1
|
Mikaeili B, Alqahtani ZA, Hincapie AL, Guo JJ. Safety of Janus kinase inhibitors in rheumatoid arthritis: a disproportionality analysis using FAERS database from 2012 to 2022. Clin Rheumatol 2025; 44:1467-1474. [PMID: 39945946 PMCID: PMC11993438 DOI: 10.1007/s10067-025-07360-9] [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: 10/15/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 04/13/2025]
Abstract
INTRODUCTION/OBJECTIVE Janus kinase (JAK) inhibitors have expanded treatment options for rheumatoid arthritis (RA), particularly for patients unresponsive to traditional disease-modifying antirheumatic drugs (DMARDs). However, safety concerns necessitate a thorough post-market evaluation. This study is aimed at comparing the safety profiles of tofacitinib, baricitinib, and upadacitinib using adverse event (AE) reports from the FDA Adverse Event Reporting System (FAERS). METHODS A retrospective disproportionality analysis was performed using the FAERS data from 2012 to 2022. The AE reports were categorized into cardiovascular, cancer, respiratory, gastrointestinal, musculoskeletal, and arthralgia-related events. Proportional reporting ratio (PRR) and reporting odds ratios (RORs) with 95% confidence intervals (CIs) were calculated to identify significant safety signals. RESULTS Of 273,657 AE reports, tofacitinib had the most (227,144), with increased musculoskeletal-related events (ROR = 1.53, 95% CI 1.49-1.57) and a reduced cancer risk (ROR = 0.44, 95% CI 0.41-0.47). Baricitinib (9305 reports) showed the highest risk of cardiovascular events (ROR = 1.63, 95% CI 1.50-1.78) and cancer (ROR = 2.17, 95% CI 1.83-2.58). Upadacitinib (37,208 reports) had elevated risks for respiratory events (ROR = 2.04, 95% CI 1.88-2.21) and cancer (ROR = 2.24, 95% CI 2.05-2.43). CONCLUSION The distinct safety profiles of these JAK inhibitors suggest that baricitinib poses higher cardiovascular and cancer risks, whereas upadacitinib increases the risk of respiratory and gastrointestinal events. Tofacitinib may be safer for patients with a history of cancer but requires monitoring for musculoskeletal AEs. Personalized risk assessments are critical for safe use of JAK inhibitors. Key Points • This study provides a comprehensive post-market safety assessment of three JAK inhibitors-tofacitinib, baricitinib, and upadacitinib-using the FAERS data from 2012 to 2022. • Distinct safety profiles were identified, with baricitinib showing a higher risk of cardiovascular events and cancer, while upadacitinib posed an increased risk of respiratory and gastrointestinal events. • Tofacitinib demonstrated a lower cancer risk than other JAK inhibitors but was associated with more musculoskeletal-related adverse events. • These findings emphasize the importance of personalized risk assessment and vigilant monitoring when prescribing JAK inhibitors for rheumatoid arthritis.
Collapse
Affiliation(s)
- Bahar Mikaeili
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA.
| | - Zuhair A Alqahtani
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
- College of Pharmacy, Health Economics and Outcome Research, King Saud University, Riyadh, Saudi Arabia
| | - Ana L Hincapie
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Jeff Jianfei Guo
- Division of Pharmacy Practice & Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
2
|
Attwa MW, Abdelhameed AS, Kadi AA. An ultra-fast UPLC-MS/MS approach for the quantification of baricitinib in the HLM matrix: greenness assessment with application to in vitro and in silico metabolic stability studies. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:2718-2732. [PMID: 40094167 DOI: 10.1039/d5ay00294j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Baricitinib (Olumiant) is a Janus kinase inhibitor utilized for the management of COVID-19, rheumatoid arthritis, and alopecia areata. It received U.S. Food and Drug Administration approval on May 31, 2018. This work developed a sensitive, rapid, environmentally friendly, and reliable UPLC-MS/MS method for quantifying baricitinib in human liver microsomes, utilized to evaluate the in vitro metabolic stability of baricitinib in HLMs. The StarDrop software, with DEREK and P450 metabolic programs, was employed to detect the structural warnings related to BCB and assess the in silico metabolic lability. The validation of the UPLC-MS/MS approach conformed to U.S. Food and Drug Administration standards for bioanalytical approach validation. The present UPLC-MS/MS method exhibited a wide range of linearity (1.0-3000 ng mL-1) and optimum separation of analytes in an ultra-fast separation time (1 min) and was reproducible and accurate in the absence of human liver microsome matrix effects. Baricitinib and encorafenib (the internal standard) were examined employing an isocratic mobile phase technique on a reversed phase (SB C18) column. This study assessed the accuracy and precision of UPLC-MS/MS methodologies for intra- and inter-day evaluations, which ranged from -1.20% to 8.67% and 0.12% to 11.67%, respectively. The intrinsic clearance of baricitinib was quantified at 27.49 mL min-1 kg-1, while the in vitro half-life was established at 29.50 minutes. In silico analysis proposes that slight structural alterations to the pyrrole ring (88%) and the pyrimidine ring (5%) in drug design may increase safety and metabolic stability related to baricitinib. The evaluation of in silico absorption, distribution, metabolism, excretion, and metabolic stability characteristics for baricitinib is crucial for advancing innovative drug discovery focused on enhancing metabolic stability.
Collapse
Affiliation(s)
- Mohamed W Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Ali S Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Adnan A Kadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
3
|
Pei W, Xu L, Zhong H, Wang Z, Yao R, Zhang L, Yang J, Li J, Feng Y, Lin Q, Li D, Zhou X, Pei D, Guo Y, Ma L, Luo Y, Zuo S, Wang L, Yan R, Su Y. Clinical features of inflammatory arthritis in daily practice-China's perspective. Clin Rheumatol 2025; 44:969-978. [PMID: 39853560 DOI: 10.1007/s10067-024-07262-2] [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: 05/31/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 01/26/2025]
Abstract
OBJECTIVE This study aimed to analyze and compare the proportion of patients with different types of inflammatory arthritis and investigate the clinical characteristics, including symptoms and signs, medication choices, and disease activity, in the daily clinical practice of China. METHODS Patients with inflammatory arthritis were recruited from 16 Grade-A tertiary hospitals between August 2021 and April 2022. The medical profiles, encompassing sociodemographic characteristics, clinical and laboratory date, were collected. RESULTS This study included 2,693 patients with arthritis, with rheumatoid arthritis (RA) accounting for the highest proportion (50.50%). Significant differences were observed in terms of age, gender, body mass index (BMI), disease duration, smoking and family history among patients with different types of inflammatory arthritis. Physical activity and cold exposure were identified as the main predisposing factors for RA, psoriatic arthritis (PsA), osteoarthritis (OA), and ankylosing spondylitis (AS), while alcohol consumption was the most common inducing factor for gout. Hypertension and hyperlipidemia were the primary concomitant diseases in RA, OA, and AS, whereas hyperuricemia and hypertension were mainly associated with gout, psoriasis and diabetes were the most common comorbidities in PsA. Peripheral joints were predominantly affected in PsA, RA, OA, and gout, while axial joints were mainly affected in AS. Methotrexate and leflunomide were the main therapeutic drugs for RA, while biologics were commonly prescribed for PsA and AS. OA and gout patients mainly utilized nonsteroidal anti-inflammatory drugs (NSAIDs). CONCLUSION Patients with different types of inflammatory arthritis exhibited varying predisposing factors, joint inflammation, concomitant diseases, and medication choices, highlighting the importance of individualized approaches in the clinic. Key Points • 2,693 patients classified and diagnosed with inflammatory arthritis were recruited in this study from 16 Grade-A tertiary hospitals in China between August 2021 and April 2022. • This study analyzed and compared the proportion of patients with different types of arthritis in routine clinical practice in China. • Joint inflammation, comorbidities, and medication choices were assessed among patients with the most common types of arthritis in this study. • This study also provided some epidemiologically relevant information about inflammatory arthritis patients in China.
Collapse
MESH Headings
- Humans
- Male
- Female
- Middle Aged
- China/epidemiology
- Adult
- Arthritis, Rheumatoid/epidemiology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/complications
- Arthritis, Psoriatic/epidemiology
- Arthritis, Psoriatic/drug therapy
- Arthritis, Psoriatic/diagnosis
- Arthritis, Psoriatic/complications
- Antirheumatic Agents/therapeutic use
- Aged
- Osteoarthritis/epidemiology
- Spondylitis, Ankylosing/drug therapy
- Spondylitis, Ankylosing/epidemiology
- Spondylitis, Ankylosing/complications
- Comorbidity
- Gout/epidemiology
- Gout/drug therapy
- Risk Factors
Collapse
Affiliation(s)
- Wenwen Pei
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Liling Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Hua Zhong
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ziye Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ranran Yao
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Lei Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Rheumatology and Immunology, Mianyang Central Hospital, Mianyang, China
| | - Jingyang Li
- Department of Rheumatology and Immunology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, China
| | - Yuan Feng
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, China
| | - Qi Lin
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dongsheng Li
- Department of Rheumatology, Ganzhou People's Hospital, Ganzhou, China
| | - Xinyao Zhou
- Division of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongxue Pei
- Department of Rheumatology, Jilin Hospital of Integrated Traditional Chinese and Western Medicine, Jilin, China
| | - Yanqiu Guo
- Department of Rheumatology and Immunology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Li Ma
- Department of Rheumatology, China-Japan Friendship Hospital, Beijing, China
| | - Yaping Luo
- Department of Rheumatology, Hebei Provincial Hospital of Traditional Chinese Medicine, Hebei, China
| | - Shufei Zuo
- Department of Rheumatology and Immunology, Xinxiang Central Hospital, Xinxiang, China
| | - Lin Wang
- Department of Rheumatology, Shaoyang Central Hospital, Shaoyang, China
| | - Rui Yan
- Department of Rheumatology and Immunology, Beijing Shunyi Hospital, Beijing, China
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| |
Collapse
|
4
|
Leo F, Ortoncelli M, Cascio Ingurgio R, Galli B, Grigolato L, Paganini C, Maurelli M, Di Brizzi EV, Lauletta G, Barei F, Fiasconaro CA, Casale Alloa M, Guanti MB, Gori N, Chiricozzi A, Napolitano M, Patruno C, Galluzzo M, Rossi M, Balato A, Ferrucci SM, Marzano AV, Pezzolo E, Foti C, Girolomoni G, Gargiulo L, Narcisi A, Quaglino P, Ribero S, Mastorino L. Safety and Effectiveness of Upadacitinib in Patients with Moderate-to-Severe Atopic Dermatitis Who Smoke: a 2-Year Real-Life Multicenter Study. Am J Clin Dermatol 2025:10.1007/s40257-025-00926-y. [PMID: 39994105 DOI: 10.1007/s40257-025-00926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin condition that significantly impairs the quality of life. Recent advancements in systemic therapies, such as Janus kinase (JAK) inhibitors, offer very effective new treatment options. However, concerns regarding potential adverse events, including cardiovascular and thromboembolic risk, have emerged from clinical studies and call for further real-life investigations. This has highlighted the need to establish specific risk categories, such as tobacco smokers. OBJECTIVE This study aims to evaluate the safety and effectiveness of upadacitinib, a JAK1 inhibitor, in patients who smoke with moderate-to-severe AD over a 2-year treatment period, comparing outcomes with patients who do not smoke. PATIENTS AND METHODS A retrospective multicenter study was conducted across 12 dermatology departments in Italy, including 375 patients treated with upadacitinib. The presence and intensity of smoking habits as well as effectiveness scores and safety data were collected. RESULTS Patients who smoke accounted for 36.8% of the sample. Two thromboembolic events in patients who do not smoke were recorded in the 2-year (median follow up of 52.6 weeks) observation period. The most common adverse event was acneiform eruption (12.4% of patients after 104 weeks). No significant differences related to safety emerged regarding the presence or absence of a smoking habit. Drug survival was very high with no differences between the two cohorts (83.5% after 104 weeks for patients who smoke). CONCLUSIONS This study suggests that upadacitinib is a safe and effective treatment for moderate-to-severe AD in presence of tobacco smoke, with no significant differences in safety or effectiveness compared with patients who do not smoke.
Collapse
Affiliation(s)
- Francesco Leo
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy.
| | - Michela Ortoncelli
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy
| | | | - Benedetta Galli
- Dermatology Department, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Laura Grigolato
- Dermatology Department, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Claudia Paganini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Martina Maurelli
- Department of Medicine, Section of Dermatology, University of Verona, Verona, Italy
| | | | - Giuseppe Lauletta
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesca Barei
- Dermatology Unit, I.R.C.C.S. Foundation Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Anna Fiasconaro
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy
| | - Marta Casale Alloa
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy
| | - Mario Bruno Guanti
- Allergology Service, Dermatology Unit, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Niccolò Gori
- Dipartimento di Scienze Mediche e Chirurgiche, UOC di Dermatologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Chiricozzi
- Dipartimento di Scienze Mediche e Chirurgiche, UOC di Dermatologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maddalena Napolitano
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Cataldo Patruno
- Magna Graecia University of Catanzaro, Catanzaro, Calabria, Italy
| | - Marco Galluzzo
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Mariateresa Rossi
- Dermatology Department, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Anna Balato
- Dermatology Unit, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Silvia Mariel Ferrucci
- Dermatology Unit, I.R.C.C.S. Foundation Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angelo Valerio Marzano
- Dermatology Unit, I.R.C.C.S. Foundation Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Elena Pezzolo
- Department of Dermatology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Caterina Foti
- Unit of Dermatology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Bari, Italy
| | - Giampiero Girolomoni
- Department of Medicine, Section of Dermatology, University of Verona, Verona, Italy
| | - Luigi Gargiulo
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | - Pietro Quaglino
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy
| | - Simone Ribero
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy
| | - Luca Mastorino
- Dermatology Section, Department of Medical Sciences, Dermatology Clinic, University of Turin, Via Cherasco 23, 10121, Torino, Italy
| |
Collapse
|
5
|
Yi RC, Akbik M, Smith LR, Klionsky Y, Feldman SR. Therapeutic Advancements in Psoriasis and Psoriatic Arthritis. J Clin Med 2025; 14:1312. [PMID: 40004842 PMCID: PMC11855982 DOI: 10.3390/jcm14041312] [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: 01/26/2025] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Within the past few years, many new therapies have emerged for psoriasis and psoriatic arthritis (PsA). Current topical therapies-including corticosteroids, vitamin D analogs, tapinarof, and roflumilast-remain the mainstay for mild disease, while oral systemic and biologic options are for moderate to severe cases. Biologics-such as Tumor necrosis factor-alpha (TNF-alpha), Interleukin 12/23 (IL-12/23), Interleukin-17 (IL-17), and Interleukin-23 (IL-23)-have revolutionized care by providing highly effective and safer alternatives. Oral small molecules, including Janus kinase (JAK) and tyrosine kinase 2 (TYK2) inhibitors, further expand the therapeutic options. Objectives: The goal for this review article was to examine current and latest treatments for psoriasis and PsA and discuss whether these emerging therapeutic options address the unmet needs of current treatments. Methods: The search for this review article included PubMed, Google Scholar, and ClinicalTrials.gov for relevant articles and current clinical trials using keywords. Results: A wide range of novel psoriatic and PsA therapies are currently undergoing clinical trials. These include selective JAK inhibitors, TYK2 inhibitors, retinoic acid-related orphan receptor (RORγT) inhibitors, oral IL-23 receptor inhibitors, oral IL-17A inhibitors, nanobody products, sphingosine-1-phosphate (S1P1R) antagonists, A3 adenosine receptor (A3AR) agonists, heat shock protein (HSP) 90 inhibitors, and rho-associated protein kinases (ROCK-2) inhibitors. Conclusions: These different mechanisms of action not only expand treatment options but may offer potential solutions for patients who do not achieve adequate response with existing therapies. However, the safety and contraindications of these newer agents remain an important consideration to ensure appropriate patient selection and minimize potential risks. Certain mechanisms may pose increased risks for infection, cardiovascular manifestations, malignancy, or other immune-related adverse events, necessitating careful monitoring and individualized treatment decisions. Ongoing clinical research aims to address unmet needs for patients who do not respond to previous agents to achieve sustained remission, monitor long-term safety outcomes, and assess patient preferences for delivery, including a preference for oral delivery. Oral IL-23 inhibitors hold potential due to their robust safety profiles. In contrast, oral IL-17 inhibitors and TYK-2 inhibitors are effective but may present side effects that could impact their acceptability. It is essential to balance efficacy, safety, and patient preferences to guide the selection of appropriate therapies.
Collapse
Affiliation(s)
- Robin C. Yi
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (L.R.S.); (S.R.F.)
| | - Maya Akbik
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Logan R. Smith
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (L.R.S.); (S.R.F.)
| | - Yael Klionsky
- Department of Internal Medicine, Division of Rheumatology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA;
| | - Steven R. Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (L.R.S.); (S.R.F.)
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
6
|
Liu J, Jiang Y, Zhang S, Liu S, Su J, Lin C, He X, Wu R, Yang L, Liu H, Duan X, Xu S, Luo H, Liu J, Xie Q, Mi C, Chen L, Zhang N, Gong H, Zhu J, Li Y, Wei H, Qian L, Wang J, Shi X, Jin H, Jiang Z, Xie X, Zhan F, Geng X, Zheng Z, Du Z, Dong G, Sun Y, Zeng X. Ivarmacitinib, a selective Janus kinase 1 inhibitor, in patients with moderate-to-severe active rheumatoid arthritis and inadequate response to conventional synthetic DMARDs: results from a phase III randomised clinical trial. Ann Rheum Dis 2025; 84:188-200. [PMID: 39919893 DOI: 10.1136/ard-2024-226385] [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: 07/08/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE To assess the efficacy/safety of ivarmacitinib, a selective Janus kinase (JAK) 1 inhibitor, in patients with moderate-to-severe active rheumatoid arthritis (RA) who had an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). METHODS Patients were randomised (1:1:1) to receive either placebo (n=188), ivarmacitinib 4 mg (n=189) or ivarmacitinib 8 mg (n=189) once daily, with background csDMARDs allowed. After 24 weeks, patients on placebo switched to ivarmacitinib 4 mg for an additional 28 weeks, while those on ivarmacitinib continued their initial dosage. The primary endpoint was the proportion of patients achieving a 20% improvement in the American College of Rheumatology response criteria (ACR20) at week 24. RESULTS At week 24, ACR20 response rates were significantly higher in the ivarmacitinib 4 mg (70.4%) and 8 mg (75.1%) groups compared with the placebo group (40.4%; both p<0.0001). Both ivarmacitinib doses achieved numerically higher Disease Activity Score 28-joint count C reactive protein of <2.6/≤3.2 response rates compared with placebo. Improvements in efficacy and patient-reported outcomes were sustained through 52 weeks and were noted in patients who switched from placebo after week 24. During the placebo-controlled period, treatment-emergent adverse events (TEAEs) occurred in 81.5% and 90.5% of patients in the ivarmacitinib 4 mg and 8 mg groups, versus 79.3% in the placebo group. Infection-related TEAEs were slightly higher in the ivarmacitinib groups. CONCLUSIONS Ivarmacitinib may offer a potential therapeutic option for patients with RA who have an inadequate response to csDMARDs, with a safety profile that was generally manageable over 1 year of treatment and similar to other JAK inhibitors. TRIAL REGISTRATION NUMBER NCT04333771.
Collapse
Affiliation(s)
- Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Ying Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Shangzhu Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Shengyun Liu
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingbo Su
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Changsong Lin
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong He
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lei Yang
- Department of Rheumatology and Immunology, Zhengzhou Central Hospital, Zhengzhou, China
| | - Huaxiang Liu
- Rheumatology Department, Qilu Hospital of Shandong University, Jinan, China
| | - Xinwang Duan
- Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengqian Xu
- Rheumatology and Immunology Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Liu
- Department of Rheumatology and Immunology Research Institute, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu, China
| | - Cundong Mi
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lin Chen
- Department of Rheumatic Immunity, Jilin Province People's Hospital, Changchun, China
| | - Ning Zhang
- Rheumatology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Huiping Gong
- Department of Rheumatology and Clinical Immunology, Peace Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Jing Zhu
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Yasong Li
- Department of Rheumatology and Clinical Immunology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Hua Wei
- Rheumatology and Immunology Department, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Long Qian
- Department of Rheumatology and Clinical Immunology, The Second Hospital of Anhui Medical University, Anhui, China
| | - Jian Wang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Anhui, China
| | - Xiaofei Shi
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Henan University of Science & Technology, Luoyang, China
| | - Hongtao Jin
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenyu Jiang
- Department of Rheumatic Immunity, The First Hospital of Jilin University, Changchun, China
| | - Xi Xie
- Department of Clinical Immunology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Zhan
- Department of Rheumatology and Immunology, Hainan General Hospital, Haikou, China
| | - Xiuqin Geng
- Department of Rheumatology and Immunology, Xinxiang Central Hospital, Xinxiang, China
| | - Zhaohui Zheng
- Department of Clinical Immunology, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhengfu Du
- Department of Rheumatology and Immunology, Yuncheng Central Hospital, Yuncheng, China
| | - Guangchao Dong
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Yuqi Sun
- Biometric Department, Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
7
|
Makhija M, Manchanda D, Sharma M. Nano-based Therapeutics for Rheumatoid Arthritis: Recent Patents and Development. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:56-75. [PMID: 37691226 DOI: 10.2174/1872210518666230905155459] [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: 02/22/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease marked by inflammation of synovium and generation of autoantibodies. Bone and cartilage are frequently damaged along with weakening of tendons and ligaments resulting in disability. An effective RA treatment needs a multi-disciplinary approach which relies upon pathophysiology that is still partially understood. In RA patients, inflammation was induced by pro-inflammatory cytokines including IL-1, IL-6 & IL-10. The conventional dosage regimens for treating RA have drawbacks such as ineffectiveness, greater doses, frequent dosing, relatively expensive and serious adverse effects. To formulate an effective treatment plan for RA, research teams have recently focused on producing several nanoformulations containing anti-inflammatory APIs with an aim to target the inflamed area. Nanomedicines have recently gained popularity in the treatment of RA. Interestingly, unbelievable improvements have been observed in current years in diagnosis and management of RA utilizing nanotechnology. Various patents and clinical trial data have been reported in relevance to RA treatment.
Collapse
Affiliation(s)
- Manish Makhija
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Deeksha Manchanda
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Manu Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
| |
Collapse
|
8
|
Loftus AEP, Romano MS, Phuong AN, McKinnel BJ, Muir MT, Furqan M, Dawson JC, Avalle L, Douglas AT, Mort RL, Byron A, Carragher NO, Pollard SM, Brunton VG, Frame MC. An ILK/STAT3 pathway controls glioblastoma stem cell plasticity. Dev Cell 2024; 59:3197-3212.e7. [PMID: 39326421 DOI: 10.1016/j.devcel.2024.09.003] [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: 08/06/2023] [Revised: 04/16/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024]
Abstract
Glioblastoma (GBM) is driven by malignant neural stem-like cells that display extensive heterogeneity and phenotypic plasticity, which drive tumor progression and therapeutic resistance. Here, we show that the extracellular matrix-cell adhesion protein integrin-linked kinase (ILK) stimulates phenotypic plasticity and mesenchymal-like, invasive behavior in a murine GBM stem cell model. ILK is required for the interconversion of GBM stem cells between malignancy-associated glial-like states, and its loss produces cells that are unresponsive to multiple cell state transition cues. We further show that an ILK/STAT3 signaling pathway controls the plasticity that enables transition of GBM stem cells to an astrocyte-like state in vitro and in vivo. Finally, we find that ILK expression correlates with expression of STAT3-regulated proteins and protein signatures describing astrocyte-like and mesenchymal states in patient tumors. This work identifies ILK as a pivotal regulator of multiple malignancy-associated GBM phenotypes, including phenotypic plasticity and mesenchymal state.
Collapse
Affiliation(s)
- Alexander E P Loftus
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK.
| | - Marianna S Romano
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Anh Nguyen Phuong
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Ben J McKinnel
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Morwenna T Muir
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Muhammad Furqan
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - John C Dawson
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Science, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Adam T Douglas
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Richard L Mort
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Adam Byron
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Neil O Carragher
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Valerie G Brunton
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Margaret C Frame
- Cancer Research UK Scotland Centre (Edinburgh), Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK.
| |
Collapse
|
9
|
Poon EK, Liu L, Wu KC, Lim J, Sweet MJ, Lohman RJ, Iyer A, Fairlie DP. A novel inhibitor of class IIa histone deacetylases attenuates collagen-induced arthritis. Br J Pharmacol 2024; 181:4804-4821. [PMID: 39223784 DOI: 10.1111/bph.17306] [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: 07/23/2023] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Most inhibitors of histone deacetylases (HDACs) are not selective and are cytotoxic. Some have anti-inflammatory activity in disease models, but cytotoxicity prevents long-term uses in non-fatal diseases. Inhibitors selective for class IIa HDACs are much less cytotoxic and may have applications in management of chronic inflammatory diseases. EXPERIMENTAL APPROACH LL87 is a novel HDAC inhibitor examined here for HDAC enzyme selectivity. It was also investigated in macrophages for cytotoxicity and for inhibition of lipopolysaccharide (LPS)-stimulated cytokine secretion. In a rat model of collagen-induced arthritis, LL87 was investigated for effects on joint inflammation in Dark Agouti rats. Histological, immunohistochemical, micro-computed tomography and molecular analyses characterise developing arthritis and anti-inflammatory efficacy. KEY RESULTS LL87 was significantly more inhibitory against class IIa than class I or IIb HDAC enzymes. In macrophages, LL87 was not cytotoxic and reduced both LPS-induced secretion of pro-inflammatory cytokines, and IL6-induced class IIa HDAC activity. In rats, LL87 attenuated paw swelling and clinical signs of arthritis, reducing collagen loss and histological damage in ankle joints. LL87 decreased immune cell infiltration, especially pro-inflammatory macrophages and osteoclasts, into synovial joints and significantly reduced expression of pro-inflammatory cytokines and tissue-degrading proteases. CONCLUSION AND IMPLICATIONS A novel inhibitor of class IIa HDACs has been shown to have an anti-inflammatory and anti-arthritic profile distinct from current therapies. It is efficacious in reducing macrophage infiltration and joint inflammation in a chronic model of rat arthritis.
Collapse
Affiliation(s)
- Eunice K Poon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Ligong Liu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kai-Chen Wu
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Junxian Lim
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Rink-Jan Lohman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Abishek Iyer
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
10
|
Shi Z, Cai J, Yang L, Tang L, She L. Safety of baricitinib 24 weeks 4 mg or 2 mg for the treatment of rheumatoid arthritis: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2024; 103:e40512. [PMID: 39560543 PMCID: PMC11576007 DOI: 10.1097/md.0000000000040512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 10/25/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUNDS Baricitinib, an oral selective inhibitor of Janus kinase 1 and 2, is approved for moderate and severe rheumatoid arthritis (RA) with insufficient response to conventional synthetic disease-modifying antirheumatic drugs. The study evaluated the safety of baricitinib 24 weeks 4 mg or 2 mg for the treatment of RA. METHODS The net change (least squares mean [LSM]) of alanine aminotransferase (ALT), creatinine, low-density lipoprotein cholesterol (LDL-C) levels from baseline with the comparison of baricitinib versus placebo was pooled, respectively. The risk ratios (RR) of serious advanced events (SAEs), major cardiovascular events (MACEs), infection, serious infection, and advanced events (AEs) at the end of treatment across groups were compared. RESULTS Five randomized controlled trials with 2901 patients were included in the summary analysis. Results showed that baricitinib 4 mg significantly increased ALT and creatinine levels, the net LSM change was respectively 3.59 U/L with 95% confidence interval (CI) (1.75-5.43), 4.25 µmol/L with 95% CI (3.38-5.12), however, baricitinib 2 mg of ALT and creatinine levels were not significantly different. Baricitinib 4 mg and 2 mg significantly increased LDL-C levels, the net LSM change was respectively 11.44 mg/dL with 95% CI (6.08-16.80), 8.70 mg/dL with 95% CI (4.19-13.20). Baricitinib 4 mg significantly increased the incidence of infection, the pooled RR (95% CI) was 1.29 (1.13-1.47), and baricitinib 2 mg was not significantly different. However, the pooled RRs of SAEs, MACEs, and serious infection were not statistically significant across groups. The pooled RRs of AEs were not statistically significant between baricitinib 4 mg and 2 mg. CONCLUSIONS This study confirmed that patients with RA taking 4 mg baricitinib increased levels of ALT, creatinine, as well as an increased risk of infections, compared with those taking 2 mg baricitinib. Both 2 mg and 4 mg also increased the level of LDL-C, but it increased the most severely at 4 mg baricitinib. However, the incidence of SAEs, MACEs, and serious infection was not significantly different in patients treated with baricitinib 4 mg and 2 mg compared with placebo, the incidence of AEs was not significantly different between baricitinib 4 mg and 2 mg.
Collapse
Affiliation(s)
- Zhihua Shi
- Department of Pharmacology, Hunan University of Medicine General Hospital, Huaihua, People’s Republic of China
| | - Junlong Cai
- Department of Pharmacology, Hunan University of Medicine General Hospital, Huaihua, People’s Republic of China
| | - Ling Yang
- Department of Pharmacology, Hunan University of Medicine General Hospital, Huaihua, People’s Republic of China
| | - Lizhi Tang
- Department of Pharmacology, Hunan University of Medicine General Hospital, Huaihua, People’s Republic of China
| | - Lang She
- Department of Pharmacology, Hunan University of Medicine General Hospital, Huaihua, People’s Republic of China
| |
Collapse
|
11
|
Bergstra SA, van Ouwerkerk L, Nevins IS, van der Pol JA, Helmich GS, Hest I, van Veen A, Bos R, Goekoop-Ruiterman YPM, Vonkeman HE, Bijsterbosch J, de Jong PHP, Güler-Yüksel M, Böhringer S, Huizinga TWJ, van Gaalen FA. Induction of Cure in Early Arthritis (I CEA): study protocol for an investigator-initiated randomized single-blind clinical trial with open-label extension to compare three treatment strategies in patients with newly diagnosed undifferentiated arthritis. Trials 2024; 25:758. [PMID: 39533416 PMCID: PMC11558825 DOI: 10.1186/s13063-024-08609-5] [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: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Undifferentiated arthritis (UA) is a term used to describe patients with inflammatory arthritis that has not differentiated into a specific rheumatic disease. UA may be a pre-stage of rheumatoid arthritis (RA) or another inflammatory disease or remain undifferentiated, but a substantial proportion of patients may also achieve spontaneous remission. As UA may be an early presentation of RA, rheumatologists often start methotrexate (or another csDMARD) as early as possible. There are however very little data on the potential benefits of early DMARD treatment, and longitudinal data suggests that long-term outcomes such as physical functioning hardly improved in these patients in the past decades. In the I CEA trial, we investigate if it is beneficial to start early treatment with MTX or baricitinib, a more rapidly acting drug with a broader mechanism of action, compared to waiting for spontaneous remission with symptomatic therapy in patients with UA. METHODS The I CEA is a multicenter single-blind (independent assessor) randomized clinical trial with a 3-month interventional and 9-month observational follow-up period. The study includes patients with early (< 1 year symptom duration) DMARD-naïve undifferentiated arthritis. Patients with an increased risk of AEs with baricitinib treatment are excluded. Participants are randomized 1:1:1 to (1) symptom relief with NSAIDs and a single injection of glucocorticoids and (waiting for spontaneous remission); (2) methotrexate and a single injection of glucocorticoids, and NSAIDs are optional; and (3) baricitinib and a single injection of glucocorticoids and NSAIDs are optional. During the observational follow-up period, patients are treated in shared decision with their rheumatologist. The primary outcome will be the change in DAS at 3 months. Secondary outcomes include radiographic progression, physical functioning, patient-reported outcomes, cost utilities, safety, progression to classifiable RA, and disease activity over time. DISCUSSION The 12-month I CEA trial studies early treatment of patients with UA with methotrexate, baricitinib, or NSAIDs. The study initially had a more complex design. Emerging safety warnings about baricitinib necessitated adjustment of the trial protocol including more extensive exclusion criteria. The number of included patients was lower than initially planned, supported by an updated sample size calculation. TRIAL REGISTRATION Dutch trial register NL73202.058.20, EudraCT 2019-004359-35, registered 10-06-2020. Protocol version 1Q, 06-05-2024.
Collapse
Affiliation(s)
- S A Bergstra
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands.
| | - L van Ouwerkerk
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - I S Nevins
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - J A van der Pol
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - G S Helmich
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - I Hest
- Patient Partner Panel, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - A van Veen
- Patient Partner Panel, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - R Bos
- Department of Rheumatology, Medical Center Leeuwarden, Leeuwarden, the Netherlands and Department of Rheumatology, Nij Smellinghe Hospital, Drachten, the Netherlands
| | | | - H E Vonkeman
- Department of Rheumatology and Clinical Immunology, Medisch Spectrum Twente, Enschede, the Netherlands and Department of Psychology, Health and Technology, University of Twente, Enschede, the Netherlands
| | - J Bijsterbosch
- Department of Rheumatology, Amphia Hospital, Breda, the Netherlands
| | - P H P de Jong
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - M Güler-Yüksel
- Department of Rheumatology and Immunology, Maasstad Hospital, Rotterdam, the Netherlands
| | - S Böhringer
- Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands and Department of Pharmacology and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - T W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - F A van Gaalen
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
12
|
Vijayan S, Margesan T. Comprehensive investigation of network pharmacology, computational modeling, and pharmacokinetic assessment to evaluate the efficacy of flavonoids in rheumatoid arthritis. Mol Divers 2024:10.1007/s11030-024-10989-4. [PMID: 39348084 DOI: 10.1007/s11030-024-10989-4] [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: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
Rheumatoid arthritis is a chronic autoimmune disease characterized by inflammation and joint damage, imposing a significant burden on affected individuals worldwide. Flavonoids, a class of natural compounds abundant in various plant-based foods, have shown promising anti-inflammatory and immunomodulatory effects, suggesting their potential as therapeutic agents for RA. In this study, we conducted a comprehensive investigation of identified LCMS compounds utilizing network pharmacology, computational modeling, in silico approaches, and pharmacokinetic assessment to evaluate the efficacy of flavonoids in RA treatment. The study identified 5 flavonoid structures with common targets via LCMS and Integration of network pharmacology approaches enabled a comprehensive evaluation of the pharmacological profile of flavonoids in the context of RA treatment, guiding the selection of promising candidates for further experimental validation and clinical development. The top 10 targets were AKT1, PI3KR1, CDK2, EGFR, CDK6, NOS2, FLT3, ALOX5, CCNB1, and PTPRS via PPI network. The investigation emphasized several pathways, including the AGE-RAGE signaling pathway, resistance to EGFR tyrosine kinase inhibitors, the PI3K-AKT signaling network, and the Rap 1 signaling pathway. In silico studies estimated binding affinities that ranged from - 7.0 to - 10.0 kcal/mol. Schaftoside and Vitexin showed no toxicity in computational approach and found suitable for further investigations. Overall, our study underscores the potential of flavonoids as therapeutic agents for RA and highlights the utility of integrative approaches combining network pharmacology, computational modeling, in silico methods, and pharmacokinetic assessment in drug discovery and development processes.
Collapse
Affiliation(s)
- Sukanya Vijayan
- Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Thirumal Margesan
- Department of Pharmacognosy, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India.
| |
Collapse
|
13
|
Faris A, Cacciatore I, Alnajjar R, Aouidate A, AL Mughram MH, Elhallaoui M. Computational insights into rational design and virtual screening of pyrazolopyrimidine derivatives targeting Janus kinase 3 (JAK3). Front Chem 2024; 12:1425220. [PMID: 39189018 PMCID: PMC11345245 DOI: 10.3389/fchem.2024.1425220] [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: 04/29/2024] [Accepted: 06/12/2024] [Indexed: 08/28/2024] Open
Abstract
The Janus kinase 3 (JAK3) family, particularly JAK3, is pivotal in initiating autoimmune diseases such as rheumatoid arthritis. Recent advancements have focused on developing antirheumatic drugs targeting JAK3, leading to the discovery of novel pyrazolopyrimidine-based compounds as potential inhibitors. This research employed covalent docking, ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) analysis, molecular dynamics modeling, and MM/GBSA (Molecular Mechanics Generalized Born Surface Area) binding free energy techniques to screen 41 in silico-designed pyrazolopyrimidine derivatives. Initially, 3D structures of the JAK3 enzyme were generated using SWISS-MODEL, followed by virtual screening and covalent docking via AutoDock4 (AD4). The selection process involved the AMES test, binding affinity assessment, and ADMET analysis, narrowing down the candidates to 27 compounds that passed the toxicity test. Further covalent docking identified compounds 21 and 41 as the most promising due to their high affinity and favourable ADMET profiles. Subsequent development led to the creation of nine potent molecules, with derivatives 43 and 46 showing exceptional affinity upon evaluation through molecular dynamics simulation and MM/GBSA calculations over 300 nanoseconds, comparable to tofacitinib, an approved RA drug. However, compounds L21 and L46 demonstrated stable performance, suggesting their effectiveness in treating rheumatoid arthritis and other autoimmune conditions associated with JAK3 inhibition.
Collapse
Affiliation(s)
- Abdelmoujoud Faris
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Radwan Alnajjar
- CADD Unit, PharmD, Faculty of Pharmacy, Libyan International Medical University, Benghazi, Libya
| | - Adnane Aouidate
- School of Applied Sciences of Ait Melloul, Ibn Zohr University, Agadir, Morocco
| | - Mohammed H. AL Mughram
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Menana Elhallaoui
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
14
|
Zheng J, Ding Y, Chen Y, Shi Y, Gao Y. Effectiveness of baricitinib in acquired reactive perforating collagenosis: a case report. Front Immunol 2024; 15:1388274. [PMID: 39076971 PMCID: PMC11284048 DOI: 10.3389/fimmu.2024.1388274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/21/2024] [Indexed: 07/31/2024] Open
Abstract
Background Acquired reactive perforating collagenosis (ARPC) poses a clinical challenge with an unclear pathogenesis. This disease has been frequently proven resistant to immunosuppressive treatments, significantly affecting the quality of life of patients. In this report, we highlight the efficacy of baricitinib as a viable option for maintenance therapy in ARPC. Case summary An 81-year-old woman presented to our hospital with recurrent pruritus and cup-like ulcerated lesions on her trunk and limbs persisting for 1 year. She exhibited limited response to oral antihistamines and topical steroids. Past medical history revealed a prolonged history of coronary heart disease and type 2 diabetes spanning several years to decades. Histopathological examination revealed cup-shaped depressions filled with necrotic inflammatory debris. In the dermis, a mixed inflammatory infiltrate composed of lymphocytes and histiocytes was observed. Van Gieson staining indicated the elimination of fibrous tissue extending from the dermis into the epidermis. Consequently, a diagnosis of ARPC was established. Due to the inadequate response to conventional treatments and the severe itching, we initiated baricitinib therapy for ARPC, resulting in gradual symptom improvement. Follow-up assessments showed no adverse reactions and normal laboratory findings. Conclusion The case report suggests that baricitinib might offer significant therapeutic benefits for ARPC.
Collapse
Affiliation(s)
- Jianfeng Zheng
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institue of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yangfeng Ding
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institue of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yuanyuan Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institue of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institue of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yunlu Gao
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institue of Psoriasis, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Paroli M, Becciolini A, Lo Gullo A, Parisi S, Bravi E, Andracco R, Nucera V, Ometto F, Lumetti F, Farina A, Del Medico P, Colina M, Ravagnani V, Scolieri P, Larosa M, Priora M, Visalli E, Addimanda O, Vitetta R, Volpe A, Bezzi A, Girelli F, Molica Colella AB, Caccavale R, Di Donato E, Adorni G, Santilli D, Lucchini G, Arrigoni E, Platè I, Mansueto N, Ianniello A, Fusaro E, Ditto MC, Bruzzese V, Camellino D, Bianchi G, Serale F, Foti R, Amato G, De Lucia F, Dal Bosco Y, Foti R, Reta M, Fiorenza A, Rovera G, Marchetta A, Focherini MC, Mascella F, Bernardi S, Sandri G, Giuggioli D, Salvarani C, De Andres MI, Franchina V, Molica Colella F, Ferrero G, Raffeiner B, Ariani A. Influence of Safety Warnings on the Prescribing Attitude of JAK Inhibitors for Rheumatoid Arthritis in Italy. J Clin Med 2024; 13:3929. [PMID: 38999494 PMCID: PMC11242683 DOI: 10.3390/jcm13133929] [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: 06/09/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Background/Objectives: The Janus kinase inhibitors (JAKi) tofacitinib (TOFA), baricitinib (BARI), upadacitinib (UPA), and filgotinib (FILGO) are effective drugs for the treatment of rheumatoid arthritis. However, the US Food and Drug Administration (FDA) raised concerns about the safety of TOFA after its approval. This prompted the European Medicines Agency (EMA) to issue two safety warnings for limiting TOFA use, then extended a third warning to all JAKi in patients at high risk of developing serious adverse effects (SAE). These include thrombosis, major adverse cardiac events (MACE), and cancer. The purpose of this work was to analyze how the first two safety warnings from the EMA affected the prescribing of JAKi by rheumatologists in Italy. Methods: All patients with rheumatoid arthritis who had been prescribed JAKi for the first time in a 36-month period from 1 July 2019, to 30 June 2022 were considered. Data were obtained from the medical records of 29 Italian tertiary referral rheumatology centers. Patients were divided into three groups of 4 months each, depending on whether the JAKi prescription had occurred before the EMA's first safety alert (1 July-31 October 2019, Group 1), between the first and second alerts (1 November 2019-29 February 2020, Group 2), or between the second and third alerts (1 March 2021-30 June 2021, Group 3). The percentages and absolute changes in the patients prescribed the individual JAKi were analyzed. Differences among the three groups of patients regarding demographic and clinical characteristics were also assessed. Results: A total of 864 patients were prescribed a JAKi during the entire period considered. Of these, 343 were identified in Group 1, 233 in Group 2, and 288 in Group 3. An absolute reduction of 32% was observed in the number of patients prescribed a JAKi between Group 1 and Group 2 and 16% between Group 1 and Group 3. In contrast, there was a 19% increase in the prescription of a JAKi in patients between Group 2 and Group 3. In the first group, BARI was the most prescribed drug (227 prescriptions, 66.2% of the total), followed by TOFA (115, 33.5%) and UPA (1, 0.3%). In the second group, the most prescribed JAKi was BARI (147, 63.1%), followed by TOFA (65, 27.9%) and UPA (33, 11.5%). In the third group, BARI was still the most prescribed JAKi (104 prescriptions, 36.1%), followed by UPA (89, 30.9%), FILGO (89, 21.5%), and TOFA (33, 11.5%). The number of patients prescribed TOFA decreased significantly between Group 1 and Group 2 and between Group 2 and Group 3 (p ˂ 0.01). The number of patients who were prescribed BARI decreased significantly between Group 1 and Group 2 and between Group 2 and Group 3 (p ˂ 0.01). In contrast, the number of patients prescribed UPA increased between Group 2 and Group 3 (p ˂ 0.01). Conclusions: These data suggest that the warnings issued for TOFA were followed by a reduction in total JAKi prescriptions. However, the more selective JAKi (UPA and FILGO) were perceived by prescribers as favorable in terms of the risk/benefit ratio, and their use gradually increased at the expense of the other molecules.
Collapse
Affiliation(s)
- Marino Paroli
- Department of Clinical, Internist, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Becciolini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy
| | | | - Simone Parisi
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Elena Bravi
- Rheumatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
| | - Romina Andracco
- Rheumatology Unit, ASL1 Liguria, 18038 Bussana di Sanremo, Italy
| | | | | | - Federica Lumetti
- Rheumatology Unit, Azienda USL of Modena and AOU Policlinico of Modena, 41100 Modena, Italy
| | - Antonella Farina
- Internal Medicine Unit, Augusto Murri Hospital, 63900 Fermo, Italy
| | - Patrizia Del Medico
- Internal Medicine Unit, Civitanova Marche Hospital, 62012 Civitanova Marche, Italy
| | - Matteo Colina
- Rheumatology Unit, Internal Medicine Division, Department of Medicine and Oncology, Santa Maria della Scaletta Hospital, 40026 Imola, Italy
- Rheumatology Unit, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy
| | - Viviana Ravagnani
- Rheumatology Unit, Santa Chiara Hospital APSS-Trento, 38122 Trento, Italy
| | - Palma Scolieri
- Rheumatology Unit, Nuovo Regina Margherita Hospital, 00154 Rome, Italy
| | - Maddalena Larosa
- Division of Rheumatology, Department of Medical Specialties, Azienda Sanitaria Locale 3 Genovese, 16132 Genoa, Italy
| | | | - Elisa Visalli
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy
| | - Olga Addimanda
- Rheumatology Unit, AUSL of Bologna-Policlinico Sant'Orsola-AOU-IRCCS of Bologna, 40138 Bologna, Italy
| | - Rosetta Vitetta
- Rheumatology Unit, ASL VC Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Alessandro Volpe
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | - Alessandra Bezzi
- Internal Medicine and Rheumatology Unit, AUSL della Romagna-Rimini, 47924 Rimini, Italy
| | - Francesco Girelli
- Rheumatology Unit, G.B. Morgagni-L. Pierantoni Hospital, 47121 Forlì, Italy
| | | | - Rosalba Caccavale
- Department of Clinical, Internist, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Eleonora Di Donato
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Giuditta Adorni
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Daniele Santilli
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Gianluca Lucchini
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Eugenio Arrigoni
- Rheumatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
| | - Ilaria Platè
- Rheumatology Unit, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy
| | - Natalia Mansueto
- Rheumatology Unit, ASL1 Liguria, 18038 Bussana di Sanremo, Italy
| | | | - Enrico Fusaro
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Maria Chiara Ditto
- Rheumatology Department, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Vincenzo Bruzzese
- Rheumatology Unit, Nuovo Regina Margherita Hospital, 00154 Rome, Italy
| | - Dario Camellino
- Division of Rheumatology, Department of Medical Specialties, Azienda Sanitaria Locale 3 Genovese, 16132 Genoa, Italy
| | - Gerolamo Bianchi
- Division of Rheumatology, Department of Medical Specialties, Azienda Sanitaria Locale 3 Genovese, 16132 Genoa, Italy
| | | | - Rosario Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy
| | - Giorgio Amato
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy
| | | | - Ylenia Dal Bosco
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy
| | - Roberta Foti
- Rheumatology Unit, Policlinico San Marco Hospital, 95121 Catania, Italy
| | - Massimo Reta
- Rheumatology Unit, AUSL of Bologna-Policlinico Sant'Orsola-AOU-IRCCS of Bologna, 40138 Bologna, Italy
| | - Alessia Fiorenza
- Rheumatology Unit, ASL VC Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Guido Rovera
- Rheumatology Unit, ASL VC Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Antonio Marchetta
- Rheumatology Unit, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar di Valpolicella, Italy
| | | | - Fabio Mascella
- Internal Medicine and Rheumatology Unit, AUSL della Romagna-Rimini, 47924 Rimini, Italy
| | - Simone Bernardi
- Rheumatology Unit, G.B. Morgagni-L. Pierantoni Hospital, 47121 Forlì, Italy
| | - Gilda Sandri
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Dilia Giuggioli
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Carlo Salvarani
- Rheumatology Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | | | - Veronica Franchina
- Medical Oncology Unit, Azienda Ospedaliera Papardo, 98158 Messina, Italy
| | | | - Giulio Ferrero
- Unit of Diagnostic and Interventional Radiology, Santa Corona Hospital, 17027 Pietra Ligure, Italy
| | - Bernd Raffeiner
- Department of Rheumatology, Bolzano Central Hospital, 39100 Bolzano, Italy
| | - Alarico Ariani
- Internal Medicine and Rheumatology Unit, University Hospital of Parma, 43126 Parma, Italy
| |
Collapse
|
16
|
Ferrara F, Zovi A, Capuozzo M, Langella R. Atopic dermatitis: treatment and innovations in immunotherapy. Inflammopharmacology 2024; 32:1777-1789. [PMID: 38581639 DOI: 10.1007/s10787-024-01453-8] [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: 12/22/2023] [Accepted: 02/25/2024] [Indexed: 04/08/2024]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disease characterized by itching and skin barrier dysfunction. Moderate to severe AD is often refractory to first-line topical treatments, and systemic immunosuppressants have been shown to be effective but have significant adverse effects. The paucity of basic treatments has contributed to the development of targeted topical and systemic immunotherapies based on the use of small molecules and biologic drugs which can directly interact with AD pathogenetic pathways. They represent a new era of therapeutic innovation. Additional new treatments are desirable since AD is a heterogeneous disease marked by different immunological phenotypes. This manuscript will review the mechanism of action, safety profile, and efficacy of promising new systemic immunological treatments for AD. Since moderate to severe AD can result in poor quality of life, the development of targeted and well-tolerated immunomodulators is a crucial purpose. The introduction of new pharmacological agents may offer new therapeutic options. However, there is the need to evaluate how "narrow-acting" agents, such as individual interleukin inhibitors, will perform under the safety and efficacy profiles compared with "broad-acting" agents, such as JAK inhibitors.
Collapse
Affiliation(s)
- Francesco Ferrara
- Pharmaceutical Department, Asl Napoli 3 Sud, Dell'Amicizia Street, 22, 80035, Nola, Naples, Italy
| | - Andrea Zovi
- Ministry of Health, Viale Giorgio Ribotta, 5, 00144, Rome, Italy.
| | - Maurizio Capuozzo
- Pharmaceutical Department, Asl Napoli 3 Sud, Marittima Street, 3, 80056, Ercolano, Naples, Italy
| | - Roberto Langella
- Italian Society of Hospital Pharmacy (SIFO), SIFO Secretariat of the Lombardy Region, Via Carlo Farini, 81, 20159, Milan, Italy
| |
Collapse
|
17
|
Recchia Luciani G, Barilli A, Visigalli R, Sala R, Dall’Asta V, Rotoli BM. IRF1 Mediates Growth Arrest and the Induction of a Secretory Phenotype in Alveolar Epithelial Cells in Response to Inflammatory Cytokines IFNγ/TNFα. Int J Mol Sci 2024; 25:3463. [PMID: 38542436 PMCID: PMC10970306 DOI: 10.3390/ijms25063463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
In COVID-19, cytokine release syndrome can cause severe lung tissue damage leading to acute respiratory distress syndrome (ARDS). Here, we address the effects of IFNγ, TNFα, IL-1β and IL-6 on the growth arrest of alveolar A549 cells, focusing on the role of the IFN regulatory factor 1 (IRF1) transcription factor. The efficacy of JAK1/2 inhibitor baricitinib has also been tested. A549 WT and IRF1 KO cells were exposed to cytokines for up to 72 h. Cell proliferation and death were evaluated with the resazurin assay, analysis of cell cycle and cycle-regulator proteins, LDH release and Annexin-V positivity; the induction of senescence and senescence-associated secretory phenotype (SASP) was evaluated through β-galactosidase staining and the quantitation of secreted inflammatory mediators. While IL-1 and IL-6 proved ineffective, IFNγ plus TNFα caused a proliferative arrest in A549 WT cells with alterations in cell morphology, along with the acquisition of a secretory phenotype. These effects were STAT and IRF1-dependent since they were prevented by baricitinib and much less evident in IRF1 KO than in WT cells. In alveolar cells, STATs/IRF1 axis is required for cytokine-induced proliferative arrest and the induction of a secretory phenotype. Hence, baricitininb is a promising therapeutic strategy for the attenuation of senescence-associated inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Valeria Dall’Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy (R.S.); (B.M.R.)
| | | |
Collapse
|
18
|
Faris A, Cacciatore I, Alnajjar R, Hanine H, Aouidate A, Mothana RA, Alanzi AR, Elhallaoui M. Revealing innovative JAK1 and JAK3 inhibitors: a comprehensive study utilizing QSAR, 3D-Pharmacophore screening, molecular docking, molecular dynamics, and MM/GBSA analyses. Front Mol Biosci 2024; 11:1348277. [PMID: 38516192 PMCID: PMC10956358 DOI: 10.3389/fmolb.2024.1348277] [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: 12/02/2023] [Accepted: 01/17/2024] [Indexed: 03/23/2024] Open
Abstract
The heterocycle compounds, with their diverse functionalities, are particularly effective in inhibiting Janus kinases (JAKs). Therefore, it is crucial to identify the correlation between their complex structures and biological activities for the development of new drugs for the treatment of rheumatoid arthritis (RA) and cancer. In this study, a diverse set of 28 heterocyclic compounds selective for JAK1 and JAK3 was employed to construct quantitative structure-activity relationship (QSAR) models using multiple linear regression (MLR). Artificial neural network (ANN) models were employed in the development of QSAR models. The robustness and stability of the models were assessed through internal and external methodologies, including the domain of applicability (DoA). The molecular descriptors incorporated into the model exhibited a satisfactory correlation with the receptor-ligand complex structures of JAKs observed in X-ray crystallography, making the model interpretable and predictive. Furthermore, pharmacophore models ADRRR and ADHRR were designed for each JAK1 and JAK3, proving effective in discriminating between active compounds and decoys. Both models demonstrated good performance in identifying new compounds, with an ROC of 0.83 for the ADRRR model and an ROC of 0.75 for the ADHRR model. Using a pharmacophore model, the most promising compounds were selected based on their strong affinity compared to the most active compounds in the studied series each JAK1 and JAK3. Notably, the pharmacokinetic, physicochemical properties, and biological activities of the selected compounds (As compounds ZINC79189223 and ZINC66252348) were found to be consistent with their therapeutic effects in RA, owing to their non-toxic, cholinergic nature, absence of P-glycoprotein, high gastrointestinal absorption, and ability to penetrate the blood-brain barrier. Furthermore, ADMET properties were assessed, and molecular dynamics and MM/GBSA analysis revealed stability in these molecules.
Collapse
Affiliation(s)
- Abdelmoujoud Faris
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ivana Cacciatore
- Department of Pharmacy, University ‘G. d’Annunzio’ of Chieti-Pescara, Chieti, Italy
| | - Radwan Alnajjar
- CADD Unit, PharmD, Faculty of Pharmacy, Libyan International Medical University, Benghazi, Libya
| | - Hadni Hanine
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Adnane Aouidate
- School of Applied Sciences of Ait Melloul, Ibn Zohr University, Fez, Morocco
| | - Ramzi A. Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah R. Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Menana Elhallaoui
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
19
|
Fiehn C, Leipe J, Weseloh C, Bergner R, Krüger K. Assessment of interactions and dosage recommendations of synthetic DMARDs-Evidence-based and consensus-based recommendations based on a systematic literature search. Z Rheumatol 2024; 83:8-19. [PMID: 37831190 DOI: 10.1007/s00393-023-01417-3] [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] [Accepted: 08/14/2023] [Indexed: 10/14/2023]
Abstract
Conventional synthetic (cs) and targeted synthetic (ts) disease-modifying antirheumatic drugs (DMARD) have potential interactions with a multitude of drugs. Furthermore, they sometimes have a lower therapeutic index, particularly in cases of limited organ functions. The aim of this work was to establish evidence-based recommendations on the therapeutic use of DMARDs in the context of drug interactions and dosage recommendations. A systematic literature search was carried out on the issue of drug interactions and dosages in cases of patients with limited kidney function and higher age and suffering from rheumatoid arthritis. A total of 2756 scientific publications were screened and 154 selected of which 68 were scrutinized in detail. Furthermore, the respective product information was also analyzed. A multitude of possible interactions of synthetic DMARDs with different drugs were detected, which were then assessed with respect to the clinical significance and consequences. A consensus process led to making recommendations with which the interactions were classified: A: dangerous combination, B: avoid combination (if possible, pausing DMARD treatment), C: possible combination requiring increased monitoring and potential adjustments in dosage and D: pharmacological interaction without relevance in DMARD standard doses. Apart from that dosage recommendations were established for each csDMARD and tsDMARD depending on kidney function and age. There are 3 primary recommendations and 11 core recommendations on interactions and dosages of csDMARDs and tsDMARDs meant as a practical help for therapeutic decision making and to improve safety in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- C Fiehn
- Kommission Pharmakotherapie, Deutsche Gesellschaft für Rheumatologie (DGRh), Berlin, Germany.
- Tätigkeitsschwerpunkt klinische Immunologie, Rheumatologie Baden-Baden, Beethovenstr. 2, 76530, Baden-Baden, Germany.
| | - J Leipe
- Kommission Pharmakotherapie, Deutsche Gesellschaft für Rheumatologie (DGRh), Berlin, Germany
- V. Medizinische Klinik, Universitätsmedizin Mannheim, Mannheim, Germany
| | - C Weseloh
- Kommission Pharmakotherapie, Deutsche Gesellschaft für Rheumatologie (DGRh), Berlin, Germany
- Deutsche Gesellschaft für Rheumatologie (DGRh), Berlin, Germany
| | - R Bergner
- Kommission Pharmakotherapie, Deutsche Gesellschaft für Rheumatologie (DGRh), Berlin, Germany
- Medizinische Klinik A, Klinikum Ludwigshafen, Ludwigshafen, Germany
| | - K Krüger
- Kommission Pharmakotherapie, Deutsche Gesellschaft für Rheumatologie (DGRh), Berlin, Germany
- Rheumatologisches Praxiszentrum St. Bonifatius, Munich, Germany
| |
Collapse
|
20
|
Martinez J, Manjaly C, Manjaly P, Ly S, Zhou G, Barbieri J, Mostaghimi A. Janus Kinase Inhibitors and Adverse Events of Acne: A Systematic Review and Meta-Analysis. JAMA Dermatol 2023; 159:1339-1345. [PMID: 37851459 PMCID: PMC10585588 DOI: 10.1001/jamadermatol.2023.3830] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/31/2023] [Indexed: 10/19/2023]
Abstract
Importance Janus kinase (JAK) inhibitors are increasingly used across a range of dermatologic conditions. Adverse events of acne have been noted in some studies in clinical practice, but the scope of this outcome across JAK inhibitors has not been established. Objective To systematically analyze all published phase 2 and 3 placebo-controlled randomized clinical trials (RCTs) of JAK inhibitors for the risk of acne as an adverse effect of these medications. Data Sources Comprehensive search of Ovid MEDLINE and PubMed databases through January 31, 2023. Study Selection Inclusion criteria were phase 2 and 3 placebo-controlled RCTs of JAK inhibitors published in English with reported adverse events of acne. Data Extraction and Synthesis Two reviewers independently reviewed and extracted information from all included studies. Main Outcomes and Measures The primary outcome of interest was the incidence of acne following JAK inhibitor use. A meta-analysis was conducted using random-effects models. Results A total of 25 unique studies (10 839 unique participants; 54% male and 46% female) were included in the final analysis. The pooled odds ratio (OR) was calculated to be 3.83 (95% CI, 2.76-5.32) with increased ORs for abrocitinib (13.47 [95% CI, 3.25-55.91]), baricitinib (4.96 [95% CI, 2.52-9.78]), upadacitinib (4.79 [95% CI, 3.61-6.37]), deucravacitinib (2.64 [95% CI, 1.44-4.86]), and deuruxolitinib (3.30 [95% CI, 1.22-8.93]). Estimated ORs were higher across studies investigating the use of JAK inhibitors for the management of dermatologic compared with nondermatologic conditions (4.67 [95% CI, 3.10-7.05]) as well as for JAK1-specific inhibitors (4.69 [95% CI, 3.56-6.18]), combined JAK1 and JAK2 inhibitors (3.43 [95% CI, 2.14-5.49]), and tyrosine kinase 2 inhibitors (2.64 [95% CI, 1.44-4.86]). Conclusions and Relevance In this systematic review and meta-analysis, JAK inhibitor use was associated with an elevated odds of acne. Patients should be properly counseled on this potential adverse effect of these medications before treatment initiation. Future studies are needed to further elucidate the pathophysiology of this association.
Collapse
Affiliation(s)
- Jeremy Martinez
- Harvard Medical School, Boston, Massachusetts
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Priya Manjaly
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
- Boston University School of Medicine, Boston, Massachusetts
| | - Sophia Ly
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock
| | - Guohai Zhou
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - John Barbieri
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Arash Mostaghimi
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
21
|
Xue Y, Mei H, Chen Y, Griffin JD, Liu Q, Weisberg E, Yang J. Repurposing clinically available drugs and therapies for pathogenic targets to combat SARS-CoV-2. MedComm (Beijing) 2023; 4:e254. [PMID: 37193304 PMCID: PMC10183156 DOI: 10.1002/mco2.254] [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: 12/02/2022] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 05/18/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has affected a large portion of the global population, both physically and mentally. Current evidence suggests that the rapidly evolving coronavirus subvariants risk rendering vaccines and antibodies ineffective due to their potential to evade existing immunity, with enhanced transmission activity and higher reinfection rates that could lead to new outbreaks across the globe. The goal of viral management is to disrupt the viral life cycle as well as to relieve severe symptoms such as lung damage, cytokine storm, and organ failure. In the fight against viruses, the combination of viral genome sequencing, elucidation of the structure of viral proteins, and identifying proteins that are highly conserved across multiple coronaviruses has revealed many potential molecular targets. In addition, the time- and cost-effective repurposing of preexisting antiviral drugs or approved/clinical drugs for these targets offers considerable clinical advantages for COVID-19 patients. This review provides a comprehensive overview of various identified pathogenic targets and pathways as well as corresponding repurposed approved/clinical drugs and their potential against COVID-19. These findings provide new insight into the discovery of novel therapeutic strategies that could be applied to the control of disease symptoms emanating from evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yiying Xue
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
- University of Science and Technology of ChinaHefeiAnhuiChina
| | - Yisa Chen
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - James D. Griffin
- Department of Medical Oncology, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medicine, Harvard Medical SchoolBostonMassachusettsUSA
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
- University of Science and Technology of ChinaHefeiAnhuiChina
- Hefei Cancer HospitalChinese Academy of SciencesHefeiChina
| | - Ellen Weisberg
- Department of Medical Oncology, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medicine, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jing Yang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
| |
Collapse
|
22
|
Freitas E, Guttman-Yassky E, Torres T. Baricitinib for the Treatment of Alopecia Areata. Drugs 2023:10.1007/s40265-023-01873-w. [PMID: 37195491 DOI: 10.1007/s40265-023-01873-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 05/18/2023]
Abstract
Alopecia areata (AA) is a relapsing, chronic, immune-mediated disease characterized by nonscarring, inflammatory hair loss that can affect any hair-bearing site. AA clinical presentation is heterogeneous. Its pathogenesis involves immune and genetic factors and several pro-inflammatory cytokines involved in AA pathogenesis, including interleukin-15 and interferon-γ, as well as Th2 cytokines, such as IL-4/IL-13, that signal through Janus kinase (JAK) pathway. AA treatment aims to stop its progression and reverse hair loss, and JAK inhibition has been shown to stop hair loss and reverse alopecia and has exhibited promising results in treating AA in clinical trials. Baricitinib, an oral, reversible, selective JAK1/JAK2 inhibitor, was shown to be superior to placebo on hair growth after 36 weeks of treatment in adults with severe AA in a phase 2 trial and recently in two phase 3 trials (BRAVE-AA1 and BRAVE-AA2). In both studies, the most common adverse events were upper respiratory tract infections, urinary tract infection, acne, headache, and elevated creatine kinase levels. On the basis of these trial results, baricitinib was recently approved by the European Medicines Agency (EMA) and US Food and Drug Administration (FDA) for the treatment of adults with severe AA. Nevertheless, longer trials are needed to determine the long-term efficacy and safety of baricitinib in AA. Current trials are ongoing and are planned to remain randomized and blinded for up to 200 weeks.
Collapse
Affiliation(s)
- Egídio Freitas
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Edifício das Consultas Externas, Ex. CICAP, Rua D. Manuel II, s/n, 4100, Porto, Portugal
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Tiago Torres
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Edifício das Consultas Externas, Ex. CICAP, Rua D. Manuel II, s/n, 4100, Porto, Portugal.
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.
| |
Collapse
|
23
|
Hartinger R, Lederer EM, Schena E, Lattanzi G, Djabali K. Impact of Combined Baricitinib and FTI Treatment on Adipogenesis in Hutchinson-Gilford Progeria Syndrome and Other Lipodystrophic Laminopathies. Cells 2023; 12:1350. [PMID: 37408186 DOI: 10.3390/cells12101350] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease that causes premature aging symptoms, such as vascular diseases, lipodystrophy, loss of bone mineral density, and alopecia. HGPS is mostly linked to a heterozygous and de novo mutation in the LMNA gene (c.1824 C > T; p.G608G), resulting in the production of a truncated prelamin A protein called "progerin". Progerin accumulation causes nuclear dysfunction, premature senescence, and apoptosis. Here, we examined the effects of baricitinib (Bar), an FDA-approved JAK/STAT inhibitor, and a combination of Bar and lonafarnib (FTI) treatment on adipogenesis using skin-derived precursors (SKPs). We analyzed the effect of these treatments on the differentiation potential of SKPs isolated from pre-established human primary fibroblast cultures. Compared to mock-treated HGPS SKPs, Bar and Bar + FTI treatments improved the differentiation of HGPS SKPs into adipocytes and lipid droplet formation. Similarly, Bar and Bar + FTI treatments improved the differentiation of SKPs derived from patients with two other lipodystrophic diseases: familial partial lipodystrophy type 2 (FPLD2) and mandibuloacral dysplasia type B (MADB). Overall, the results show that Bar treatment improves adipogenesis and lipid droplet formation in HGPS, FPLD2, and MADB, indicating that Bar + FTI treatment might further ameliorate HGPS pathologies compared to lonafarnib treatment alone.
Collapse
Affiliation(s)
- Ramona Hartinger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Eva-Maria Lederer
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Elisa Schena
- Unit of Bologna, CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giovanna Lattanzi
- Unit of Bologna, CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| |
Collapse
|
24
|
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
|
25
|
Yu Y, Ding X, Guo F, Ze K, Sun X, Li X. Perifolliculitis capitis abscedens et suffodiens treatment with tumor necrosis factor inhibitors and baricitinib: A case report and literature review. Front Med (Lausanne) 2023; 10:1132574. [PMID: 37056728 PMCID: PMC10086126 DOI: 10.3389/fmed.2023.1132574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
RationalePerifolliculitis capitis abscedens et suffodiens (PCAS), also known as dissecting cellulitis of the scalp (DCS), is a part of the “follicular occlusion tetrad” that also includes acne conglobate (AC), hidradenitis suppurativa (HS), and pilonidal sinus, which share the same pathogenic mechanism, such as follicular occlusions, follicular ruptures, and follicular infections.Patient concernsA 15-year-old boy had multiple rashes on the scalp accompanied by pain.DiagnosisThe patient was diagnosed with PCAS or DCS based on the clinical manifestations and laboratory examinations.InterventionsThe patient was initially administered adalimumab 40 mg biweekly and oral isotretinoin 30 mg daily for 5 months. Because the initial results were insufficient, the interval between adalimumab injections was extended to 4 weeks, and isotretinoin was changed to baricitinib 4 mg daily for 2 months. When the condition became more stable, adalimumab 40 mg and baricitinib 4 mg were administered every 20 and 3 days, respectively, for two more months until now.OutcomesAfter 9 months of treatment and follow-up, the original skin lesions of the patient were almost cured, and most inflammatory alopecia patches disappeared.ConclusionOur literature review did not find any previous reports on treating PCAS with TNF-α inhibitors and baricitinib. Accordingly, we presented the first successful treatment of PCAS with this regimen.
Collapse
Affiliation(s)
- Yuanting Yu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojie Ding
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kan Ze
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoying Sun
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Xiaoying Sun,
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Xin Li,
| |
Collapse
|
26
|
[Assessment of interactions and dosage recommendations of synthetic DMARDs-Evidence-based and consensus-based recommendations based on a systematic literature search]. Z Rheumatol 2023; 82:151-162. [PMID: 36633662 DOI: 10.1007/s00393-022-01308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2022] [Indexed: 01/13/2023]
Abstract
Conventional synthetic (cs) and targeted synthetic (ts) disease-modifying antirheumatic drugs (DMARD) have potential interactions with a multitude of drugs. Furthermore, they sometimes have a lower therapeutic index, particularly in cases of limited organ functions. The aim of this work was to establish evidence-based recommendations on the therapeutic use of DMARDs in the context of drug interactions and dosage recommendations. A systematic literature search was carried out on the issue of drug interactions and dosages in cases of patients with limited kidney function and higher age and suffering from rheumatoid arthritis. A total of 2756 scientific publications were screened and 154 selected of which 68 were scrutinized in detail. Furthermore, the respective specialist subject information was also analyzed. A multitude of possible interactions of synthetic DMARDs with different drugs were detected, which were then assessed with respect to the clinical significance and consequences. A consensus process led to making recommendations with which the interactions were classified: A: dangerous combination, B: avoid combination (if possible, pausing DMARD treatment), C: possible combination requiring increased monitoring and potential adjustments in dosage and D: pharmacological interaction without relevance in DMARD standard doses. Apart from that dosage recommendations were established for each csDMARD and tsDMARD depending on kidney function and age. There are 3 primary recommendations and 11 core recommendations on interactions and dosages of csDMARDs and tsDMARDs meant as a practical help for therapeutic decision making and to improve safety in the treatment of rheumatoid arthritis.
Collapse
|
27
|
Atluri K, Manne S, Nalamothu V, Mantel A, Sharma PK, Babu RJ. Advances in Current Drugs and Formulations for the Management of Atopic Dermatitis. Crit Rev Ther Drug Carrier Syst 2023; 40:1-87. [PMID: 37585309 DOI: 10.1615/critrevtherdrugcarriersyst.2023042979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease with a complex pathophysiology. Treatment of AD remains challenging owing to the presence of a wide spectrum of clinical phenotypes and limited response to existing therapies. However, recent genetic, immunological, and pathophysiological insights into the disease mechanism resulted in the invention of novel therapeutic drug candidates. This review provides a comprehensive overview of current therapies and assesses various novel drug delivery strategies currently under clinical investigation. Further, this review majorly emphasizes on various topical treatments including emollient therapies, barrier repair agents, topical corticosteroids (TCS), phosphodiesterase 4 (PDE4) inhibitors, calcineurin inhibitors, and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway inhibitors. It also discusses biological and systemic therapies, upcoming treatments based on ongoing clinical trials. Additionally, this review scrutinized the use of pharmaceutical inactive ingredients in the approved topical dosage forms for AD treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
28
|
Rheumatoid arthritis: advances in treatment strategies. Mol Cell Biochem 2023; 478:69-88. [PMID: 35725992 DOI: 10.1007/s11010-022-04492-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/31/2022] [Indexed: 01/17/2023]
Abstract
Rheumatoid arthritis (RA) is characterised by severe joint and bone damage due to heightened autoimmune response at the articular sites. Worldwide annual incidence and prevalence rate of RA is 3 cases per 10,000 population and 1%, respectively. Several genetic and environmental (microbiota, smoking, infectious agents) factors contribute to its pathogenesis. Although convention treatment strategies, predominantly Disease Modifying Anti Rheumatic Drugs (DMARDs) and Glucocorticoids (GC), are unchanged as the primary line of treatment; novel strategies consisting of biological DMARDs, are being developed and explored. Personalized approaches using biologicals targetspecific pathways associated with disease progression. However, considering the economic burden and side-effects associated with these, there is an unmet need on strategies for early stratification of the inadequate responders with cDMARDs. As RA is a complex disease with a variable remission rate, it is important not only to evaluate the current status of drugs in clinical practice but also those with the potential of personalised therapeutics. Here, we provide comprehensive data on the treatment strategies in RA, including studies exploring various combination strategies in clinical trials. Our systematic analysis of current literature found that conventional DMARDs along with glucocorticoid may be best suited for early RA cases and a combination of conventional and targeted DMARDs could be effective for treating seronegative patients with moderate to high RA activity. Clinical trials with insufficient responders to Methotrexate suggest that adding biologicals may help in such cases. However, certain adverse events associated with the current therapy advocate exploring novel therapeutic approaches such as gene therapy, mesenchymal stem cell therapy in future.
Collapse
|
29
|
Dogra S, Shah S, Sharma A, Chhabra S, Narang T. Emerging role of baricitinib in dermatology practice: All we need to know! Indian Dermatol Online J 2023; 14:153-162. [PMID: 37089829 PMCID: PMC10115327 DOI: 10.4103/idoj.idoj_542_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/13/2022] [Accepted: 12/31/2022] [Indexed: 03/12/2023] Open
Abstract
Baricitinib is a competitive inhibitor of the Janus Kinase family of non-receptor protein kinases, predominantly acting against JAK-1 and JAK-2 subtypes. By downregulating transcription of various pro-inflammatory cytokines, this drug has shown efficacy across various dermatoses. Approved for severe cases of alopecia areata and moderate-severe atopic dermatitis in adults, baricitinib is being increasingly tried across many other indications with promising results. It is prudent that dermatologists remain aware of boxed warnings and precautions with the use of this much-discussed molecule, including its infectious, thrombotic, cardiovascular, and malignant ramifications. Long-term data on the use of baricitinib in dermatological conditions are lacking and further research is warranted since most data on safety profile is extrapolated from its use in rheumatology. The present review aims to highlight the immunopathogenic mechanisms of JAK-1/2 blockade, approved and off-label uses in dermatology, along with a concise review of laboratory monitoring and the side-effect profile of baricitinib.
Collapse
|
30
|
Agashe RP, Lippman SM, Kurzrock R. JAK: Not Just Another Kinase. Mol Cancer Ther 2022; 21:1757-1764. [PMID: 36252553 PMCID: PMC10441554 DOI: 10.1158/1535-7163.mct-22-0323] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/07/2022] [Accepted: 10/07/2022] [Indexed: 01/12/2023]
Abstract
The JAK/STAT axis is implicated in cancer, inflammation, and immunity. Numerous cytokines/growth factors affect JAK/STAT signaling. JAKs (JAK1, JAK2, JAK3, and TYK2) noncovalently associate with cytokine receptors, mediate receptor tyrosine phosphorylation, and recruit ≥1 STAT proteins (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6). Tyrosine-phosphorylated STATs dimerize and are then transported into the nucleus to function as transcription factors. Signaling is attenuated by specific suppressor of cytokine signaling proteins, creating a negative feedback loop. Both germline mutations and polymorphisms of JAK family members correlate with specific diseases: Systemic lupus erythematosus (TYK2 polymorphisms); severe combined immunodeficiency (JAK3 mutations); pediatric acute lymphoblastic leukemia (TYK2 mutations); and hereditary thrombocytosis (JAK2 mutations). Somatic gain-of-function JAK mutations mainly occur in hematologic malignancies, with the activating JAK2 V617F being a myeloproliferative disorder hallmark; it is also seen in clonal hematopoiesis of indeterminate potential. Several T-cell malignancies, as well as B-cell acute lymphoblastic leukemia, and acute megakaryoblastic leukemia also harbor JAK family somatic alterations. On the other hand, JAK2 copy-number loss is associated with immune checkpoint inhibitor resistance. JAK inhibitors (jakinibs) have been deployed in many conditions with JAK activation; they are approved in myeloproliferative disorders, rheumatoid and psoriatic arthritis, atopic dermatitis, ulcerative colitis, graft-versus-host disease, alopecia areata, ankylosing spondylitis, and in patients hospitalized for COVID-19. Clinical trials are investigating jakinibs in multiple other autoimmune/inflammatory conditions. Furthermore, dermatologic and neurologic improvements have been observed in children with Aicardi-Goutieres syndrome (a genetic interferonopathy) treated with JAK inhibitors.
Collapse
Affiliation(s)
| | | | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, Wisconsin
- Win Consortium, Paris, France
| |
Collapse
|
31
|
Sk MF, Kar P. Finding inhibitors and deciphering inhibitor-induced conformational plasticity in the Janus kinase via multiscale simulations. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2022; 33:833-859. [PMID: 36398489 DOI: 10.1080/1062936x.2022.2145352] [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: 08/17/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
The Janus kinase (JAK) is a master regulator of the JAK/STAT pathway. Dysregulation of this signalling cascade causes neuroinflammation and autoimmune disorders. Therefore, JAKs have been characterized as an attractive target for developing anti-inflammatory drugs. Nowadays, designing efficient, effective, and specific targeted therapeutics without being cytotoxic has gained interest. We performed the virtual screening of natural products in combination with pharmacological analyses. Subsequently, we performed molecular dynamics simulations to study the stability of the ligand-bound complexes and ligand-induced inactive conformations. Notably, inactive kinases display remarkable conformational plasticity; however, ligand-induced molecular mechanisms of these conformations are still poorly understood. Herein, we performed a free energy landscape analysis to explore the conformational plasticity of the JAK1 kinase. Leonurine, STOCK1N-68642, STOCK1N-82656, and STOCK1N-85809 bound JAK1 exhibited a smooth transition from an active (αC-in) to a completely inactive conformation (αC-out). Ligand binding induces disorders in the αC-helix. Molecular mechanics Poisson Boltzmann surface area (MM/PBSA) calculation suggested three phytochemicals, namely STOCK1N-68642, Epicatechin, and STOCK1N-98615, have higher binding affinity compared to other ligand molecules. The ligand-induced conformational plasticity revealed by our simulations differs significantly from the available crystal structures, which might help in designing allosteric drugs.
Collapse
Affiliation(s)
- M F Sk
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, India
| | - P Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, India
| |
Collapse
|
32
|
Berthe P, Scailteux LM, Lescoat A, Staumont D, Coiffier G, Guéret P, Dupuy A, Oger E, Droitcourt C. Oral Janus kinase inhibitors and venous thromboembolic events in atopic dermatitis: protocols for a case-time control study and a nested case-control study based on the French national health insurance (SNDS) cohort. BMJ Open 2022; 12:e059979. [PMID: 36130766 PMCID: PMC9494565 DOI: 10.1136/bmjopen-2021-059979] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 07/23/2022] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Atopic dermatitis (AD) is a highly prevalent, chronic, inflammatory skin disease. Several orally administered Janus kinase inhibitors (JAKis, including baricitinib, upadacitinib and abrocitinib) have received a marketing authorisation for AD.Clinical trials in rheumatoid arthritis (RA) have flagged up a potential risk of JAKi-induced venous thromboembolic events (VTEs). Accordingly, the summary of product characteristics for a JAKi must mention VTEs as potential adverse drug reactions. In contrast to RA, AD per se is not associated with an elevated risk of VTEs. Assessing this potential risk among patients with AD would shed further light on the putative underlying relationship between JAKis and VTEs.Our research question is to investigate whether JAKi administration increases the risk of VTEs in adults with AD. Our primary objective is to assess the risk of VTEs in adults with AD exposed to JAKis compared to AD adults not exposed to JAKis, and our secondary objective is to evaluate whether JAKi initiation acts as a trigger of VTEs in adults with AD within 3 months. METHODS AND ANALYSIS Hence, we have designed (1) a nested case-control study and (2) a case-time control study in a cohort of adults with AD with data from the French national health insurance system (2017-2025).Here, we describe the study protocol, our methodological choices and certain novel aspects, including the combined value of the two assumptions and the use of an exhaustive national health insurance database with potentially greater statistical power for studying rare events in the population of patients with AD at a low risk of VTEs (thus limiting the influence of confounding factors). ETHICS AND DISSEMINATION The protocol has been approved by an independent ethics committee and registered with the French National Data Protection Commission. The study's findings will be published in peer-reviewed scientific journals and presented at international conferences.
Collapse
Affiliation(s)
| | - Lucie-Marie Scailteux
- Pharmacovigilance and Pharmacoepidemiology, CHU Rennes, Rennes, France
- Univ Rennes, INSERM, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Alain Lescoat
- Univ Rennes, INSERM, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- Internal Medicine and Clinical Immunology, CHU Rennes, Rennes, France
| | - Delphine Staumont
- Department of Dermatology, Lille University Hospital Center, Lille, France
| | - Guillaume Coiffier
- Department of Rheumatology, CH Dinan, Dinan, France
- INSERM, INRA, Institut NUMECAN (Nutrition Metabolism and Cancer), Rennes, France
| | | | - Alain Dupuy
- Department of Dermatology, CHU Rennes, Rennes, France
- Univ Rennes, INSERM, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Emmanuel Oger
- Pharmacovigilance and Pharmacoepidemiology, CHU Rennes, Rennes, France
- Univ Rennes, INSERM, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Catherine Droitcourt
- Department of Dermatology, CHU Rennes, Rennes, France
- Univ Rennes, INSERM, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
33
|
Schlöder J, Shahneh F, Schneider FJ, Wieschendorf B. Boosting regulatory T cell function for the treatment of autoimmune diseases – That’s only half the battle! Front Immunol 2022; 13:973813. [PMID: 36032121 PMCID: PMC9400058 DOI: 10.3389/fimmu.2022.973813] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 01/04/2023] Open
Abstract
Regulatory T cells (Treg) represent a subset of specialized T cells that are essential for the regulation of immune responses and maintenance of peripheral tolerance. Once activated, Treg exert powerful immunosuppressive properties, for example by inhibiting T cell-mediated immune responses against self-antigens, thereby protecting our body from autoimmunity. Autoimmune diseases such as multiple sclerosis, rheumatoid arthritis or systemic lupus erythematosus, exhibit an immunological imbalance mainly characterized by a reduced frequency and impaired function of Treg. In addition, there has been increasing evidence that – besides Treg dysfunction – immunoregulatory mechanisms fail to control autoreactive T cells due to a reduced responsiveness of T effector cells (Teff) for the suppressive properties of Treg, a process termed Treg resistance. In order to efficiently treat autoimmune diseases and thus fully induce immunological tolerance, a combined therapy aimed at both enhancing Treg function and restoring Teff responsiveness could most likely be beneficial. This review provides an overview of immunomodulating drugs that are currently used to treat various autoimmune diseases in the clinic and have been shown to increase Treg frequency as well as Teff sensitivity to Treg-mediated suppression. Furthermore, we discuss strategies on how to boost Treg activity and function, and their potential use in the treatment of autoimmunity. Finally, we present a humanized mouse model for the preclinical testing of Treg-activating substances in vivo.
Collapse
Affiliation(s)
- Janine Schlöder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- *Correspondence: Janine Schlöder,
| | - Fatemeh Shahneh
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Franz-Joseph Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Björn Wieschendorf
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
34
|
Zhang P, Miyata KN, Nast CC, LaPage JA, Mahoney M, Nguyen S, Khan K, Wu Q, Adler SG, Dai T. Dual therapy with an angiotensin receptor blocker and a JAK1/2 inhibitor attenuates dialysate-induced angiogenesis and preserves peritoneal membrane structure and function in an experimental CKD rat model. ARCH ESP UROL 2022; 43:159-167. [PMID: 35946050 DOI: 10.1177/08968608221116956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Peritoneal dialysis (PD) is limited by reduced efficacy over time. We previously showed that a Janus kinase 1/2 inhibitor (JAK1/2i) reduced inflammation, hypervascularity and fibrosis induced by 4.25% dextrose dialysate (4.25%D) intraperitoneally (IP) infused for 10 days in rats with normal kidney function. JAK/STAT signalling mediates inflammatory pathways, including angiotensin signalling. We now tested the effect of long-term JAK1/2i and/or an angiotensin receptor blocker (ARB) on peritoneal membrane (PM) in polycystic kidneys (PCK) rats infused with 4.25%D. METHODS Except for controls, all PCK rats had a tunnelled PD catheter: (1) no infusions; (2) 4.25%D; (3) 4.25%D + JAK1/2i (5 mg/kg); (4) 4.25%D +losartan (5 mg/kg); and (5) 4.25%D + losartan +JAK1/2i (5 mg/kg each) IP BID × 16 weeks (N = 5/group). PM VEGFR2 staining areas and submesothelial compact zone (SMCZ) width were morphometrically measured. Peritoneal equilibration testing measured peritoneal ultrafiltration (UF) by calculating dialysate glucose at time 0 and 90 min (D/D0 glucose). RESULTS 4.25%D caused hypervascularity, SMCZ widening, fibrosis and UF functional decline in PCK rats. Angiogenesis was significantly attenuated by JAK1/2i ± ARB but not by ARB monotherapy. Both treatments reduced SMCZ area. UF was preserved consistently by dual therapy (p < 0.05) but with inconsistent responses by monotherapies. CONCLUSION Long-term JAK1/2i ± ARB reduced angiogenesis and fibrosis, and the combination consistently maintained UF. In clinical practice, angiotensin inhibition has been advocated to maintain residual kidney function. Our study suggests that adding JAK1/2i to angiotensin inhibition may preserve PM structure and UF.
Collapse
Affiliation(s)
- Pei Zhang
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.,Department of Nephrology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Kana N Miyata
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.,Division of Nephrology, Department of Internal Medicine, Saint Louis University, St Louis, MO, USA
| | - Cynthia C Nast
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Janine A LaPage
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Madisyn Mahoney
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sonny Nguyen
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kamran Khan
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Qiaoyuan Wu
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.,Department of Nephrology, the First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Sharon G Adler
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Tiane Dai
- Division of Nephrology and Hypertension, the Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
35
|
Fang YF, Liu JR, Chang SH, Kuo CF, See LC. Comparative safety of Janus kinase inhibitors and tumor necrosis factor inhibitors in patients undergoing treatment for rheumatoid arthritis. Int J Rheum Dis 2022; 25:1254-1262. [PMID: 35923107 DOI: 10.1111/1756-185x.14414] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Since 2010, biological disease-modifying antirheumatic drugs (bDMARDs) have been the dominant mode of treatment for rheumatoid arthritis (RA). However, the safety of DMARDs, such as tumor necrosis factor inhibitors (TNFis) and Janus kinase inhibitors (JAKis), in treating patients with RA is a concern. We compared the safety outcomes of JAKis and TNFis in RA patients in clinical settings. METHODS Patients diagnosed with RA between 2015 and 2017 were identified from the Taiwan National Health Insurance Research Database and followed till 2018. Propensity score stabilized weighting was used to balance the baseline characteristics of the JAKis and TNFis groups. The incidences of safety outcomes, namely cardiovascular (CV) events, tuberculosis (TB), total hip replacement (THR), total knee replacement (TKR), and all-cause mortality, were compared between the 2 study groups. RESULTS A total of 3179 patients with RA who were administered JAKis (n = 822) and TNFis (n = 2357) were included in this study. The mean follow-up duration was 2.02 years in the JAKis group and 2.10 in the TNFis group. All-cause mortality had the highest incidence rate, followed by TKR, THR, CV events, and TB. A lower incidence rate of the study outcomes was observed in the JAKis group than in the TNFis group but without statistical significance. CONCLUSION Comparable safety issues and mortality rates were observed for JAKis and TNFis in RA patients treated in real-world settings.
Collapse
Affiliation(s)
- Yao-Fan Fang
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Jia-Rou Liu
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Shu-Hao Chang
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chang-Fu Kuo
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
| | - Lai-Chu See
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan.,Department of Public Health, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Biostatistics Core Laboratory, Molecular Medicine Research Center, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
36
|
Ali E, Owais R, Sheikh A, Shaikh A. Olumniant (Baricitinib) oral tablets: An insight into FDA-approved systemic treatment for Alopecia Areata. Ann Med Surg (Lond) 2022; 80:104157. [PMID: 36045780 PMCID: PMC9422172 DOI: 10.1016/j.amsu.2022.104157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Eman Ali
- Corresponding author. Department of Internal Medicine Dow University of Health Sciences, Baba-e-Urdu Road, Saddar Karachi, Pakistan.
| | | | | | | |
Collapse
|
37
|
Efficacy and Safety of JAK Inhibitors for Rheumatoid Arthritis: A Meta-Analysis. J Clin Med 2022; 11:jcm11154459. [PMID: 35956078 PMCID: PMC9369647 DOI: 10.3390/jcm11154459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/14/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Background: More and more trials have been conducted. We aimed to assess the efficacy and safety of different JAKinibs in RA. Methods: A systematic search of randomized controlled trials (RCTs) with JAKinib treatment in RA published in the Medline, Embase, and Cochrane databases up to May 2021 was performed. Results: 37 trials involving 15,174 patients were identified. Pooled analysis revealed that JAKinibs were associated with significant therapeutic improvement in RA patients as determined by ACR20 (RR = 2.03, 95% CI: 1.85 to 2.28) and HAQ-DI (MD = −0.31, 95% CI: −0.33 to −0.28) over placebo. Compared to placebo, JAKinib treatment was also associated with more adverse events (RR = 1.10, p < 0.001; RR = 1.29, p < 0.001; RR = 1.59, p = 0.02). Baricitinib and upadacitinib were related to more frequent adverse events (RR = 1.10; 95% CI: 1.01, 1.21; RR = 1.19; 95% CI: 1.11, 1.28) and infection (RR = 1.22; 95% CI: 1.09, 1.37; RR = 1.38; 95% CI: 1.22, 1.56), whereas only baricitinib was associated with more herpes zoster (RR = 3.15; 95% CI: 1.19, 8.33). Conclusions: JAKinibs were superior to placebo for improving signs, symptoms, and health-related quality of life in RA patients at short term, whereas the overall risk of adverse events and infections were greater with baricitinib and upadacitinib, and a higher risk of herpes zoster was only associated with baricitinib. More trials are needed to investigate the long-term safety.
Collapse
|
38
|
Laux J, Forster M, Riexinger L, Schwamborn A, Guezguez J, Pokoj C, Kudolo M, Berger LM, Knapp S, Schollmeyer D, Guse J, Burnet M, Laufer SA. Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune Cells. ACS PHARMACOLOGY & TRANSLATIONAL SCIENCE 2022; 5:573-602. [DOI: 10.1021/acsptsci.2c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Julian Laux
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Michael Forster
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
| | - Laura Riexinger
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Anna Schwamborn
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Jamil Guezguez
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Christina Pokoj
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Mark Kudolo
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
| | - Lena M. Berger
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Dieter Schollmeyer
- Institute for Organic Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55099 Mainz, Germany
| | - Jan Guse
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Michael Burnet
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, 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 and Development (TüCAD2), 72076 Tübingen, Germany
| |
Collapse
|
39
|
Poudel G, Tolland MG, Hughes TP, Pagani IS. Mechanisms of Resistance and Implications for Treatment Strategies in Chronic Myeloid Leukaemia. Cancers (Basel) 2022; 14:cancers14143300. [PMID: 35884363 PMCID: PMC9317051 DOI: 10.3390/cancers14143300] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Chronic myeloid leukaemia (CML) is a type of blood cancer that is currently well-managed with drugs that target cancer-causing proteins. However, a significant proportion of CML patients do not respond to those drug treatments or relapse when they stop those drugs because the cancer cells in those patients stop relying on that protein and instead develop a new way to survive. Therefore, new treatment strategies may be necessary for those patients. In this review, we discuss those additional survival pathways and outline combination treatment strategies to increase responses and clinical outcomes, improving the lives of CML patients. Abstract Tyrosine kinase inhibitors (TKIs) have revolutionised the management of chronic myeloid leukaemia (CML), with the disease now having a five-year survival rate over 80%. The primary focus in the treatment of CML has been on improving the specificity and potency of TKIs to inhibit the activation of the BCR::ABL1 kinase and/or overcoming resistance driven by mutations in the BCR::ABL1 oncogene. However, this approach may be limited in a significant proportion of patients who develop TKI resistance despite the effective inhibition of BCR::ABL1. These patients may require novel therapeutic strategies that target both BCR::ABL1-dependent and BCR::ABL1-independent mechanisms of resistance. The combination treatment strategies that target alternative survival signalling, which may contribute towards BCR::ABL1-independent resistance, could be a successful strategy for eradicating residual leukaemic cells and consequently increasing the response rate in CML patients.
Collapse
Affiliation(s)
- Govinda Poudel
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
| | - Molly G. Tolland
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Timothy P. Hughes
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital and SA Pathology, Adelaide, SA 5000, Australia
| | - Ilaria S. Pagani
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
- Correspondence:
| |
Collapse
|
40
|
Wang K, Wei J, Ma J, Jia Q, Liu Y, Chai J, Xu J, Xu T, Zhao D, Wang Y, Yan Q, Guo S, Guo X, Zhu F, Fan L, Li M, Wang Z. Phosphorylation of PBK/TOPK Tyr74 by JAK2 promotes Burkitt lymphoma tumor growth. Cancer Lett 2022; 544:215812. [PMID: 35780928 DOI: 10.1016/j.canlet.2022.215812] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/08/2022] [Accepted: 06/27/2022] [Indexed: 11/02/2022]
Abstract
Burkitt lymphoma (BL), which is characterized by high invasiveness, is a subgroup of non-Hodgkin lymphoma. Although BL is regarded as a highly curable disease, especially for children, some patients unfortunately still do not respond adequately. The understanding of the etiology and molecular mechanisms of BL is still limited, and targeted therapies are still lacking. Here, we found that T-LAK cell-derived protein kinase (TOPK) and phosphorylated Janus kinase 2 (p-JAK2) are highly expressed in the tissues of BL patients. We report that TOPK directly binds to and is phosphorylated at Tyr74 by JAK2. Histone H3, one of the downstream targets of TOPK, is also phosphorylated in vivo and in vitro. Furthermore, we report that the phosphorylation of TOPK at Tyr74 by JAK2 plays a vital role in the proliferation of BL cells and promotes BL tumorigenesis in vivo. Phosphorylation of TOPK at Tyr74 by JAK2 enhances the stability of TOPK. Collectively, our results suggest that the JAK2/TOPK/histone H3 axis plays a key role in the proliferation of BL cells and BL tumorigenesis in vivo.
Collapse
Affiliation(s)
- Kaijing Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Jie Wei
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Jing Ma
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Qingge Jia
- Department of Reproductive Endocrinology, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China
| | - Yixiong Liu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Jia Chai
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Junpeng Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Danhui Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Qingguo Yan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Shuangping Guo
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Xinjian Guo
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining City, Qinghai Province, China
| | - Feng Zhu
- Cancer Research Institute, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Linni Fan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China.
| | - Zhe Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Air Force Medical University, Xi'an, China.
| |
Collapse
|
41
|
Fé LXSGM, Cipolatti EP, Pinto MCC, Branco S, Nogueira FCS, Ortiz GMD, Pinheiro ADS, Manoel EA. Enzymes in the time of COVID-19: An overview about the effects in the human body, enzyme market, and perspectives for new drugs. Med Res Rev 2022; 42:2126-2167. [PMID: 35762498 PMCID: PMC9350392 DOI: 10.1002/med.21919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 01/27/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022]
Abstract
The rising pandemic caused by a coronavirus, resulted in a scientific quest to discover some effective treatments against its etiologic agent, the severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2). This research represented a significant scientific landmark and resulted in many medical advances. However, efforts to understand the viral mechanism of action and how the human body machinery is subverted during the infection are still ongoing. Herein, we contributed to this field with this compilation of the roles of both viral and human enzymes in the context of SARS‐CoV‐2 infection. In this sense, this overview reports that proteases are vital for the infection to take place: from SARS‐CoV‐2 perspective, the main protease (Mpro) and papain‐like protease (PLpro) are highlighted; from the human body, angiotensin‐converting enzyme‐2, transmembrane serine protease‐2, and cathepsins (CatB/L) are pointed out. In addition, the influence of the virus on other enzymes is reported as the JAK/STAT pathway and the levels of lipase, enzymes from the cholesterol metabolism pathway, amylase, aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and glyceraldehyde 3‐phosphate dehydrogenase are also be disturbed in SARS‐CoV‐2 infection. Finally, this paper discusses the importance of detailed enzymatic studies for future treatments against SARS‐CoV‐2, and how some issues related to the syndrome treatment can create opportunities in the biotechnological market of enzymes and the development of new drugs.
Collapse
Affiliation(s)
- Luana Xavier Soares Gomes Moura Fé
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliane Pereira Cipolatti
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Engenharia Química, Instituto de Tecnologia, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Rio de Janeiro, Brazil
| | - Martina Costa Cerqueira Pinto
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil.,Chemical Engineering Program, Instituto Alberto Luiz Coimbra de Pós-graduação e Pesquisa de Engenharia (COPPE), Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suema Branco
- Biofísica Ambiental, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio César Sousa Nogueira
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisela Maria Dellamora Ortiz
- Departamento de Fármacos e Medicamentos, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anderson de Sá Pinheiro
- Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelin Andrade Manoel
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Centro de Ciências da Saúde (CCS), Universidade Federal do Rio de Janeiro (UFRJ)-Cidade Universitária, Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Bioquímica, Instituto de Química, Centro de Tecnologia (CT), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Li B, Yang L, Bai F, Tong B, Liu X. Indications and effects of biological agents in the treatment of noninfectious uveitis. Immunotherapy 2022; 14:985-994. [PMID: 35695019 DOI: 10.2217/imt-2021-0303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Noninfectious uveitis is a common blinding eye disease, and an autoimmune response is involved in its pathogenesis. Biological agents have gradually been introduced into the treatment of noninfectious uveitis. The authors reviewed the clinical application and side effects of different biological agents on noninfectious uveitis. Biological agents that target TNF-α are widely used in the clinic. Other biological agents, such as IL-6- and IL-1-neutralizing antibodies, are used in patients who do not respond to TNF inhibitors. The efficacy of IL-17 neutralizing antibodies in noninfectious uveitis is controversial. Biological agents targeting T cells and signaling pathways provide new drug options for treatment of noninfectious uveitis. However, it cannot be ignored that these biological agents have side effects, such as increasing risk of infection.
Collapse
Affiliation(s)
- Bowen Li
- Ophthalmologic Center of the Second Hospital, Jilin University, Ziqiang Street 218, Changchun, 130000, P.R. China.,Clinical College, Jilin University, Xinmin Street 70, Changchun, 130000, P.R. China
| | - Li Yang
- Ophthalmologic Center of the Second Hospital, Jilin University, Ziqiang Street 218, Changchun, 130000, P.R. China
| | - Feng Bai
- Ophthalmologic Center of the Second Hospital, Jilin University, Ziqiang Street 218, Changchun, 130000, P.R. China
| | - Bainan Tong
- Ophthalmologic Center of the Second Hospital, Jilin University, Ziqiang Street 218, Changchun, 130000, P.R. China
| | - Xiaoli Liu
- Ophthalmologic Center of the Second Hospital, Jilin University, Ziqiang Street 218, Changchun, 130000, P.R. China
| |
Collapse
|
43
|
Nooreen R, Nene S, Jain H, Prasannanjaneyulu V, Chitlangya P, Otavi S, Khatri DK, Raghuvanshi RS, Singh SB, Srivastava S. Polymer nanotherapeutics: A versatile platform for effective rheumatoid arthritis therapy. J Control Release 2022; 348:397-419. [PMID: 35660632 DOI: 10.1016/j.jconrel.2022.05.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 11/27/2022]
Abstract
Rheumatoid arthritis is an aggressive and severely debilitating disorder that is characterized by joint pain and cartilage damage. It restricts mobility in patients, leaving them unable to carry out simple tasks. RA presents itself with severe lasting pain, swelling and stiffness in the joints and may cause permanent disability in patients. Treatment regimens currently employed for rheumatoid arthritis revolve around keeping clinical symptoms like joint pain, inflammation, swelling and stiffness at bay. The current therapeutic interventions in rheumatoid arthritis involve the use of non-steroidal anti-inflammatory drugs, glucocorticoids, disease-modifying anti-rheumatic drugs and newer biological drugs that are engineered for inhibiting the expression of pro-inflammatory mediators. These conventional drugs are plagued with severe adverse effects because of their higher systemic distribution, lack of specificity and higher doses. Oral, intra-articular, and intravenous routes are routinely used for drug delivery which is associated with decreased patient compliance, high cost, poor bioavailability and rapid systemic clearance. All these drawbacks have enticed researchers to create novel strategies for drug delivery, the main approach being nanocarrier-based systems. In this article, we aim to consolidate the remarkable contributions of polymeric carrier systems including microneedle technology and smart trigger-responsive polymeric carriers in the management of rheumatoid arthritis along with its detailed pathophysiology. This review also briefly describes the safety and regulatory aspects of polymer therapeutics.
Collapse
Affiliation(s)
- Rimsha Nooreen
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shweta Nene
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Harsha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Velpula Prasannanjaneyulu
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Palak Chitlangya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shivam Otavi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, India
| | - Shashi Bala Singh
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
44
|
Wang Z, Liu W, Li X, Chen H, Qi D, Pan F, Liu H, Yu S, Yi B, Wang G, Liu Y. Physiologically based pharmacokinetic combined JAK2 occupancy modelling to simulate PK and PD of baricitinib with kidney transporter inhibitors and in patients with hepatic/renal impairment. Regul Toxicol Pharmacol 2022; 133:105210. [PMID: 35700864 DOI: 10.1016/j.yrtph.2022.105210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/05/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
PURPOSE Our aim is to build a physiologically based pharmacokinetic and JAK2 occupancy model (PBPK-JO) to simultaneously predict pharmacokinetic (PK) and pharmacodynamic (PD) changes of baricitinib (BAR) in healthy humans when co-administrated with kidney transporters OAT3 and MATE2-K inhibitors, and in patients with hepatic and renal impairment. METHODS Probenecid and vandetanib were selected as OAT3 and MATE2-K competitive inhibitors, respectively. The PBPK-JO model was built using physicochemical and biochemical properties of BAR, and then verified by observed clinical PK. Finally, the model was applied to determine optimal dosing regimens in various clinical situations. RESULTS Here, we have successfully simulated PK and JAK2 occupancy profiles in humans by PBPK-JO model. Moreover, this modelling reproduced every observed PK data, and every mean relative deviation (MRD) was below 2. The simulation suggested that PK of BAR had a significant change (2.22-fold increase), however PD only had a slight increase of 1.14-fold. Additionally, the simulation also suggested that vandetanib was almost unlikely to affect the PK and PD of BAR. In simulations of hepatic and renal impairment patients, the predictions suggested that significant changes in the PK and PD of BAR occurred. However, there was a lower fold increase in JAK2 occupancy than in PK in patients relative to healthy individuals. CONCLUSION Administration dose adjustment of BAR when co-administrated with OAT3 inhibitors or in patients with hepatic or renal impairment should combine PK and PD changes of BAR, instead of only considering PK alteration.
Collapse
Affiliation(s)
- Zhongjian Wang
- Pharnexcloud Digital Technology Co., Ltd., Chengdu, Sichuan, 610093, China
| | - Wei Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Xueyan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Hongjiao Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Dongying Qi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Fulu Pan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Huining Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Shuang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Bowen Yi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Guopeng Wang
- Zhongcai Health (Beijing) Biological Technology Development Co., Ltd., Beijing, 101500, China.
| | - Yang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100102, China.
| |
Collapse
|
45
|
Srivastava S, Rasool M. Underpinning IL-6 biology and emphasizing selective JAK blockade as the potential alternate therapeutic intervention for rheumatoid arthritis. Life Sci 2022; 298:120516. [DOI: 10.1016/j.lfs.2022.120516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/08/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023]
|
46
|
Park JW, Kim JH, Kim SE, Jung JH, Jang MK, Park SH, Lee MS, Kim HS, Suk KT, Kim DJ. Primary Biliary Cholangitis and Primary Sclerosing Cholangitis: Current Knowledge of Pathogenesis and Therapeutics. Biomedicines 2022; 10:biomedicines10061288. [PMID: 35740310 PMCID: PMC9220082 DOI: 10.3390/biomedicines10061288] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/24/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023] Open
Abstract
Cholangiopathies encompass various biliary diseases affecting the biliary epithelium, resulting in cholestasis, inflammation, fibrosis, and ultimately liver cirrhosis. Primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) are the most important progressive cholangiopathies in adults. Much research has broadened the scope of disease biology to genetic risk, epigenetic changes, dysregulated mucosal immunity, altered biliary epithelial cell function, and dysbiosis, all of which interact and arise in the context of ill-defined environmental triggers. An in-depth understanding of the molecular pathogenesis of these cholestatic diseases will help clinicians better prevent and treat diseases. In this review, we focus on the main underlying mechanisms of disease initiation and progression, and novel targeted therapeutics beyond currently approved treatments.
Collapse
Affiliation(s)
- Ji-Won Park
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Jung-Hee Kim
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Sung-Eun Kim
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Jang Han Jung
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Myoung-Kuk Jang
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Sang-Hoon Park
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
| | - Myung-Seok Lee
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
| | - Hyoung-Su Kim
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Ki Tae Suk
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
| | - Dong Joon Kim
- Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon-si 24252, Korea; (J.-W.P.); (J.-H.K.); (S.-E.K.); (J.H.J.); (M.-K.J.); (S.-H.P.); (M.-S.L.); (H.-S.K.); (K.T.S.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 200-010, Korea
- Correspondence: ; Tel.: +82-33-240-5646
| |
Collapse
|
47
|
Lee J, Bae Y, Kim NJ, Lim S, Kim YM, Kim J, Chin YW. Anti-rheumatic, and analgesic effects by the parent tuberous roots of Aconitum jaluense in adjuvant induced arthritis rats. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:114518. [PMID: 34637968 DOI: 10.1016/j.jep.2021.114518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
AIM OF THE STUDY The aim of this study was to test the anti-rheumatic effects of A. jaluense tubers in acute and chronic arthritis rats, and to assign its ingredients through UHPLC-TOF/MS. MATERIALS AND METHODS Subcutaneous injection of carrageenan for acute arthritis and complete Freund's adjuvant (CFA) for chronic arthritis was carried out in the hind paw of SD rats. The paw volume was measured by a plethysmometer thermal hyperalgesia was tested using a thermal plantar tester, and mechanical hyperalgesia was evaluated by ankle flexion evoked vocalizations. The expression of c-Fos in the brain hippocampus was measured with the avidin-biotin-peroxidase technique. The ingredients were assigned by UHPLC-TOF/MS, chromatography was performed by UHPLC system with DAD detector and BEH C18 column, and spectroscopy was conducted by ESI-MS system. RESULTS AND DISCUSSION The 80% ethanoic extract of A. jaluense tubers showed an acute anti-inflammatory effect by suppressing the edema volume in the hind paw of carrageenan-stimulated rats. In addition, A. jaluense tubers exerted an anti-rheumatic activity by reducing the secondary swelling volume from an immunological reaction in the left hind paw of CFA-induced chronic arthritis rats. Additionally, oral treatment with the 80% ethanoic extract -showed potent analgesic effects in the arthritis rats by recovering the paw withdrawal latency stimulated by the thermal hyperalgesia and by reducing the vocalization scores evoked by ankle flexion on both hind paws. Moreover, its treatment also indicated an anti-psychiatric effect by controlling the c-Fos protein expression of the brain hippocampus in CFA-stimulated arthritis rats. These results suggested that these therapeutic effects were exhibited by less toxic mono-esterified diterpenoid alkaloids (MDAs), and nontoxic non-esterified diterpenoid alkaloids (NDAs). CONCLUSION A. jaluense tubers may act as viable therapeutic or preventive candidates for acute and chronic arthritis, particularly, for immune-inflammatory rheumatoid arthritis to suppress the pain and psychiatric condition.
Collapse
Affiliation(s)
- JiSuk Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, South Korea.
| | - YoungChul Bae
- Research Group of Pain and Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, 02447, South Korea
| | - Nam Jae Kim
- Research Group of Pain and Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, 02447, South Korea
| | - Sabina Lim
- Research Group of Pain and Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, 02447, South Korea.
| | - Young-Mi Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jinwoong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, South Korea.
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
48
|
Saha A, Hyzy S, Lamothe T, Hammond K, Clark N, Lanieri L, Bhattarai P, Palchaudhuri R, Gillard GO, Proctor J, Riddle MJ, Panoskaltsis-Mortari A, MacMillan ML, Wagner JE, Kiem HP, Olson LM, Blazar BR. A CD45-targeted antibody-drug conjugate successfully conditions for allogeneic hematopoietic stem cell transplantation in mice. Blood 2022; 139:1743-1759. [PMID: 34986233 PMCID: PMC8931510 DOI: 10.1182/blood.2021012366] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment of patients with nonmalignant or malignant blood disorders. Its success has been limited by graft-versus-host disease (GVHD). Current systemic nontargeted conditioning regimens mediate tissue injury and potentially incite and amplify GVHD, limiting the use of this potentially curative treatment beyond malignant disorders. Minimizing systemic nontargeted conditioning while achieving alloengraftment without global immune suppression is highly desirable. Antibody-drug-conjugates (ADCs) targeting hematopoietic cells can specifically deplete host stem and immune cells and enable alloengraftment. We report an anti-mouse CD45-targeted-ADC (CD45-ADC) that facilitates stable murine multilineage donor cell engraftment. Conditioning with CD45-ADC (3 mg/kg) was effective as a single agent in both congenic and minor-mismatch transplant models resulting in full donor chimerism comparable to lethal total body irradiation (TBI). In an MHC-disparate allo-HSCT model, pretransplant CD45-ADC (3 mg/kg) combined with low-dose TBI (150 cGy) and a short course of costimulatory blockade with anti-CD40 ligand antibody enabled 89% of recipients to achieve stable alloengraftment (mean value: 72%). When CD45-ADC was combined with pretransplant TBI (50 cGy) and posttransplant rapamycin, cyclophosphamide (Cytoxan), or a JAK inhibitor, 90% to 100% of recipients achieved stable chimerism (mean: 77%, 59%, 78%, respectively). At a higher dose (5 mg/kg), CD45-ADC as a single agent was sufficient for rapid, high-level multilineage chimerism sustained through the 22 weeks observation period. Therefore, CD45-ADC has the potential utility to confer the benefit of fully myeloablative conditioning but with substantially reduced toxicity when given as a single agent or at lower doses in conjunction with reduced-intensity conditioning.
Collapse
Affiliation(s)
- Asim Saha
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | | | | | | | | | | | | | | | | | - Megan J Riddle
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Angela Panoskaltsis-Mortari
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Margaret L MacMillan
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - John E Wagner
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Hans-Peter Kiem
- Fred Hutchinson Cancer Research Center and Department of Medicine, University of Washington, Seattle, WA
| | | | - Bruce R Blazar
- Division of Blood & Marrow Transplant & Cellular Therapy, Masonic Cancer Center and Department of Pediatrics, University of Minnesota, Minneapolis, MN
| |
Collapse
|
49
|
Maitz T, Parfianowicz D, Vojtek A, Rajeswaran Y, Vyas AV, Gupta R. COVID-19 Cardiovascular Connection: A Review of Cardiac Manifestations in COVID-19 Infection and Treatment Modalities. Curr Probl Cardiol 2022:101186. [PMID: 35351486 PMCID: PMC8957382 DOI: 10.1016/j.cpcardiol.2022.101186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 01/08/2023]
Abstract
The coronavirus pandemic has crippled healthcare system since its outbreak in 2020, and has led to over 2.6 million deaths worldwide. Clinical manifestations of COVID-19 range from asymptomatic carrier to severe pneumonia, to life-threatening acute respiratory distress syndrome (ARDS). The early efforts of the pandemic surrounded treating the pulmonary component of COVID-19, however, there has been robust data surrounding the cardiac complications associated with the virus. This is suspected to be from a marked inflammatory response as well as direct viral injury. Arrhythmias, acute myocardial injury, myocarditis, cardiomyopathy, thrombosis, and myocardial fibrosis are some of the observed cardiac complications. There have been high morbidity and mortality rates in those affected by cardiac conditions associated with COVID-19. Additionally, there have been documented cases of patients presenting with typical cardiac symptoms who are subsequently discovered to have COVID-19 infection. In those who test positive for COVID-19, clinical awareness of the significant cardiac components of the virus is pertinent to prevent morbidity and mortality. Unfortunately, treatment and preventative measures developed for COVID-19 have been shown to be also be associated with cardiac complications. This is a comprehensive review of the cardiac complications and manifestations of COVID-19 infection in addition to those associated with both treatment and vaccination.
Collapse
Affiliation(s)
- Theresa Maitz
- Department of Medicine, Lehigh Valley Health Network, Allentown, PA
| | | | - Ashley Vojtek
- Department of Medicine, Lehigh Valley Health Network, Allentown, PA
| | | | - Apurva V Vyas
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA
| | - Rahul Gupta
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA.
| |
Collapse
|
50
|
Yang T, Cui X, Tang M, Qi W, Zhu Z, Shi M, Yang L, Pei H, Zhang W, Xie L, Xu Y, Yang Z, Chen L. Identification of a Novel 2,8-Diazaspiro[4.5]decan-1-one Derivative as a Potent and Selective Dual TYK2/JAK1 Inhibitor for the Treatment of Inflammatory Bowel Disease. J Med Chem 2022; 65:3151-3172. [PMID: 35113547 DOI: 10.1021/acs.jmedchem.1c01137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this study, we described a series of 2,8-diazaspiro[4.5]decan-1-one derivatives as selective TYK2/JAK1 inhibitors. Systematic exploration of the structure-activity relationship through the introduction of spirocyclic scaffolds based on the reported selective TYK2 inhibitor 14l led to the discovery of the superior derivative compound 48. Compound 48 showed excellent potency on TYK2/JAK1 kinases with IC50 values of 6 and 37 nM, respectively, and exhibited more than 23-fold selectivity for JAK2. Compound 48 also demonstrated excellent metabolic stability and more potent anti-inflammatory efficacy than tofacitinib in acute ulcerative colitis models. Moreover, the excellent anti-inflammatory effect of compound 48 was mediated by regulating the expression of related TYK2/JAK1-regulated genes, as well as the formation of Th1, Th2, and Th17 cells. Taken together, these findings suggest that compound 48 is a selective dual TYK2/JAK inhibitor, deserving to be developed as a clinical candidate.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xue Cui
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Wenyan Qi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Heying Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Wanhua Zhang
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lixin Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yaohui Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.,Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu 610041, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.,Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu 610041, China
| |
Collapse
|