies on macroscopic appearance and histopathological symptoms. RESULTS Exosomes encapsulating TFC showed lower cytotoxicity in MTT assay, higher suppression the expression of TNF-a, IL-23, IL-6, and IL-15 genes in real-time PCR and better therapeutic effect on animal models compered to free TFC. CONCLUSIONS This method of drug delivery for TFC may be effective on enhancing its therapeutic effects and reduction its side effects favorably in chronic administration.
Collapse
Affiliation(s)
- Pouya Dehghani
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Isfahan University of Medical Sciences, PO Box 81745-359, Isfahan, Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Isfahan University of Medical Sciences, PO Box 81745-359, Isfahan, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Minaiyan
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Reproductive Sciences and Sexual Health Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdi Bodaghi
- Department of Engineering School of Science and Technology Nottingham Trent University, Nottingham, NG11 8NS, UK.
| |
Collapse
|
34
|
Cuccia G, Privitera G, Di Vincenzo F, Monastero L, Parisio L, Carbone L, Scaldaferri F, Pugliese D. Predictors of Efficacy of Janus Kinase Inhibitors in Patients Affected by Ulcerative Colitis. J Clin Med 2024; 13:766. [PMID: 38337460 PMCID: PMC10856140 DOI: 10.3390/jcm13030766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Personalised medicine and the identification of predictors of the efficacy of specific drugs represent the ultimate goal for the treatment of ulcerative colitis (UC) in order to break the current therapeutic ceiling. JAK inhibitors are a new class of advanced therapies, orally administered, showing a good profile of efficacy and safety in both randomised controlled trials (RCTs) and real-world studies. Unfortunately, to date, it is not possible to draw the ideal profile of a patient maximally benefiting from this class of drugs to guide clinicians' therapeutic choices. Baseline clinical activities and inflammatory biomarkers, as well as their early variation after treatment initiation, emerged as the main predictors of efficacy from post hoc analyses of RCTs with tofacitinib. Similar findings were also observed in the real-life studies including mainly patients with a history of pluri-refractoriness to biological therapies. At last, a few new biomarkers have been explored, even though they have not been validated in large cohorts. This paper provides a review of the current knowledge on clinical variables and biomarkers predicting response to JAK inhibitors in UC.
Collapse
Affiliation(s)
- Giuseppe Cuccia
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, L. Go A. Gemelli 8, 00168 Rome, Italy; (G.C.); (F.D.V.); (L.M.); (F.S.)
| | - Giuseppe Privitera
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20122 Milan, Italy;
| | - Federica Di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, L. Go A. Gemelli 8, 00168 Rome, Italy; (G.C.); (F.D.V.); (L.M.); (F.S.)
| | - Lucia Monastero
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, L. Go A. Gemelli 8, 00168 Rome, Italy; (G.C.); (F.D.V.); (L.M.); (F.S.)
| | - Laura Parisio
- IBD UNIT-CEMAD (Centro Malattie Apparato Digerente), Medicina Interna e Gastroenterologia, Fondazione Policlinico A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Luigi Carbone
- UOC Pronto Soccorso, Medicina d’Urgenza e Medicina Interna, Ospedale Isola Tiberina Gemelli Isola, 00186 Rome, Italy;
| | - Franco Scaldaferri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, L. Go A. Gemelli 8, 00168 Rome, Italy; (G.C.); (F.D.V.); (L.M.); (F.S.)
- IBD UNIT-CEMAD (Centro Malattie Apparato Digerente), Medicina Interna e Gastroenterologia, Fondazione Policlinico A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Daniela Pugliese
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, L. Go A. Gemelli 8, 00168 Rome, Italy; (G.C.); (F.D.V.); (L.M.); (F.S.)
- IBD UNIT-CEMAD (Centro Malattie Apparato Digerente), Medicina Interna e Gastroenterologia, Fondazione Policlinico A. Gemelli IRCCS, 00168 Rome, Italy;
- UOS Gastroenterologia, Ospedale Isola Tiberina Gemelli Isola, 00186 Rome, Italy
| |
Collapse
|
35
|
Bonelli M, Kerschbaumer A, Kastrati K, Ghoreschi K, Gadina M, Heinz LX, Smolen JS, Aletaha D, O'Shea J, Laurence A. Selectivity, efficacy and safety of JAKinibs: new evidence for a still evolving story. Ann Rheum Dis 2024; 83:139-160. [PMID: 37923366 PMCID: PMC10850682 DOI: 10.1136/ard-2023-223850] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/18/2023] [Indexed: 11/07/2023]
Abstract
Fundamental insight gained over the last decades led to the discovery of cytokines as pivotal drivers of inflammatory diseases such as rheumatoid arthritis, psoriasis/psoriasis arthritis, inflammatory bowel diseases, atopic dermatitis and spondylarthritis. A deeper understanding of the pro-inflammatory and anti-inflammatory effects of various cytokines has prompted new cytokine-targeting therapies, which revolutionised the treatment options in the last years for patients with inflammatory disorders. Disease-associated immune responses typically involve a complex interplay of multiple cytokines. Therefore, blockade of one single cytokine does not necessarily lead to a persistent remission in all patients with inflammatory disorders and fostered new therapeutic strategies targeting intracellular pathways shared by multiple cytokines. By inhibiting JAK-STAT signalling pathways common to families of cytokines, JAK-inhibitors (JAKinibs) have created a new paradigm for the treatment of inflammatory diseases. Multiple agents have been approved for various disorders and more are being investigated for several new indications. Second-generation selective JAKinibs have been devised with the aim to achieve an increased selectivity and a possible reduced risk of side effects. In the current review, we will summarise the current body of evidence of pan versus selective JAKinibs and the most recent insights on new side effects and indications, including COVID-19.
Collapse
Affiliation(s)
- Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kastriot Kastrati
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Massimo Gadina
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - John O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Arian Laurence
- Translational Gastroenterology Unit, Department of Haematology, University College Hospital, UCLH Hospitals NHS Trust, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Leal I, Steeples LR, Wong SW, Giuffrè C, Pockar S, Sharma V, Green EKY, Payne J, Jones NP, Chieng ASE, Ashworth J. Update on the systemic management of noninfectious uveitis in children and adolescents. Surv Ophthalmol 2024; 69:103-121. [PMID: 36682467 DOI: 10.1016/j.survophthal.2023.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/22/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023]
Abstract
Noninfectious uveitis (NIU) in children and adolescents is a rare but treatable cause of visual impairment in children. Treatments for pediatric NIU and their side effects, along with the risks of vision loss and the need for long-term disease monitoring, pose significant challenges for young patients and their families. Treatment includes local and systemic approaches and this review will focus on systemic therapies that encompass corticosteroids, conventional synthetic disease-modifying antirheumatic drugs (csDMARD), and biological disease-modifying antirheumatic drugs (bDMARD). Treatment is generally planned in a stepwise approach. Methotrexate is well-established as the preferential csDMARD in pediatric NIU. Adalimumab, an antitumor necrosis factor (TNF) agent, is the only bDMARD formally approved for pediatric NIU and has a good safety and efficacy profile. Biosimilars are gaining increasing visibility in the treatment of pediatric NIU. Other bDMARD with some evidence in literature for the treatment of pediatric NIU include infliximab, tocilizumab, abatacept, rituximab and, more recently, Janus kinase inhibitors. Important aspects of managing children on these systemic therapies include vaccination issues, risk of infection, and psychological distress. Also, strategies need to address regarding primary nonresponse/secondary loss of response to anti-TNF treatment, biological switching, and monitoring regimens for these drugs. Optimal management of pediatric uveitis involves a multidisciplinary team, including specialist pediatric uveitis and rheumatology nurses, pediatric rheumatologists, psychological support, orthoptic and optometry support, and play specialists.
Collapse
Affiliation(s)
- Inês Leal
- Ophthalmology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal; Visual Sciences Study Centre, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Laura R Steeples
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK; Academic Health Science Centre, Manchester, UK
| | - Shiao Wei Wong
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Chiara Giuffrè
- Centro Europeo di Oftalmologia, Palermo, Italy; Ophthalmology Department, San Raffaele Scientific Institute, University Vita-Salute, Milan, Italy
| | - Sasa Pockar
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Vinod Sharma
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Elspeth K Y Green
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Janine Payne
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Nicholas P Jones
- School of Biological Sciences, University of Manchester, Manchester, UK
| | | | - Jane Ashworth
- Manchester Royal Eye Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, UK; Division of Evolution & Genomic Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
37
|
Davoutis E, Panou C, Stachika N, Dalla C, Kokras N. Drug-drug interactions between COVID-19 drug therapies and antidepressants. Expert Opin Drug Metab Toxicol 2023; 19:937-950. [PMID: 37934891 DOI: 10.1080/17425255.2023.2280750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Antidepressants are widely used for the pharmacological treatment of anxiety and mood disorders. Since the eruption of the SARS-COV-2 pandemic and the later development of targeted treatments against COVID-19, inevitably many patients receive antidepressants as well as targeted treatments against COVID-19 against COVID-19. Co-administration of antidepressants with COVID-19 therapeutics has the potential of drug-drug interactions, of varying severity and clinical significance. AREAS COVERED This is a curated narrative review of the current state of the art regarding drug-drug interactions between COVID-19 therapeutics and medications licensed for the pharmacotherapy of depression. A systematic search of electronic databases, using as keywords the international nonproprietaty names of currently approved COVID-19 therapeutics and antidepressants was performed, and additionally online interaction checker tools were consulted. Derived data were synthesized for each COVID-19 therapeutic and presented with up-to-date guidance. EXPERT OPINION Several COVID-19 therapeutics have potential for drug-drug interactions with antidepressants. Remdesivir and Nirmatrelvir-Ritonavir have the higher risk, whereas several monoclonal antibodies appear safer. The most serious drug-drug interactions (serotonin syndrome and QTc prolongation) require close monitoring; however, DDI toward reducing the efficacy of antidepressants may be difficult to recognize. As COVID-19 treatment protocols take precedence, psychiatrists should exert flexibility in antidepressant use and proactively monitor treatment progress.
Collapse
Affiliation(s)
- Efstathia Davoutis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Chrysa Panou
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolina Stachika
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
38
|
Castelo-Soccio L, Kim H, Gadina M, Schwartzberg PL, Laurence A, O'Shea JJ. Protein kinases: drug targets for immunological disorders. Nat Rev Immunol 2023; 23:787-806. [PMID: 37188939 PMCID: PMC10184645 DOI: 10.1038/s41577-023-00877-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/17/2023]
Abstract
Protein kinases play a major role in cellular activation processes, including signal transduction by diverse immunoreceptors. Given their roles in cell growth and death and in the production of inflammatory mediators, targeting kinases has proven to be an effective treatment strategy, initially as anticancer therapies, but shortly thereafter in immune-mediated diseases. Herein, we provide an overview of the status of small molecule inhibitors specifically generated to target protein kinases relevant to immune cell function, with an emphasis on those approved for the treatment of immune-mediated diseases. The development of inhibitors of Janus kinases that target cytokine receptor signalling has been a particularly active area, with Janus kinase inhibitors being approved for the treatment of multiple autoimmune and allergic diseases as well as COVID-19. In addition, TEC family kinase inhibitors (including Bruton's tyrosine kinase inhibitors) targeting antigen receptor signalling have been approved for haematological malignancies and graft versus host disease. This experience provides multiple important lessons regarding the importance (or not) of selectivity and the limits to which genetic information informs efficacy and safety. Many new agents are being generated, along with new approaches for targeting kinases.
Collapse
Affiliation(s)
- Leslie Castelo-Soccio
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanna Kim
- Juvenile Myositis Pathogenesis and Therapeutics Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela L Schwartzberg
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arian Laurence
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK.
- University College London Hospitals NHS Foundation Trust, London, UK.
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Kim Y, Yang HI, Kim KS. Etiology and Pathogenesis of Rheumatoid Arthritis-Interstitial Lung Disease. Int J Mol Sci 2023; 24:14509. [PMID: 37833957 PMCID: PMC10572849 DOI: 10.3390/ijms241914509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/19/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Interstitial lung disease (ILD) is one of the most serious extra-articular complications of rheumatoid arthritis (RA), which increases the mortality of RA. Because the pathogenesis of RA-ILD remains poorly understood, appropriate therapeutic strategies and biomarkers have not yet been identified. Thus, the goal of this review was to summarize and analyze the reported data on the etiology and pathogenesis of RA-ILD. The incidence of RA-ILD increases with age, and is also generally higher in men than in women and in patients with specific genetic variations and ethnicity. Lifestyle factors associated with an increased risk of RA-ILD include smoking and exposure to pollutants. The presence of an anti-cyclic citrullinated peptide antibody, high RA disease activity, and rheumatoid factor positivity also increase the risk of RA-ILD. We also explored the roles of biological processes (e.g., fibroblast-myofibroblast transition, epithelial-mesenchymal transition, and immunological processes), signaling pathways (e.g., JAK/STAT and PI3K/Akt), and the histopathology of RA involved in RA-ILD pathogenesis based on published preclinical and clinical models of RA-ILD in animal and human studies.
Collapse
Affiliation(s)
- Yerin Kim
- Department of Medicine, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea;
| | - Hyung-In Yang
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Kyoung-Soo Kim
- East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| |
Collapse
|
40
|
Li H, Wang H, Qiao G, Liu Y, Zhang F, Pan F. Concurrent bullous pemphigoid and psoriasis vulgaris successfully treated with Janus kinase inhibitor tofacitinib: A case report and review of the literature. Int Immunopharmacol 2023; 122:110591. [PMID: 37441809 DOI: 10.1016/j.intimp.2023.110591] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
Bullous pemphigoid (BP) and psoriasis are both immune-related skin diseases. Still, the comorbidities between the two are rare, and there is no consensus on the optimal treatment strategy for BP combined with psoriasis. JAK inhibitors are emerging, molecularly targeted therapeutic agents that target the molecule of Janus kinase, a signal transducer and activator of transcription (JAK/STAT). JAK inhibitors block intracellular signaling pathways by blocking the gene transcription of key pro-inflammatory cytokines that play a central role in the pathogenesis of many inflammatory and autoimmune diseases. Tofacitinib is a first-generation JAK inhibitor. The purpose of this article is to describe the first report of the use of tofacitinib in treating BP combined with psoriasis vulgaris with significant results. According to our findings, tofacitinib may be a safe and effective treatment option for patients suffering from BP and psoriasis together. The implications of this are substantial for the guidance of treatment strategies for both comorbid conditions.
Collapse
Affiliation(s)
- Hongda Li
- Shandong University of Traditional Chinese Medicine, People's Republic of China; Shandong Provincial Hospital for Skin Diseases, Shandong First Medical University, People's Republic of China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, People's Republic of China
| | - Honglei Wang
- Shandong Provincial Hospital for Skin Diseases, Shandong First Medical University, People's Republic of China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, People's Republic of China
| | - Guizhi Qiao
- Jinan Dermatology Prevention and Treatment Hospital, People's Republic of China
| | - Yongxia Liu
- Shandong Provincial Hospital for Skin Diseases, Shandong First Medical University, People's Republic of China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, People's Republic of China
| | - Furen Zhang
- Shandong University of Traditional Chinese Medicine, People's Republic of China; Shandong Provincial Hospital for Skin Diseases, Shandong First Medical University, People's Republic of China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, People's Republic of China.
| | - Futang Pan
- Shandong Provincial Hospital for Skin Diseases, Shandong First Medical University, People's Republic of China; Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, People's Republic of China.
| |
Collapse
|
41
|
Laux J, Martorelli M, Späth N, Maier F, Burnet M, Laufer SA. Selective Inhibitors of Janus Kinase 3 Modify Responses to Lipopolysaccharides by Increasing the Interleukin-10-to-Tumor Necrosis Factor α Ratio. ACS Pharmacol Transl Sci 2023; 6:892-906. [PMID: 37325444 PMCID: PMC10262334 DOI: 10.1021/acsptsci.3c00043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Indexed: 06/17/2023]
Abstract
Janus kinase (JAK) inhibitors act at low doses (e.g., tofacitinib, 0.2-0.4 μmol/kg bid) in clinical use, suggesting an efficient underlying mode of action. We hypothesized that their effectiveness is due to their ability to raise the ratio of IL-10 to TNFα. Unlike other JAK isoforms, JAK3 is expressed mainly in hematopoietic cells and is essential for immune function. We used JAK3 selective inhibitors with preferential distribution to immune cells. Inhibition of JAK3 in human leukocytes reduced TNFα and IL-6 but maintained levels of IL-10, while pan-JAK inhibitors increased TNFα, IL-6, and IL-10. JAK1 is required for IL-10 receptor signaling, which suggests that, at exposure above the IC50 (55 nM for tofacitinib on JAK1), there is less feedback control of TNFα levels. This leads to self-limiting effects of JAK1 inhibitors and could place an upper limit on appropriate doses. In vivo, treating mice with JAK3 inhibitors before LPS administration decreased plasma TNFα and increased IL-10 above vehicle levels, suggesting that JAK3 inhibition may limit TNFα release by increasing IL-10 while leaving the IL-10 receptor functional. This mechanism should have general utility in controlling autoimmune diseases and can be conveniently observed by measuring the ratio of IL-10 to TNFα. In summary, our targeted, "leukotropic" inhibitors more effectively increased IL-10/TNFα ratios than unselective control compounds and could, therefore, be ideal for autoimmune therapy.
Collapse
Affiliation(s)
- Julian Laux
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
| | - Mariella Martorelli
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
| | - Nadja Späth
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Florian Maier
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Michael Burnet
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Stefan A. Laufer
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany
- Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| |
Collapse
|
42
|
Seal R, Schwab LSU, Chiarolla CM, Hundhausen N, Klose GH, Reu-Hofer S, Rosenwald A, Wiest J, Berberich-Siebelt F. Delayed and limited administration of the JAKinib tofacitinib mitigates chronic DSS-induced colitis. Front Immunol 2023; 14:1179311. [PMID: 37275854 PMCID: PMC10235777 DOI: 10.3389/fimmu.2023.1179311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/25/2023] [Indexed: 06/07/2023] Open
Abstract
In inflammatory bowel disease, dysregulated T cells express pro-inflammatory cytokines. Using a chronic azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced colitis model resembling ulcerative colitis, we evaluated whether and when treatment with the Janus kinase (JAK) inhibitor tofacitinib could be curative. Comparing the treatment with two and three cycles of tofacitinib medication in drinking water - intermittently with DSS induction - revealed that two cycles were not only sufficient but also superior over the 3-x regimen. The two cycles of the 2-x protocol paralleled the second and third cycles of the longer protocol. T cells were less able to express interferon gamma (IFN-γ) and the serum levels of IFN-γ, interleukin (IL)-2, IL-6, IL-17, and tumor necrosis factor (TNF) were significantly reduced in sera, while those of IL-10 and IL-22 increased under the 2-x protocol. Likewise, the frequency and effector phenotype of regulatory T cells (Tregs) increased. This was accompanied by normal weight gain, controlled clinical scores, and restored stool consistency. The general and histologic appearance of the colons revealed healing and tissue intactness. Importantly, two phases of tofacitinib medication completely prevented AOM-incited pseudopolyps and the hyper-proliferation of epithelia, which was in contrast to the 3-x regimen. This implies that the initial IBD-induced cytokine expression is not necessarily harmful as long as inflammatory signaling can later be suppressed and that time-restricted treatment allows for anti-inflammatory and tissue-healing cytokine activities.
Collapse
Affiliation(s)
- Rishav Seal
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Lara S. U. Schwab
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | | - Nadine Hundhausen
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Georg Heinrich Klose
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Simone Reu-Hofer
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Johannes Wiest
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | |
Collapse
|
43
|
Ojha AA, Srivastava A, Votapka LW, Amaro RE. Selectivity and Ranking of Tight-Binding JAK-STAT Inhibitors Using Markovian Milestoning with Voronoi Tessellations. J Chem Inf Model 2023; 63:2469-2482. [PMID: 37023323 PMCID: PMC10131228 DOI: 10.1021/acs.jcim.2c01589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Janus kinases (JAK), a group of proteins in the nonreceptor tyrosine kinase (NRTKs) family, play a crucial role in growth, survival, and angiogenesis. They are activated by cytokines through the Janus kinase-signal transducer and activator of a transcription (JAK-STAT) signaling pathway. JAK-STAT signaling pathways have significant roles in the regulation of cell division, apoptosis, and immunity. Identification of the V617F mutation in the Janus homology 2 (JH2) domain of JAK2 leading to myeloproliferative disorders has stimulated great interest in the drug discovery community to develop JAK2-specific inhibitors. However, such inhibitors should be selective toward JAK2 over other JAKs and display an extended residence time. Recently, novel JAK2/STAT5 axis inhibitors (N-(1H-pyrazol-3-yl)pyrimidin-2-amino derivatives) have displayed extended residence times (hours or longer) on target and adequate selectivity excluding JAK3. To facilitate a deeper understanding of the kinase-inhibitor interactions and advance the development of such inhibitors, we utilize a multiscale Markovian milestoning with Voronoi tessellations (MMVT) approach within the Simulation-Enabled Estimation of Kinetic Rates v.2 (SEEKR2) program to rank order these inhibitors based on their kinetic properties and further explain the selectivity of JAK2 inhibitors over JAK3. Our approach investigates the kinetic and thermodynamic properties of JAK-inhibitor complexes in a user-friendly, fast, efficient, and accurate manner compared to other brute force and hybrid-enhanced sampling approaches.
Collapse
Affiliation(s)
- Anupam Anand Ojha
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Ambuj Srivastava
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Lane William Votapka
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Rommie E Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
44
|
Zou J, Lin CH, Wang Y, Shen Y, Guan JL. Correspondence on 'A pilot study of tofacitinib for refractory Behçet's syndrome'. Ann Rheum Dis 2023; 82:e100. [PMID: 33495153 DOI: 10.1136/annrheumdis-2020-219810] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 01/15/2023]
Affiliation(s)
- Jun Zou
- Division of Rheumatology and Immunology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Chen-Hong Lin
- Division of Rheumatology and Immunology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yan Wang
- Department of Ophthalmology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yan Shen
- Division of Rheumatology and Immunology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jian-Long Guan
- Division of Rheumatology and Immunology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
45
|
Kim MH, Lee CW. Phosphatase Ssu72 Is Essential for Homeostatic Balance Between CD4 + T Cell Lineages. Immune Netw 2023; 23:e12. [PMID: 37179750 PMCID: PMC10166661 DOI: 10.4110/in.2023.23.e12] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/01/2022] [Accepted: 12/21/2022] [Indexed: 05/15/2023] Open
Abstract
Ssu72, a dual-specificity protein phosphatase, not only participates in transcription biogenesis, but also affects pathophysiological functions in a tissue-specific manner. Recently, it has been shown that Ssu72 is required for T cell differentiation and function by controlling multiple immune receptor-mediated signals, including TCR and several cytokine receptor signaling pathways. Ssu72 deficiency in T cells is associated with impaired fine-tuning of receptor-mediated signaling and a defect in CD4+ T cell homeostasis, resulting in immune-mediated diseases. However, the mechanism by which Ssu72 in T cells integrates the pathophysiology of multiple immune-mediated diseases is still poorly elucidated. In this review, we will focus on the immunoregulatory mechanism of Ssu72 phosphatase in CD4+ T cell differentiation, activation, and phenotypic function. We will also discuss the current understanding of the correlation between Ssu72 in T cells and pathological functions which suggests that Ssu72 might be a therapeutic target in autoimmune disorders and other diseases.
Collapse
Affiliation(s)
- Min-Hee Kim
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
- SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
46
|
Subramanyam SH, Hriczko JT, Pappas A, Schippers A, Wagner N, Ohl K, Tenbrock K. Tofacitinib fails to prevent T cell transfer colitis in mice but ameliorates disease activity. Sci Rep 2023; 13:3762. [PMID: 36882462 PMCID: PMC9992375 DOI: 10.1038/s41598-023-30616-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Tofactinib is a JAK inhibitor approved for ulcerative colitis in humans. Despite of its' proven effectiveness in humans, mechanistic data are scarce on the effectiveness of Tofactinib in experimental colitis in mice. We induced experimental colitis by transfer of CD4+CD25- isolated T cells into RAG2-/- (T and B cell deficient) mice and treated these mice with tofacitinib for 5-6 weeks either with a dosage of 10 or 40 mg/kg body weight immediately after CD4+ transfer or started treatment after first symptoms of disease for several weeks. While treatment with tofacitinib immediately after transfer resulted in an enhanced expansion of CD4+ T cells and did not prevent occurrence of colitis, treatment after start of symptoms of colitis ameliorated disease activity on a clinical basis and in histological analyses. Tofacitinib is effective in the treatment of murine experimental T cell transfer colitis, however does not prevent occurrence of disease.
Collapse
Affiliation(s)
| | - Judit Turyne Hriczko
- Department of Pediatrics, RWTH Aachen University, Pauwelsstr 30, 52074, Aachen, Germany
| | - Angeliki Pappas
- Department of Pediatrics, RWTH Aachen University, Pauwelsstr 30, 52074, Aachen, Germany
| | - Angela Schippers
- Department of Pediatrics, RWTH Aachen University, Pauwelsstr 30, 52074, Aachen, Germany
| | - Nobert Wagner
- Department of Pediatrics, RWTH Aachen University, Pauwelsstr 30, 52074, Aachen, Germany
| | - Kim Ohl
- Department of Pediatrics, RWTH Aachen University, Pauwelsstr 30, 52074, Aachen, Germany
| | - Klaus Tenbrock
- Department of Pediatrics, RWTH Aachen University, Pauwelsstr 30, 52074, Aachen, Germany.
| |
Collapse
|
47
|
Herrera-deGuise C, Serra-Ruiz X, Lastiri E, Borruel N. JAK inhibitors: A new dawn for oral therapies in inflammatory bowel diseases. Front Med (Lausanne) 2023; 10:1089099. [PMID: 36936239 PMCID: PMC10017532 DOI: 10.3389/fmed.2023.1089099] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition of the gastrointestinal tract that requires chronic treatment and strict surveillance. Development of new monoclonal antibodies targeting one or a few single cytokines, including anti-tumor necrosis factor agents, anti-IL 12/23 inhibitors, and anti-α4β7 integrin inhibitors, have dominated the pharmacological armamentarium in IBD in the last 20 years. Still, many patients experience incomplete or loss of response or develop serious adverse events and drug discontinuation. Janus kinase (JAK) is key to modulating the signal transduction pathway of several proinflammatory cytokines directly involved in gastrointestinal inflammation and, thus, probably IBD pathogenesis. Targeting the JAK-STAT pathway offers excellent potential for the treatment of IBD. The European Medical Agency has approved three JAK inhibitors for treating adults with moderate to severe Ulcerative Colitis when other treatments, including biological agents, have failed or no longer work or if the patient cannot take them. Although there are currently no approved JAK inhibitors for Crohn's disease, upadacitinib and filgotinib have shown increased remission rates in these patients. Other JAK inhibitors, including gut-selective molecules, are currently being studied IBD. This review will discuss the JAK-STAT pathway, its implication in the pathogenesis of IBD, and the most recent evidence from clinical trials regarding the use of JAK inhibitors and their safety in IBD patients.
Collapse
Affiliation(s)
| | | | | | - Natalia Borruel
- Unitat d’Atenció Crohn-Colitis, Digestive System Research Unit, Hospital Universitari Vall d’Hebrón, Barcelona, Spain
| |
Collapse
|
48
|
Springer JM, Kermani TA. Recent advances in the treatment of giant cell arteritis. Best Pract Res Clin Rheumatol 2023; 37:101830. [PMID: 37328409 DOI: 10.1016/j.berh.2023.101830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 06/18/2023]
Abstract
Giant cell arteritis (GCA) is a systemic, granulomatous, large-vessel vasculitis that affects individuals over the age of 50 years. Morbidity from disease includes cranial manifestations which can cause irreversible blindness, while extra-cranial manifestations can cause vascular damage with large-artery stenosis, occlusions, aortitis, aneurysms, and dissections. Glucocorticoids while efficacious are associated with significant adverse effects. Furthermore, despite treatment with glucocorticoids, relapses are common. An understanding of the pathogenesis of GCA has led to the discovery of tocilizumab as an efficacious steroid-sparing therapy while additional therapeutic targets affecting different inflammatory pathways are under investigation. Surgical treatment may be indicated in cases of refractory ischemia or aortic complications but data on surgical outcomes are limited. Despite the recent advances, many unmet needs exist, including the identification of patients or subsets of GCA who would benefit from earlier initiation of adjunctive therapies, patients who may warrant long-term immunosuppression and medications that sustain permanent remission. The impact of medications like tocilizumab on long-term outcomes, including the development of aortic aneurysms and vascular damage also warrants investigation.
Collapse
Affiliation(s)
- Jason M Springer
- Vanderbilt University Medical Center, 1161 21st Avenue Sound, T3113 Medical Center North, Nashville, TN, 37232, USA.
| | - Tanaz A Kermani
- University of California Los Angeles, 2020 Santa Monica Boulevard, Suite 540, Santa Monica, CA, 90404, USA.
| |
Collapse
|
49
|
Szegvari G, Dora D, Lohinai Z. Effective Reversal of Macrophage Polarization by Inhibitory Combinations Predicted by a Boolean Protein–Protein Interaction Model. BIOLOGY 2023; 12:biology12030376. [PMID: 36979068 PMCID: PMC10045914 DOI: 10.3390/biology12030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Background: The function and polarization of macrophages has a significant impact on the outcome of many diseases. Targeting tumor-associated macrophages (TAMs) is among the greatest challenges to solve because of the low in vitro reproducibility of the heterogeneous tumor microenvironment (TME). To create a more comprehensive model and to understand the inner workings of the macrophage and its dependence on extracellular signals driving polarization, we propose an in silico approach. Methods: A Boolean control network was built based on systematic manual curation of the scientific literature to model the early response events of macrophages by connecting extracellular signals (input) with gene transcription (output). The network consists of 106 nodes, classified as 9 input, 75 inner and 22 output nodes, that are connected by 217 edges. The direction and polarity of edges were manually verified and only included in the model if the literature plainly supported these parameters. Single or combinatory inhibitions were simulated mimicking therapeutic interventions, and output patterns were analyzed to interpret changes in polarization and cell function. Results: We show that inhibiting a single target is inadequate to modify an established polarization, and that in combination therapy, inhibiting numerous targets with individually small effects is frequently required. Our findings show the importance of JAK1, JAK3 and STAT6, and to a lesser extent STK4, Sp1 and Tyk2, in establishing an M1-like pro-inflammatory polarization, and NFAT5 in creating an anti-inflammatory M2-like phenotype. Conclusions: Here, we demonstrate a protein–protein interaction (PPI) network modeling the intracellular signalization driving macrophage polarization, offering the possibility of therapeutic repolarization and demonstrating evidence for multi-target methods.
Collapse
Affiliation(s)
- Gabor Szegvari
- Translational Medicine Institute, Semmelweis University, 1094 Budapest, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (D.D.); (Z.L.); Tel.: +36-1-2156920 (D.D.)
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, 1094 Budapest, Hungary
- Pulmonary Hospital Torokbalint, 2045 Torokbalint, Hungary
- Correspondence: (D.D.); (Z.L.); Tel.: +36-1-2156920 (D.D.)
| |
Collapse
|
50
|
Ferrarini A, Vacca A, Solimando AG, Tavio M, Acquaviva R, Rocchi M, Nitti C, Salvi A, Menditto V, Luchetti Gentiloni MM, Russo A, Moretti M, Pavani M, Giacometti A, Bonifazi M, Zuccatosta L, Romani L, Racanelli V, Moroncini G, Gabrielli A, Pomponio G. Early administration of tofacitinib in COVID-19 pneumonitis: An open randomised controlled trial. Eur J Clin Invest 2023; 53:e13898. [PMID: 36380693 DOI: 10.1111/eci.13898] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Controversies on sub-populations most sensitive to therapy and the best timing of starting the treatment still surround the use of immunomodulatory drugs in COVID-19. OBJECTIVES We designed a multicentre open-label randomised controlled trial to test the effect of prompt adding of tofacitinib to standard therapy for hospitalised patients affected by mild/moderate COVID-19 pneumonitis. METHODS Patients admitted to three Italian hospitals affected by COVID-19 pneumonitis not requiring mechanical ventilation were randomised to receive standard treatment alone or tofacitinib (10 mg/bid) for 2 weeks, starting within the first 24 h from admission. RESULTS A total of 116 patients were randomised; 49 in the experimental arm completed the 14-day treatment period, 9 discontinued tofacitinib as the disease worsened and were included in the analysis, and 1 died of respiratory failure. All 58 control patients completed the study. Clinical and demographic characteristics were similar between the study groups. In the tofacitinib group, 9/58 (15.5%) patients progressed to noninvasive ventilation (CPAP) to maintain SO2 > 93%, invasive mechanical ventilation or death by day 14 was 15.5%, significantly less than in the control group (20/58, 34.4%, RR 0,45, RRR -55%, NNT 5; p = .018). No differences in severe adverse effect incidence had been observed across the groups. CONCLUSION High-dose tofacitinib therapy in patients with COVID pneumonitis is safe and may prevent deterioration to respiratory failure.
Collapse
Affiliation(s)
- Alessia Ferrarini
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Gastroenterologia ed Endoscopia Digestiva, Ospedali Riuniti Marche Nord, Fano, Italy
| | - Angelo Vacca
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Antonio Giovanni Solimando
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy.,IRCCS Istituto Tumori "Giovanni Paolo II" Bari, Bari, Italy
| | - Marcello Tavio
- Malattie Infettive, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Rossella Acquaviva
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Marco Rocchi
- Statistica Medica, Dipartimento di Scienze Biomolecolari, Università di Urbino, Urbino, Italy
| | - Cinzia Nitti
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Aldo Salvi
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vincenzo Menditto
- Medicina Interna e Sub Intensiva, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | - Alessandro Russo
- Clinica di Malattie Infettive e Tropicali Dipartimento di Scienze Mediche e Chirurgiche Università "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Marco Moretti
- SOD Medicina di Laboratorio Ospedali Riuniti di Ancona, Ancona, Italy
| | - Marianna Pavani
- SOD Medicina di Laboratorio Ospedali Riuniti di Ancona, Ancona, Italy
| | - Andrea Giacometti
- Clinica di Malattie Infettive, Ospedali Riuniti di Ancona, Ancona, Italy
| | | | | | - Laura Romani
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Vito Racanelli
- Dipartimento di Scienze Biomediche e Oncologia Umana U.O.C, Medicina Interna Universitaria "G. Baccelli" A.O.U.C, Policlinico di Bari, Bari, Italy
| | - Gianluca Moroncini
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Armando Gabrielli
- Clinica Medica, Ospedali Riuniti di Ancona, Ancona, Italy.,Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | | |
Collapse
|