1
|
Mok TC, Mok CC. The Potential Use of Arsenic Trioxide in the Treatment of Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:9577. [PMID: 39273522 PMCID: PMC11394723 DOI: 10.3390/ijms25179577] [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: 08/11/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Arsenic trioxide (ATO) is now part of the standard regimen for the treatment of newly diagnosed and relapsed acute promyelocytic leukemia. The availability of an oral form of ATO has greatly reduced the incidence of cardiotoxicity as compared to intravenous (IV) administration. Increasing evidence suggests that ATO has anti-inflammatory properties that may be useful for the treatment of autoimmune diseases. These include the modulation of Treg cell activation, Th1/Th2 and Th17/Treg balance, depletion of activated T cells and plasmacytoid dendritic cells, and influence of B-cell differentiation, leading to reduced autoantibody and cytokine production. ATO has also been shown to induce apoptosis of activated fibroblast-like synoviocytes through the generation of reactive oxygen species and alter the gut microbiota in collagen-induced arthritis. Despite the emergence of newer treatment modalities, the treatment of systemic lupus erythematosus (SLE), especially refractory manifestations, remains a challenge, owing to the paucity of effective biological and targeted therapies that are devoid of adverse effects. Oral ATO is an attractive option for the treatment of SLE because of the lower cost of production, convenience of administration, and reduced cardiotoxicity. This article summarizes the anti-inflammatory mechanisms of ATO and its potential application in the treatment of SLE and other rheumatic diseases.
Collapse
Affiliation(s)
- Tsz Ching Mok
- Department of Medicine, Ruttonjee Hospital, Hong Kong SAR, China
| | - Chi Chiu Mok
- Department of Medicine and Geriatrics, Tuen Mun Hospital, Hong Kong SAR, China
| |
Collapse
|
2
|
Kwon S, Judson MA. Clinical Pharmacology in Sarcoidosis: How to Use and Monitor Sarcoidosis Medications. J Clin Med 2024; 13:1250. [PMID: 38592130 PMCID: PMC10932410 DOI: 10.3390/jcm13051250] [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: 01/15/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 04/10/2024] Open
Abstract
When sarcoidosis needs treatment, pharmacotherapy is usually required. Although glucocorticoids work reliably and relatively quickly for sarcoidosis, these drugs are associated with numerous significant side effects. Such side effects are common in sarcoidosis patients, as the disease frequently has a chronic course and glucocorticoid treatment courses are often prolonged. For these reasons, corticosteroid-sparing and corticosteroid-replacing therapies are often required for sarcoidosis. Unfortunately, many healthcare providers who care for sarcoidosis patients are not familiar with the use of these agents. In this manuscript, we provide a review of the pharmacotherapy of sarcoidosis. We discuss the mechanism of action, dosing, side-effect profile, approach to monitoring and patient counselling concerning glucocorticoids, and the common alternative drugs recommended for use in the recent European Respiratory Society (Lausanne, Switzerland) Sarcoidosis Treatment Guidelines. We also discuss the use of these agents in special situations including hepatic insufficiency, renal insufficiency, pregnancy, breastfeeding, vaccination, and drug-drug interactions. It is hoped that this manuscript will provide valuable practical guidance to clinicians who care for sarcoidosis patients.
Collapse
Affiliation(s)
- Sooyeon Kwon
- Samuel S. Stratton Veterans Affairs Medical Center, Albany, NY 12208, USA
| | - Marc A. Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY 12208, USA;
| |
Collapse
|
3
|
Merrill JT, Guthridge J, Smith M, June J, Koumpouras F, Machua W, Askanase A, Khosroshahi A, Sheikh SZ, Rathi G, Burington B, Foster P, Matijevic M, Arora S, Wang X, Gao M, Wax S, James JA, Zack DJ. Obexelimab in Systemic Lupus Erythematosus With Exploration of Response Based on Gene Pathway Co-Expression Patterns: A Double-Blind, Randomized, Placebo-Controlled, Phase 2 Trial. Arthritis Rheumatol 2023; 75:2185-2194. [PMID: 37459248 DOI: 10.1002/art.42652] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/17/2023] [Accepted: 07/11/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVE Obexelimab is an investigational, bifunctional, noncytolytic monoclonal antibody that binds CD19 and FcyRIIb to inhibit B cells, plasmablasts, and plasma cells. This trial evaluated the efficacy and safety of obexelimab in the treatment of patients with systemic lupus erythematosus (SLE). METHODS During screening, patients with active, non-organ-threatening SLE received corticosteroid injections to ameliorate symptoms while immunosuppressants were withdrawn (≤10 mg/day prednisone equivalent and ≤400 mg/day hydroxychloroquine allowed). Patients with improved disease activity were randomized 1:1 to obexelimab 5 mg/kg intravenously or placebo once every 2 weeks until week 32 or loss of improvement (LOI). RESULTS In this study, 104 patients were randomized. Analysis of the primary endpoint, proportion of patients reaching week 32 without LOI, used an efficacy-evaluable (EE) population defined as patients who completed the study or withdrew for flare or treatment-related toxicity. This endpoint did not reach statistical significance: 21 of 50 obexelimab-treated patients (42.0%) versus 12 of 42 patients (28.6%) treated with a placebo (P = 0.183). Time to LOI was increased in obexelimab-treated patients versus patients treated with a placebo in the EE (hazard ratio [HR] 0.53, P = 0.025) and intention-to-treat (HR 0.59, P = 0.062) populations. In obexelimab-treated patients, B cells decreased approximately 50%, and trough concentration and inclusion in baseline gene expression clusters with high B cell pathway modules were associated with increased time to LOI. Obexelimab was associated with infusion reactions but was generally safe and well-tolerated. CONCLUSION Although the primary endpoint was not reached, secondary analysis showed time to LOI was significantly increased in obexelimab-treated patients, and analysis of patient subsets defined by gene expression patterns at baseline suggests a responding subpopulation.
Collapse
Affiliation(s)
- Joan T Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Joel Guthridge
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Miles Smith
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Joshua June
- Great Lakes Center of Rheumatology, Lansing, Michigan
| | | | | | - Anca Askanase
- Columbia University Medical Center, New York City, New York
| | | | - Saira Z Sheikh
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | | - Judith A James
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | | |
Collapse
|
4
|
Huang X, Shu Q, Luo X, Ge W, Xie H, Zhou Y. Analysis of Factors Influencing Whole Blood Hydroxychloroquine Concentration in Patients with Systemic Lupus Erythematosus in China. Rheumatol Ther 2023; 10:1597-1607. [PMID: 37755649 PMCID: PMC10654291 DOI: 10.1007/s40744-023-00598-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023] Open
Abstract
INTRODUCTION The aim of this study was to determine the factors associated with the concentrations of hydroxychloroquine (HCQ) and its major metabolite, desethylhydroxychloroquine (DHCQ), in patients with systemic lupus erythematosus (SLE). METHODS Patients with SLE taking oral HCQ for at least 3 months were recruited from the Department of Rheumatology and Immunology of Nanjing Drum Tower Hospital. Clinical characteristics and laboratory values were examined. The concentrations of HCQ and DHCQ were measured by high-performance liquid chromatography, and the effects of various factors on the concentrations were investigated. RESULTS A total of 272 patients were included in this study. The average concentration of HCQ was 690.90 ng/ml and the average concentration of DHCQ was 431.84 ng/ml. Multivariate analysis indicated that gender (P = 0.015), age (year) (P < 0.001), weight (kg) (P = 0.013), duration of HCQ use (month) (P < 0.001), systemic lupus erythematosus disease activity index (SLEDAI) (P < 0.001), platelet count (× 109/l) (P < 0.001), immunoglobulin G levels (g/l) (P = 0.014) were associated with low HCQ concentrations. Gender (P = 0.006), duration of HCQ use (month) (P < 0.001), SLEDAI (P = 0.007), and platelet count (× 109/l) (P < 0.001) were associated with low DHCQ concentrations. CONCLUSIONS Patients with SLE require long-term administration of HCQ, but blood levels vary widely between individuals. Studying the factors influencing the blood HCQ and DHCQ concentrations and optimizing the dose according to individual characteristics might help to improve the efficacy of HCQ. TRIAL REGISTRATION ChiCTR2300070628.
Collapse
Affiliation(s)
- Xuan Huang
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
| | - Qing Shu
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu Province, China
| | - Xuemei Luo
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu Province, China
| | - Weihong Ge
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu Province, China.
| | - Han Xie
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu Province, China.
| | - Yujie Zhou
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
5
|
Cheng DH, Huang ZG. Therapeutic drug monitoring of disease-modifying antirheumatic drugs in circulating leukocytes in immune-mediated inflammatory diseases. Inflammopharmacology 2023:10.1007/s10787-023-01243-8. [PMID: 37160525 DOI: 10.1007/s10787-023-01243-8] [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: 01/25/2023] [Accepted: 04/07/2023] [Indexed: 05/11/2023]
Abstract
The treatment of immune-mediated inflammatory diseases (IMIDs) is one of the main challenges of modern medicine. Although a number of disease-modifying antirheumatic drugs (DMARDs) are available, there is wide variability in clinical response to treatment among individuals. Therapeutic drug monitoring (TDM) has been proposed to optimize treatment; however, some patients still experience unsatisfactory outcomes, although the blood concentrations of drugs in these patients remain in the therapeutic range. One possible reason for this is that the conventional samples (e.g., whole blood or plasma) used in TDM may not accurately reflect drug concentrations or concentrations of their metabolites at the target site. Hence, more refined TDM approaches to guide clinical decisions related to dose optimization are necessary. Circulating leukocytes or white blood cells have a critical role in driving the inflammatory process. They are recruited to the site of injury, infection and inflammation, and the main target of small molecule DMARDs is within immune cells. Given this, assaying drug concentrations in leukocytes has been proposed to be of possible relevance to the interpretation of outcomes. This review focuses on the clinical implications and challenges of drug monitoring of DMARDs in peripheral blood leukocytes from therapeutic or toxicological perspectives in IMIDs.
Collapse
Affiliation(s)
- Dao-Hai Cheng
- Department of Pharmacy, First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Zhen-Guang Huang
- Department of Pharmacy, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Zeng L, Yang T, Yang K, Yu G, Li J, Xiang W, Chen H. Curcumin and Curcuma longa Extract in the Treatment of 10 Types of Autoimmune Diseases: A Systematic Review and Meta-Analysis of 31 Randomized Controlled Trials. Front Immunol 2022; 13:896476. [PMID: 35979355 PMCID: PMC9376628 DOI: 10.3389/fimmu.2022.896476] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/04/2022] [Indexed: 01/30/2023] Open
Abstract
Objective To evaluate the randomized controlled trials (RCTs) of Curcumin and Curcuma longa Extract in the treatment of autoimmune diseases. Methods Databases such as Embase, Web of Science, PubMed and The Cochrane Library were searched from the database establishment to February 2022 to collect RCTs of Curcumin and Curcuma longa Extract in the treatment of autoimmune diseases. Then the literature was screened and the data were extracted. Meta-analysis was performed using RevMan 5.3 software. Results A total of 34 records were included, involving 31 RCTs and 10 types of autoimmune disease. Among them, ankylosing spondylitis (AS) involves one RCT, Behcet ‘s disease (BD) involves one RCT, Crohn ‘s disease involves two RCTs, multiple sclerosis (MS) involves two RCTs, oral lichen planus involves six RCTs, psoriasis involves two RCTs, rheumatoid arthritis (RA) involves five RCTs, systemic lupus erythematosus (SLE) involves two RCTs, arteritis involves one RCT, ulcerative colitis (UC) involves nine RCTs. Among them, most of the RCTs of ulcerative colitis (UC), oral lichen planus, RA showed that curcumin and curcumin extracts improved clinical or laboratory results. Crohn ‘ s disease, MS, SLE, psoriasis included two RCTs; they all showed improvements (at least one RCT reported improvements in clinical outcomes). AS, BD and arteritis included only one RCT, and the clinical results showed improvement. However, due to the small number of RCTs and the small number of patients involved in each disease, there is still a need for more high-quality RCTs. Conclusion Curcumin and Curcuma longa Extract had good clinical efficacy in the treatment of Psoriasis, UC and RA, so Curcumin and Curcuma longa Extract could be used in the treatment of the above diseases in the future. The results of Meta-analysis showed that Curcumin and Curcuma longa Extract did not show efficacy in the treatment of oral lichen planus, while Takayasu arteritis, SLE, MS, AS, BD and CD did not report sufficient clinical data for meta-analysis. Therefore, large-sample, multi-center clinical trials are still needed for revision or validation.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| | - Tiejun Yang
- Department of Orthopedics, People’s Hospital of Ningxiang City, Ningxiang, Hunan, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| | - Ganpeng Yu
- Department of Orthopedics, People’s Hospital of Ningxiang City, Ningxiang, Hunan, China
| | - Jun Li
- Department of Orthopedics, People’s Hospital of Ningxiang City, Ningxiang, Hunan, China
| | - Wang Xiang
- Department of Rheumatology, The First people’s Hospital Changde City, Changde, Hunan, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| |
Collapse
|
7
|
Gelder T, Lerma E, Engelke K, Huizinga RB. Voclosporin: a novel calcineurin inhibitor for the treatment of lupus nephritis. Expert Rev Clin Pharmacol 2022; 15:515-529. [PMID: 35763288 DOI: 10.1080/17512433.2022.2092470] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Lupus nephritis (LN) is a severe manifestation of systemic lupus erythematosus. Standard-of-care immunosuppressive therapies achieve poor complete renal response (CRR) rates, with considerable toxicity. This article reviews voclosporin, a novel oral calcineurin inhibitor (CNI) approved for treatment of adults with active LN by the US Food and Drug Administration (the FDA) in January 2021. AREAS COVERED : This review summarizes the chemical properties, pharmacokinetics, and pharmacodynamics of voclosporin, and its efficacy and safety in LN, based on literature review covering PubMed searches, manufacturers' websites and documents produced by the FDA. EXPERT OPINION : Voclosporin is a CNI with a consistent pharmacokinetic-pharmacodynamic relationship resulting from enhanced calcineurin binding and reduced drug and metabolite load. This profile permits therapeutic efficacy in LN at a dose associated with relatively low calcineurin inhibition, and therefore a potentially improved safety profile. Pivotal trials demonstrated a significant benefit of adding voclosporin to standard therapy, with rapid reduction in proteinuria, and a clinically meaningful and significantly higher CRR rate at 1 year. At approved doses for LN, potential advantages of voclosporin versus historical experience with CNIs include lack of need for therapeutic drug monitoring, benign metabolic, lipid and electrolyte profile, and no impact on mycophenolate mofetil levels.
Collapse
Affiliation(s)
- Teun Gelder
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Edgar Lerma
- Section of Nephrology, University of Illinois at Chicago College of Medicine/Advocate Christ Medical Center, Chicago, IL, USA
| | | | | |
Collapse
|
8
|
Arora S, Rovin BH. Expert Perspective: An Approach to Refractory Lupus Nephritis. Arthritis Rheumatol 2022; 74:915-926. [PMID: 35166048 PMCID: PMC9156543 DOI: 10.1002/art.42092] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/24/2022] [Accepted: 02/09/2022] [Indexed: 11/06/2022]
Abstract
Systemic lupus erythematosus affects the kidneys in ~50% of all patients, and lupus nephritis (LN) is the most common manifestation of kidney involvement. Despite prompt diagnosis and treatment with aggressive immunosuppression, a significant proportion of LN patients do not respond to treatment and are considered to have refractory LN. Several factors other than drug resistance, such as nonadherence to treatment, undertreatment with conventional drugs, the effects of accumulated chronic damage, and genetic factors, may contribute to a poor response to treatment and should be considered. We define refractory LN as no change in (or worsening of) proteinuria and/or estimated glomerular filtration rate in response to 2 different standard-of-care induction regimens after 4-6 months in patients who are adherent to treatment. For patients who have LN that is truly refractory to standard of care, B cell-targeted therapy, specifically rituximab (RTX), is the most common next step. There is limited evidence available on alternative rescue therapies that may be used when there is no response to RTX. These include anti-CD38, leflunomide, intravenous immunoglobulin, plasma exchange, autologous stem cell transplantation, chimeric antigen receptor T cell therapy, anticomplement therapy, and interleukin-2 therapy.
Collapse
Affiliation(s)
| | - Brad H. Rovin
- The Ohio State University Wexner Medical Center, Columbus OH
| |
Collapse
|
9
|
Djabarouti S, Mora P, Lahouati M, Gigan M, d’Houdain N, Sourisseau B, Chambord J, Xuereb F. Intérêt des dosages pharmacologiques d’immunosuppresseurs et immunomodulateurs dans la prise en charge des maladies autoimmunes. Rev Med Interne 2022; 43:412-418. [DOI: 10.1016/j.revmed.2022.03.343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
10
|
Beaulieu Q, Zhang D, Melki I, Baudouin V, Goldwirst L, Woillard JB, Jacqz-Aigrain E. Pharmacokinetics of mycophenolic acid and external evaluation of two limited sampling strategies of drug exposure in patients with juvenile systematic lupus erythematosus. Eur J Clin Pharmacol 2022; 78:1003-1010. [PMID: 35294622 DOI: 10.1007/s00228-022-03295-1] [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: 11/23/2021] [Accepted: 02/14/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Mycophenolate mofetil (MMF), a pro-drug of mycophenolic acid (MPA), has become a major therapeutic option in juvenile systemic lupus erythematosus (jSLE). Monitoring MPA exposure using area under curve (AUC) has proved its value to increase efficacy and safety in solid organ transplantation both in children and adults, but additional data are required in patients with autoimmune diseases. In order to facilitate MMF therapeutic drug monitoring (TDM) in children, Bayesian estimators (BE) of MPA AUC0-12 h using limited sampling strategies (LSS) have been developed. Our aim was to conduct an external validation of these LSS using rich pharmacokinetics and compare their predictive performance. METHODS Pharmacokinetic blood samples were collected from jSLE treated by MMF and MPA plasma concentrations were determined using high-performance liquid chromatography system with ultraviolet detection (HPLC-UV). Individual AUC0-12 h at steady state was calculated using the trapezoid rule and compared with two LSS: (1) ISBA, a two-stage Bayesian approach developed for jSLE and (2) ADAPT, a non-linear mixed effects model with a parametric maximum likelihood approach developed with data from renal transplanted adults. RESULTS We received 41 rich pediatric PK at steady state from jSLE and calculated individual AUC0-12 h. The external validation MPA AUC0-12 h was conducted by selecting the concentration-time points adapted to ISBA and ADAPT: (1) ISBA showed good accuracy (bias: - 0.8 mg h/L), (2) ADAPT resulted in a bias of 6.7 mg L/h. The corresponding relative root mean square prediction error (RSME) was 23% and 43% respectively. CONCLUSION According to our external validation of two LSS of drug exposure, the ISBA model is recommended for Bayesian estimation of MPA AUC0-12 h in jSLE. In the literature focusing on MMF TDM, an efficacy cut-off for MPA AUC0-12 h between 30 and 45 mg h/L is proposed in jSLE but this requires additional validation.
Collapse
Affiliation(s)
- Quentin Beaulieu
- Paediatric Pharmacology, Department of Biological Pharmacology, Saint-Louis University Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Daolun Zhang
- Paediatric Pharmacology, Department of Biological Pharmacology, Saint-Louis University Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Isabelle Melki
- General Pediatrics, Infectious Disease and Internal Medicine Department, Robert Debre University Hospital, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), AP-HP, Paris, France.,Pediatric Hematology-Immunology and Rheumatology Department, Necker-Enfants Malades University Hospital, Reference center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), AP-HP, Paris, France.,Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris, France
| | - Véronique Baudouin
- Department of Pediatric Nephrology, Robert Debré University Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Lauriane Goldwirst
- Paediatric Pharmacology, Department of Biological Pharmacology, Saint-Louis University Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-Baptiste Woillard
- IPPRITT, INSERM, U1248, Limoges, France.,IPPRITT, University of Limoges, Limoges, France.,Department of Pharmacology and Toxicology, CHU Limoges, Limoges, France
| | - Evelyne Jacqz-Aigrain
- Paediatric Pharmacology, Department of Biological Pharmacology, Saint-Louis University Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France. .,University of Paris, Paris, France. .,Department of Biological Pharmacology, Saint-Louis University Hospital, Assistance Publique - Hôpitaux de Paris, FranceHôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, 1 avenue Charles Vellefaux, Paris, 75010, France.
| |
Collapse
|
11
|
Kawato Y, Fukahori H, Nakamura K, Kanno A, Kubo K, Hiramitsu M, Matsuda T, Hanada Y, Furukawa T, Nakajima Y, Kinugasa F, Morokata T. Potential benefit of the cathepsin S inhibitor, ASP1617, as a treatment for systemic lupus erythematosus. Eur J Pharmacol 2022; 919:174826. [DOI: 10.1016/j.ejphar.2022.174826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 11/03/2022]
|
12
|
A rapid and sensitive LC-MS/MS method for determination of the active component K6 in serum of patients with depression. J Pharm Biomed Anal 2022; 213:114691. [DOI: 10.1016/j.jpba.2022.114691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/07/2022] [Accepted: 02/24/2022] [Indexed: 11/21/2022]
|
13
|
Mai K, Singer P, Fahmy AE, Teperman LW, Molmenti EP, Grodstein EI, Castellanos L, Sethna CB. Kidney transplant outcomes in children and adolescents with systemic lupus erythematosus. Pediatr Transplant 2022; 26:e14178. [PMID: 34687584 DOI: 10.1111/petr.14178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/17/2021] [Accepted: 10/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Literature supports equivalent kidney transplant outcomes in adults with systemic lupus erythematosus (SLE) compared with those without SLE. However, there are conflicting and scant data on kidney transplant outcomes, as well as controversy over optimal timing of transplantation, in children and adolescents with SLE. METHODS Analysis included kidney-only transplant recipients aged 2-21 years from 2000 to 2017 enrolled in the Organ Procurement and Transplant Network (OPTN). The relationship between diagnosis (SLE n = 457, non-SLE glomerular disease n = 4492, and non-SLE non-glomerular disease n = 5605) and transplant outcomes was evaluated. The association between dialysis time and outcomes was analyzed in the SLE group only. RESULTS In adjusted models, SLE had higher mortality compared with non-SLE glomerular recipients (HR 1.24 CI 1.07-1.44) and non-glomerular recipients (HR 1.42 CI 1.20-1.70). SLE was associated with higher graft failure compared with non-SLE glomerular (HR 1.42 CI 1.20-1.69) and non-glomerular disease (HR 1.67 CI 1.22-2.28). SLE had a higher risk of acute rejection at 1 year compared with non-glomerular disease (HR 1.39 CI 1.03-1.88). There was a decreased risk of delayed graft function compared with non-SLE glomerular disease (HR 0.54, CI 0.36-0.82). There were no significant associations between dialysis time and transplant outcomes in the SLE group. CONCLUSION SLE in children and adolescents is associated with worse patient and graft survival compared with non-SLE diagnoses. Outcomes in children and adolescents with SLE are not associated with dialysis time. Further studies are needed to assess implications of potential earlier transplantation and shorter time on dialysis prior to transplantation.
Collapse
Affiliation(s)
- Katherine Mai
- Department of Pediatrics, Division of Nephrology, Cohen Children's Medical Center of New York, New Hyde Park, New York, USA
| | - Pamela Singer
- Department of Pediatrics, Division of Nephrology, Cohen Children's Medical Center of New York, New Hyde Park, New York, USA.,Department of Transplantation, Northwell Health, Great Neck, New York, USA
| | - Ahmed E Fahmy
- Department of Transplantation, Northwell Health, Great Neck, New York, USA
| | - Lewis W Teperman
- Department of Transplantation, Northwell Health, Great Neck, New York, USA
| | - Ernesto P Molmenti
- Department of Transplantation, Northwell Health, Great Neck, New York, USA
| | - Elliot I Grodstein
- Department of Transplantation, Northwell Health, Great Neck, New York, USA
| | - Laura Castellanos
- Department of Pediatrics, Division of Nephrology, Cohen Children's Medical Center of New York, New Hyde Park, New York, USA.,Department of Transplantation, Northwell Health, Great Neck, New York, USA
| | - Christine B Sethna
- Department of Pediatrics, Division of Nephrology, Cohen Children's Medical Center of New York, New Hyde Park, New York, USA.,Department of Transplantation, Northwell Health, Great Neck, New York, USA
| |
Collapse
|
14
|
Zheng P, Yu Z, Li L, Liu S, Lou Y, Hao X, Yu P, Lei M, Qi Q, Wang Z, Gao F, Zhang Y, Li Y. Predicting Blood Concentration of Tacrolimus in Patients With Autoimmune Diseases Using Machine Learning Techniques Based on Real-World Evidence. Front Pharmacol 2021; 12:727245. [PMID: 34630104 PMCID: PMC8497784 DOI: 10.3389/fphar.2021.727245] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Tacrolimus is a widely used immunosuppressive drug in patients with autoimmune diseases. It has a narrow therapeutic window, thus requiring therapeutic drug monitoring (TDM) to guide the clinical regimen. This study included 193 cases of tacrolimus TDM data in patients with autoimmune diseases at Southern Medical University Nanfang Hospital from June 7, 2018, to December 31, 2020. The study identified nine important variables for tacrolimus concentration using sequential forward selection, including height, tacrolimus daily dose, other immunosuppressants, low-density lipoprotein cholesterol, mean corpuscular volume, mean corpuscular hemoglobin, white blood cell count, direct bilirubin, and hematocrit. The prediction abilities of 14 models based on regression analysis or machine learning algorithms were compared. Ultimately, a prediction model of tacrolimus concentration was established through eXtreme Gradient Boosting (XGBoost) algorithm with the best predictive ability (R2 = 0.54, mean absolute error = 0.25, and root mean square error = 0.33). Then, SHapley Additive exPlanations was used to visually interpret the variable’s impacts on tacrolimus concentration. In conclusion, the XGBoost model for predicting blood concentration of tacrolimus on the basis of real-world evidence has good predictive performance, providing guidance for the adjustment of regimen in clinical practice.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ze Yu
- Beijing Medicinovo Technology Co. Ltd., Beijing, China
| | - Liren Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shiting Liu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Lou
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Hao
- Dalian Medicinovo Technology Co. Ltd., Dalian, China
| | - Peng Yu
- Beijing Medicinovo Technology Co. Ltd., Beijing, China
| | - Ming Lei
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaona Qi
- Beijing Medicinovo Technology Co. Ltd., Beijing, China
| | - Zeyuan Wang
- Beijing Medicinovo Technology Co. Ltd., Beijing, China
| | - Fei Gao
- Beijing Medicinovo Technology Co. Ltd., Beijing, China
| | - Yuqing Zhang
- Zhongshan School of Medicine, SYSU, Guangzhou, China
| | - Yilei Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Doudka N, Giocanti M, Basso M, Ugdonne R, Barthelemy K, Lacarelle B, Blin O, Solas C, Guilhaumou R. Development and Validation of a Simple and Rapid Ultrahigh-Performance Liquid Chromatography Tandem Spectrometry Method for the Quantification of Hydroxychloroquine in Plasma and Blood Samples in the Emergency Context of SARS-CoV-2 Pandemic. Ther Drug Monit 2021; 43:570-576. [PMID: 33165216 PMCID: PMC8277042 DOI: 10.1097/ftd.0000000000000836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/18/2020] [Indexed: 12/18/2022]
Abstract
ABSTRACT Therapeutic drug monitoring of hydroxychloroquine (HCQ) has been recommended to optimize the treatment of patients with COVID-19. The authors describe an ultrahigh-performance liquid chromatography tandem spectrometry method developed in a context of emergency, to analyze HCQ in both human plasma and blood samples. After adding the labeled internal standard and simple protein precipitation, plasma samples were analyzed using a C18 column. Blood samples required evaporation before analysis. The total chromatographic run time was 4 minutes (including 1.5 minutes of column equilibration). The assay was linear over the calibration range (r2 > 0.99) and up to 1.50 mcg/mL for the plasma samples (5.00 mcg/mL for the blood matrix). The limit of quantification was 0.0150 mcg/mL for plasma samples (0.05 mcg/mL blood matrix) with accuracy and precision ranging from 91.1% to 112% and from 0.750% to 11.1%, respectively. Intraday and interday precision and accuracy values were within 15.0%. No significant matrix effect was observed in the plasma or blood samples. This method was successfully applied to patients treated for COVID-19 infection. A simple and rapid ultrahigh-performance liquid chromatography tandem spectrometry method adapted to HCQ therapeutic drug monitoring in the context of SARS-CoV-2 infection was successfully developed and validated.
Collapse
Affiliation(s)
- Natalia Doudka
- Service de Pharmacologie Clinique et Pharmacovigilance, Hôpital de la Timone
- Aix Marseille Université, Institut de Neurosciences des Systèmes, Aix-Marseille University, The Institut de Neurosciences des Systèmes
| | - Madeleine Giocanti
- Laboratoire de Pharmacocinétique et Toxicologie, Hôpital de la Timone, Clinical and Pharmacological Unit, Timone Hospital, Laboratory of Pharmacokinetic and Toxicology; and
| | - Manon Basso
- Laboratoire de Pharmacocinétique et Toxicologie, Hôpital de la Timone, Clinical and Pharmacological Unit, Timone Hospital, Laboratory of Pharmacokinetic and Toxicology; and
| | - Renée Ugdonne
- Laboratoire de Pharmacocinétique et Toxicologie, Hôpital de la Timone, Clinical and Pharmacological Unit, Timone Hospital, Laboratory of Pharmacokinetic and Toxicology; and
| | - Karine Barthelemy
- Aix-Marseille Univ, Unité des Virus Emergents (UVE) IRD 190, Aix-Marseille University, Unité des Virus Emergents (UVE) IRD 190, Marseille, France
| | - Bruno Lacarelle
- Laboratoire de Pharmacocinétique et Toxicologie, Hôpital de la Timone, Clinical and Pharmacological Unit, Timone Hospital, Laboratory of Pharmacokinetic and Toxicology; and
| | - Olivier Blin
- Service de Pharmacologie Clinique et Pharmacovigilance, Hôpital de la Timone
- Aix Marseille Université, Institut de Neurosciences des Systèmes, Aix-Marseille University, The Institut de Neurosciences des Systèmes
| | - Caroline Solas
- Laboratoire de Pharmacocinétique et Toxicologie, Hôpital de la Timone, Clinical and Pharmacological Unit, Timone Hospital, Laboratory of Pharmacokinetic and Toxicology; and
- Aix-Marseille Univ, Unité des Virus Emergents (UVE) IRD 190, Aix-Marseille University, Unité des Virus Emergents (UVE) IRD 190, Marseille, France
| | - Romain Guilhaumou
- Service de Pharmacologie Clinique et Pharmacovigilance, Hôpital de la Timone
- Aix Marseille Université, Institut de Neurosciences des Systèmes, Aix-Marseille University, The Institut de Neurosciences des Systèmes
| |
Collapse
|
16
|
Wabitsch S, McVey JC, Ma C, Ruf B, Kamenyeva O, McCallen JD, Diggs LP, Heinrich B, Greten TF. Hydroxychloroquine can impair tumor response to anti-PD1 in subcutaneous mouse models. iScience 2020; 24:101990. [PMID: 33490900 PMCID: PMC7807148 DOI: 10.1016/j.isci.2020.101990] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/23/2020] [Accepted: 12/21/2020] [Indexed: 01/03/2023] Open
Abstract
Hydroxychloroquine (HCQ) is a well-known anti-inflammatory drug but is also known as an anti-inflammatory drug. Here, we evaluate the influence of HCQ treatment on the effect of anti-PD1 tumor immunotherapy. Anti-PD1 therapy-sensitive tumor lines MC38, CT26, and RIL-175 were used to investigate the impact of HCQ on anti-PD1 therapy efficacy. In vitro assays demonstrated that HCQ directly inhibited tumor cell growth in all the tested tumor cell lines. HCQ treatment impaired both antigen-specific and nonspecific T-cell production of TNFα and IFNγ in vitro and in vivo. Importantly, in all the three tumor models, HCQ treatment significantly impaired the response to anti-PD1 treatment, accompanying diminished in vivo T-cell activation and reduced tumor-infiltrating, antigen-specific CD8+ T cells. This study shows that HCQ treatment can result in immunotherapy failure due to its immunosuppressive effects that offset both increased MHC-I expression by tumor cell and direct cytotoxicity. HCQ inhibits both antigen-specific and nonspecific CD4+ and CD8+ T function HCQ inhibits tumor cell growth in vitro HCQ impairs anti-PD1 therapy
Collapse
Affiliation(s)
- Simon Wabitsch
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| | - John C McVey
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA
| | - Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| | - Benjamin Ruf
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Justin D McCallen
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| | - Laurence P Diggs
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| | - Bernd Heinrich
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, 10/3B43, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Blanchet B, Jallouli M, Allard M, Ghillani-Dalbin P, Galicier L, Aumaître O, Chasset F, Le Guern V, Lioté F, Smail A, Limal N, Perard L, Desmurs-Clavel H, Le Thi Huong D, Asli B, Kahn JE, Sailler L, Ackermann F, Papo T, Sacré K, Fain O, Stirnemann J, Cacoub P, Leroux G, Cohen-Bittan J, Sellam J, Mariette X, Goulvestre C, Hulot JS, Amoura Z, Vidal M, Piette JC, Jourde-Chiche N, Costedoat-Chalumeau N. Hydroxychloroquine levels in patients with systemic lupus erythematosus: whole blood is preferable but serum levels also detect non-adherence. Arthritis Res Ther 2020; 22:223. [PMID: 32977856 PMCID: PMC7517694 DOI: 10.1186/s13075-020-02291-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/07/2020] [Indexed: 12/26/2022] Open
Abstract
Background Hydroxychloroquine (HCQ) levels can be measured in both serum and whole blood. No cut-off point for non-adherence has been established in serum nor have these methods ever been compared. The aims of this study were to compare these two approaches and determine if serum HCQ cut-off points can be established to identify non-adherent patients. Methods HCQ levels were measured in serum and whole blood from 573 patients with systemic lupus erythematosus (SLE). The risk factors for active SLE (SLEDAI score > 4) were identified by multiple logistic regression. Serum HCQ levels were measured in 68 additional patients known to be non-adherent, i.e. with whole-blood HCQ < 200 ng/mL. Results The mean (± SD) HCQ levels were 469 ± 223 ng/mL in serum and 916 ± 449 ng/mL in whole blood. The mean ratio of serum/whole-blood HCQ levels was 0.53 ± 0.15. In the multivariate analysis, low whole-blood HCQ levels (P = 0.023), but not serum HCQ levels, were independently associated with active SLE. From the mean serum/whole-blood level ratio, a serum HCQ level of 106 ng/mL was extrapolated as the corresponding cut-off to identify non-adherent patients with a sensitivity of 0.87 (95% CI 0.76–0.94) and specificity of 0.89 (95% CI 0.72–0.98). All serum HCQ levels of patients with whole-blood HCQ below the detectable level (< 20 ng/mL) were also undetectable (< 20 ng/mL). Conclusions These data suggest that whole blood is better than serum for assessing the pharmacokinetic/pharmacodynamic relation of HCQ. Our results support the use of serum HCQ levels to assess non-adherence when whole blood is unavailable.
Collapse
Affiliation(s)
- Benoit Blanchet
- AP-HP, Hôpital Cochin, Biologie du médicament - Toxicologie, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.,UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Moez Jallouli
- Service de Médecine interne, Hôpital Hédi Chaker, Sfax, Tunisie
| | - Marie Allard
- Université Paris-Diderot, Sorbonne Paris-Cité, F-75205, Paris, France.,AP-HP, Hôpital Bichat Claude-Bernard, service de médecine interne, 46 rue Henri-Huchard, 75018, Paris, France
| | - Pascale Ghillani-Dalbin
- AP-HP, Hôpital Pitié-Salpêtrière, Département d'immunologie, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Lionel Galicier
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Saint Louis, service d'immunologie clinique, 1 avenue Claude Vellefaux, 75010, Paris, France
| | - Olivier Aumaître
- Université de Clermont-Ferrand, 63003, Clermont-Ferrand, France.,CHU Clermont-Ferrand, Hôpital Gabriel Montpied, service de médecine interne, 58 rue Montalembert, 63003, Clermont-Ferrand cedex1, France
| | - François Chasset
- UPMC, Université Paris 6, Paris, France.,AP-HP, Hôpital Tenon, service de dermatologie allergologie, 4 rue de la Chine, 75020, Paris, France
| | - Véronique Le Guern
- AP-HP, Hôpital Cochin, Centre de référence maladies auto-immunes et systémiques rares, service de médecine interne, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Frédéric Lioté
- Université de Paris, F-75205, Paris, France.,AP-HP, Hôpital Lariboisière, service de rhumatologie, DMU Locomotion, 2 rue Ambroise Paré, 75010, Paris, France
| | - Amar Smail
- CHU Amiens, Hôpital Nord, service de médecine interne, Place Victor Pauchet, 80000, Amiens, France
| | - Nicolas Limal
- AP-HP, Hôpital Henri Mondor, service de médecine interne, 51 avenue du Maréchal de Tassigny, 94000, Créteil, France
| | - Laurent Perard
- Centre Hospitalier Saint Joseph Saint Luc, service de médecine interne, 20 quai Claude Bernard, 69007, Lyon, France
| | - Hélène Desmurs-Clavel
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, service de médecine interne, 5 place d'Arsonval, 69003, Lyon, France
| | - Du Le Thi Huong
- UPMC, Université Paris 6, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Centre de référence pour le Lupus Systémique et le syndrome des Antiphospholipides, service de médecine interne, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Bouchra Asli
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Saint Louis, service d'immunologie clinique, 1 avenue Claude Vellefaux, 75010, Paris, France
| | - Jean-Emmanuel Kahn
- Servie de Médecine Interne, Hôpital Ambroise Paré, Université Paris Saclay, 9 Avenue Charles de Gaulle, 92104, Boulogne-Billancourt, France
| | - Laurent Sailler
- Université Paul-Sabatier, Toulouse, France.,CHU Toulouse, Hôpital Purpan, Service de Médecine Interne, Place Dr Baylac, F-31059, Toulouse, France
| | - Félix Ackermann
- Hôpital Foch, Service de médecine interne, 92150, Suresnes, France
| | - Thomas Papo
- Université Paris-Diderot, Sorbonne Paris-Cité, F-75205, Paris, France.,AP-HP, Hôpital Bichat Claude-Bernard, service de médecine interne, 46 rue Henri-Huchard, 75018, Paris, France
| | - Karim Sacré
- Université Paris-Diderot, Sorbonne Paris-Cité, F-75205, Paris, France.,AP-HP, Hôpital Bichat Claude-Bernard, service de médecine interne, 46 rue Henri-Huchard, 75018, Paris, France
| | - Olivier Fain
- Sorbonne Université, Hôpital Saint Antoine, APHP, service de médecine interne, F 75012, Paris, France
| | - Jérôme Stirnemann
- Hôpitaux Universitaires de Genève, Service de Médecine interne Générale, Avenue Gabrielle Perret Gentil 4, CH-1211, Geneva, Switzerland
| | - Patrice Cacoub
- UPMC, Université Paris 6, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Centre de référence maladies auto-immunes et systémiques rares, service de médecine interne 2, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Gaelle Leroux
- UPMC, Université Paris 6, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Centre de référence maladies auto-immunes et systémiques rares, service de médecine interne 2, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Judith Cohen-Bittan
- AP-HP, Hôpital Pitié-Salpêtrière, service de gériatrie, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Jérémie Sellam
- AP-HP, Hôpital Saint Antoine, Service de Rhumatologie, 184 Rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Xavier Mariette
- Service de Rhumatologie, Hôpitaux Universitaires Paris-Sud, AP-HP, Université Paris-Sud, INSERM UMR 1184, Paris, France
| | - Claire Goulvestre
- AP-HP, Hôpital Cochin, service d'immunologie biologique, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | | | - Zahir Amoura
- UPMC, Université Paris 6, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Centre de référence pour le Lupus Systémique et le syndrome des Antiphospholipides, service de médecine interne, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Michel Vidal
- AP-HP, Hôpital Cochin, Biologie du médicament - Toxicologie, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.,UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Jean-Charles Piette
- UPMC, Université Paris 6, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Centre de référence maladies auto-immunes et systémiques rares, service de médecine interne 2, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | | | - Noémie Jourde-Chiche
- Aix-Marseille Univ, C2VN, INSERM 1263, INRA 1260 ; AP-HM, Centre de Néphrologie et Transplantation Rénale, Marseille, France
| | - Nathalie Costedoat-Chalumeau
- AP-HP, Hôpital Cochin, Centre de référence maladies auto-immunes et systémiques rares, service de médecine interne, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France. .,Université Paris-Descartes, Paris, France. .,INSERM U 1153, Center for Epidemiology and Statistics Sorbonne Paris Cité (CRESS), Paris, France.
| |
Collapse
|
18
|
Roustit M, Guilhaumou R, Molimard M, Drici MD, Laporte S, Montastruc JL. Chloroquine and hydroxychloroquine in the management of COVID-19: Much kerfuffle but little evidence. Therapie 2020; 75:363-370. [PMID: 32473812 PMCID: PMC7244425 DOI: 10.1016/j.therap.2020.05.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/18/2020] [Indexed: 12/27/2022]
Abstract
Chloroquine and hydroxychloroquine are drugs that have shown in vitro activity on the replication of certain coronaviruses. In the context of the SARS-Cov-2 epidemic, the virus responsible for the novel coronavirus disease (COVID-19), these two drugs have been proposed as possible treatments. The results of the first clinical studies evaluating the effect of hydroxychloroquine do not support any efficacy of this drug in patients with COVID-19, due to major methodological weaknesses. Yet, these preliminary studies have aroused considerable media interest, raising fears of massive and uncontrolled use. In the absence of evidence of clinical benefits, the main risk is of exposing patients unnecessarily to the well-known adverse effects of hydroxychloroquine, with a possibly increased risk in the specific setting of COVID-19. In addition, widespread use outside of any recommendation risks compromising the completion of good quality clinical trials. The chloroquine hype, fueled by low-quality studies and media announcements, has yielded to the implementation of more than 150 studies worldwide. This represents a waste of resources and a loss of opportunity for other drugs to be properly evaluated. In the context of emergency, rigorous trials are more than ever needed in order to have, as soon as possible, reliable data on drugs that are possibly effective against the disease. Meanwhile, serious adverse drug reactions have been reported in patients with COVID-19 receiving hydroxychloroquine, justifying to limit its prescription, and to perform suitable cardiac and therapeutic drug monitoring.
Collapse
Affiliation(s)
- M Roustit
- Pharmacologie clinique, Université Grenoble Alpes, CHU de Grenoble, 38043 Grenoble, France.
| | - R Guilhaumou
- Aix Marseille Université, hôpital de la Timone, institut de neuroscience des systèmes, 13005 Marseille, France
| | - M Molimard
- Service de pharmacologie médicale, University Bordeaux, Inserm U1219, 33076 Bordeaux, France
| | - M-D Drici
- Pharmacovigilance - Department of Pharmacology, Pasteur Hospital, 06001 Nice, France
| | - S Laporte
- Université Jean Monnet, Université de Lyon, et unité de recherche clinique, innovation, pharmacologie, CHU Saint-Etienne, 42055 Saint-Étienne, France
| | - J-L Montastruc
- Service de pharmacologie médicale et clinque, centre de pharmacovigilance, de pharmacoépidémiologie et d'informations sur le médicament, CIC INSERM 1436, faculté de Médecine, centre hospitalier universitaire de Toulouse, 31000 Toulouse, France
| |
Collapse
|
19
|
Potential Molecular Mechanisms of Zhibai Dihuang Wan in Systemic Lupus Erythematosus Based on Network Biology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7842179. [PMID: 32351603 PMCID: PMC7178533 DOI: 10.1155/2020/7842179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/25/2020] [Indexed: 12/24/2022]
Abstract
Systemic lupus erythematosus (SLE) is a refractory autoimmune disease. Zhibai Dihuang Wan (ZDW) has frequently been used for treating SLE in China and been proved to have a prominent role in decreasing SLE patients’ morality rate. However, the active substances in ZDW and the molecular mechanisms of ZDW in SLE remain unclear. This study identified the bioactive compounds and delineated the molecular targets and potential pathways of ZDW by using a network biology approach. First, we collected putative targets of ZDW based on TCMSP, GeneCards, and STITCH databases and built a network containing the interactions between the putative targets of ZDW and known therapeutic targets of SLE. Then, the key hubs were imported to DAVID Bioinformatics Resources 6.7 to perform gene ontology biological process (GOBP) and pathway enrichment analysis. A total of 95 nodes including 73 putative targets of ZDW were determined as major hubs in terms of their node degree. The results of GOBP and pathway enrichment analysis indicated that putative targets of ZDW mostly were involved in various pathways associated with inflammatory response and apoptosis. More importantly, eleven putative targets of ZDW (CASP3, BCL2, BAX, CYCS, NFKB1, NFKBIA, IL-6, IL-1β, PTGS2, CCL2, and TNF-α) were recognized as active factors involved in the main biological functions of treatment, implying the underlying mechanisms of ZDW acting on SLE. This study provides novel insights into the mechanisms of ZDW in SLE, from the molecular level to the pathway level.
Collapse
|
20
|
Therapeutic Drug Monitoring of Tacrolimus-Personalized Therapy: Second Consensus Report. Ther Drug Monit 2019; 41:261-307. [DOI: 10.1097/ftd.0000000000000640] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Brunet M, van Gelder T, Åsberg A, Haufroid V, Hesselink DA, Langman L, Lemaitre F, Marquet P, Seger C, Shipkova M, Vinks A, Wallemacq P, Wieland E, Woillard JB, Barten MJ, Budde K, Colom H, Dieterlen MT, Elens L, Johnson-Davis KL, Kunicki PK, MacPhee I, Masuda S, Mathew BS, Millán O, Mizuno T, Moes DJAR, Monchaud C, Noceti O, Pawinski T, Picard N, van Schaik R, Sommerer C, Vethe NT, de Winter B, Christians U, Bergan S. Therapeutic Drug Monitoring of Tacrolimus-Personalized Therapy: Second Consensus Report. Ther Drug Monit 2019. [DOI: 10.1097/ftd.0000000000000640
expr 845143713 + 809233716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
22
|
Llanos S, Megias D, Blanco-Aparicio C, Hernández-Encinas E, Rovira M, Pietrocola F, Serrano M. Lysosomal trapping of palbociclib and its functional implications. Oncogene 2019; 38:3886-3902. [PMID: 30692638 PMCID: PMC6756094 DOI: 10.1038/s41388-019-0695-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 12/31/2018] [Accepted: 01/04/2019] [Indexed: 01/10/2023]
Abstract
Palbociclib is a selective inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6) approved for the treatment of some cancers. The main mechanism of action of palbociclib is to induce cell cycle arrest and senescence on responsive cells. Here, we report that palbociclib concentrates in intracellular acidic vesicles, where it can be readily observed due to its intrinsic fluorescence, and it is released from these vesicles upon dilution or washing out of the extracellular medium. This reversible storage of drugs into acidic vesicles is generally known as lysosomal trapping and, based on this, we uncover novel properties of palbociclib. In particular, a short exposure of cells to palbociclib is sufficient to produce a stable cell-cycle arrest and long-term senescence. Moreover, after washing out the drug, palbociclib-treated cells release the drug to the medium and this conditioned medium is active on susceptible cells. Interestingly, cancer cells resistant to palbociclib also accumulate and release the drug producing paracrine senescence on susceptible cells. Finally, other lysosomotropic drugs, such as chloroquine, interfere with the accumulation of palbociclib into lysosomes, thereby reducing the minimal dose of palbociclib required for cell-cycle arrest and senescence. In summary, lysosomal trapping explains the prolonged temporal activity of palbociclib, the paracrine activity of exposed cells, and the cooperation with lysosomotropic drugs. These are important features that may help to improve the therapeutic dosing and efficacy of palbociclib. Finally, two other clinically approved CDK4/6 inhibitors, ribociclib and abemaciclib, present a similar behavior as palbociclib, suggesting that lysosomal trapping is a property common to all three clinically-approved CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Susana Llanos
- Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| | - Diego Megias
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | | | - Miguel Rovira
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Federico Pietrocola
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Manuel Serrano
- Spanish National Cancer Research Center (CNIO), Madrid, Spain.
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
23
|
Zhou T, Lin S, Yang S, Lin W. Efficacy and safety of tacrolimus in induction therapy of patients with lupus nephritis. Drug Des Devel Ther 2019; 13:857-869. [PMID: 30880918 PMCID: PMC6420100 DOI: 10.2147/dddt.s189156] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The purpose of this study was to detect the efficacy and safety of tacrolimus (TAC) in induction therapy of patients with lupus nephritis. METHODS Associated studies were extracted from the PubMed and the Cochrane Library on July 10, 2018, and applicable investigations were pooled and analyzed by meta-analysis. Data on complete remission (CR), total remission (TR; complete plus partial remission), proteinuria levels, urine erythrocyte number, albumin, glomerular filtration rate, negative rate of ds-DNA, C3 levels, C4 levels, systemic lupus erythematosus disease activity index (SLE-DAI), etc, were extracted and pooled using RevMan 5.3. RESULTS In the therapeutic regimen of TAC + glucocorticoids (GC) vs cyclophosphamide (CYC) + GC, the results indicated that the TAC group had high values of CR, TR, albumin, and negative rate of ds-DNA, and low values of proteinuria levels and SLE-DAI when compared with those in CYC group (all P<0.05). In the therapeutic regimen comprising TAC + GC vs mycophenolate mofetil (MMF) + GC, the results indicated that the difference of CR, TR, proteinuria levels, and albumin between TAC group and MMF group were not significant (all P>0.05). In the therapeutic regimen comprising TAC + MMF + GC vs CYC + GC, multitarget therapy group showed higher values of CR, TR, urinary protein decline, and rise of serum albumin when compared with CYC group (all P<0.05). CONCLUSION TAC is an effective and safe agent in induction therapy of patients with lupus nephritis.
Collapse
Affiliation(s)
- Tianbiao Zhou
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China,
| | - Shujun Lin
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China,
| | - Shen Yang
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| | - Wenshan Lin
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, Shantou 515041, China,
| |
Collapse
|
24
|
Abstract
Although it has been widely acknowledged for more than two decades that transition from pediatric to adult care is a vulnerable time for adolescents and young adults with rheumatic diseases, current primary and subspecialty care transition and transfer processes remain inadequate. Barriers to improving transition include complex health care systems, neurodevelopmental challenges of adolescents and young adults, and insufficient transition-related education and resources for health care providers. Standardized, evidence-based transition interventions are sorely needed to establish best practices. Quality improvement approaches such as the Six Core Elements of Health Care Transition offer opportunities to improve transition care for teens and young adults.
Collapse
|
25
|
Morassi A, Rivera-Vélez SM, Slovak JE, Court MH, Villarino NF. Ex vivo binding of the immunosuppressant mycophenolic acid to dog and cat plasma proteins and the effect of co-incubated dexamethasone and prednisolone. J Vet Pharmacol Ther 2018; 41:513-521. [PMID: 29687456 DOI: 10.1111/jvp.12507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 03/26/2018] [Indexed: 11/30/2022]
Abstract
Mycophenolic acid (MPA) has been shown to be promising for the treatment of autoimmune diseases in dogs and cats. In humans, MPA is highly bound to plasma proteins (~97%). It has been recommended to monitor free drug plasma concentrations because the free MPA correlates with its immunosuppressive effect. However, it is unknown if MPA is highly bound to plasma proteins in dogs and cats. The objectives of this study were to determine the extent of plasma protein binding of MPA and evaluate the effect of prednisolone and dexamethasone on the extent of protein binding of MPA in dogs and cats. The extent of plasma protein binding of MPA was determined in plasma collected from clinically healthy adult cats (n = 13) and dogs (n = 14) by combining high-throughput dialysis and ultra-high-liquid chromatography. This study reveals that MPA is highly bound to plasma proteins (>90%) in dogs and cats, mean extent of binding of MPA at 15 μg/ml to plasma proteins being 96% (range, 95%-97%) and 92% (range, 90%-93%) for dogs and cats, respectively. In dog plasma, MPA is primarily bound to albumin. In vitro, prednisolone increased the unbound MPA in dogs (p < .01) but not in cats (p = .07) while dexamethasone had no effect on MPA plasma binding in either species (p > .05). Results of this study provide valuable information for designing future pharmacokinetic and pharmacodynamic studies and also therapeutic monitoring programs for dogs and cats.
Collapse
Affiliation(s)
- A Morassi
- Department of Veterinary Clinical Sciences, Program in Individualized Medicine, Washington State University, Pullman, Washington
| | - S M Rivera-Vélez
- Department of Veterinary Clinical Sciences, Program in Individualized Medicine, Washington State University, Pullman, Washington
| | - J E Slovak
- Department of Veterinary Clinical Sciences, Program in Individualized Medicine, Washington State University, Pullman, Washington
| | - M H Court
- Department of Veterinary Clinical Sciences, Program in Individualized Medicine, Washington State University, Pullman, Washington
| | - N F Villarino
- Department of Veterinary Clinical Sciences, Program in Individualized Medicine, Washington State University, Pullman, Washington
| |
Collapse
|