1
|
Patel A, Calkins TL, Vidal JD, Coder PS, Carrier S, Gokulrangan G, Chakilam ASR, Akare S, Briscoe RJ, Mondal MS. Effects of dopamine agonists on the estradiol-induced prolactin surge in ovariectomized female Wistar Han rats. Toxicol Sci 2025; 205:65-73. [PMID: 40070087 DOI: 10.1093/toxsci/kfaf027] [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] [Indexed: 04/30/2025] Open
Abstract
Dopamine agonists (DAs) are approved for the treatment of hypodopaminergic pathologies, including Parkinson's disease, restless legs syndrome, and periodic limb movement disorder. During drug development, drugs acting on dopaminergic receptors are often associated with a rat-specific endocrine tumor response, including changes in fertility, which are ascribed to DA-induced suppression of pituitary prolactin release. Although these effects are not observed in or relevant to humans, given species differences in the effects of prolactin on reproductive organs, modeling DA-mediated changes in prolactin and the reproductive system remains important for preclinical drug development. We investigated the effects of 2 D2/D3 DAs, pergolide and rotigotine, on the estradiol (E2)-induced prolactin surge in ovariectomized (OVX) female Wistar Han rats. Daily treatment with DAs over 7 days led to a reduction in the prolactin surge in E2-implanted OVX rats. Specifically, pergolide induced a significant decrease in prolactin levels at all time points compared with the OVX-E2 control group. Similarly, rotigotine dose-dependently suppressed plasma prolactin levels compared with the OVX-E2 control group. This study demonstrates the utility of the OVX rat model in evaluating the effects of DAs on the E2-induced prolactin surge. These results support the use of rotigotine, a DA with a long history of safe human use without significant endocrine-related adverse events, as a positive control at a dose level of 5.0 mg/kg/day for future nonclinical toxicity studies investigating the effects of novel DAs on reproductive hormones in rats.
Collapse
Affiliation(s)
- Atish Patel
- Cerevel Therapeutics, LLC, Cambridge, MA 02141, United States
| | - Travis L Calkins
- Charles River Laboratories Ashland, LLC, Ashland, OH 44805, United States
| | - Justin D Vidal
- Charles River Laboratories Ashland, LLC, Ashland, OH 44805, United States
| | - Pragati S Coder
- Charles River Laboratories Ashland, LLC, Ashland, OH 44805, United States
| | - Scott Carrier
- Cerevel Therapeutics, LLC, Cambridge, MA 02141, United States
| | | | | | - Sandeep Akare
- Cerevel Therapeutics, LLC, Cambridge, MA 02141, United States
| | | | - Madhu S Mondal
- Cerevel Therapeutics, LLC, Cambridge, MA 02141, United States
| |
Collapse
|
2
|
Deleuran M, Dézfoulian B, Elberling J, Knutar I, Lapeere H, Lossius AH, Schuttelaar MLA, Stockman A, Wikström E, Bradley M, de Bruin-Weller M, Gutermuth J, Mandelin JM, Schmidt MC, Thyssen JP, Vestergaard C. Systemic anti-inflammatory treatment of atopic dermatitis during conception, pregnancy and breastfeeding: Interdisciplinary expert consensus in Northern Europe. J Eur Acad Dermatol Venereol 2024; 38:31-41. [PMID: 37818828 DOI: 10.1111/jdv.19512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/09/2023] [Indexed: 10/13/2023]
Abstract
Treating atopic dermatitis (AD) in pregnant or breastfeeding women, and in women and men with AD aspiring to be parents is difficult and characterized by uncertainty, as evidence to inform decision-making on systemic anti-inflammatory treatment is limited. This project mapped consensus across dermatologists, obstetricians and patients in Northwestern Europe to build practical advice for managing AD with systemic anti-inflammatory treatment in men and women of reproductive age. Twenty-one individuals (sixteen dermatologists, two obstetricians and three patients) participated in a two-round Delphi process. Full consensus was reached on 32 statements, partial consensus on four statements and no consensus on four statements. Cyclosporine A was the first-choice long-term systemic AD treatment for women preconception, during pregnancy and when breastfeeding, with short-course prednisolone for flare management. No consensus was reached on second-choice systemics preconception or during pregnancy, although during breastfeeding dupilumab and azathioprine were deemed suitable. It may be appropriate to discuss continuing an existing systemic AD medication with a woman if it provides good disease control and its benefits in pregnancy outweigh its risks. Janus kinase (JAK) inhibitors, methotrexate and mycophenolate mofetil should be avoided by women during preconception, pregnancy and breastfeeding, with medication-specific washout periods advised. For men preconception: cyclosporine A, azathioprine, dupilumab and corticosteroids are appropriate; a 3-month washout prior to conception is desirable for methotrexate and mycophenolate mofetil; there was no consensus on JAK inhibitors. Patient and clinician education on appropriate (and inappropriate) AD treatments for use in pregnancy is vital. A shared-care framework for interdisciplinary management of AD patients is advocated and outlined. This consensus provides interdisciplinary clinical guidance to clinicians who care for patients with AD before, during and after pregnancy. While systemic AD medications are used uncommonly in this patient group, considerations in this article may help patients with severe refractory AD.
Collapse
Affiliation(s)
- M Deleuran
- Department of Dermatology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| | - B Dézfoulian
- Dermatology Department, Liège University Hospital, Liège, Belgium
| | - J Elberling
- Department of Dermatology and Allergy, Department of Clinical Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - I Knutar
- Department of Dermatology, Vaasa Central Hospital, Vaasa, Finland
| | - H Lapeere
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - A H Lossius
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
| | - M L A Schuttelaar
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A Stockman
- Department of Dermatology, AZ Delta, Torhout, Belgium
| | - E Wikström
- Dermatology Health Clinic, Oulu, Finland
| | - M Bradley
- Dermatology and Venereology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
| | - M de Bruin-Weller
- Department of Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J Gutermuth
- Vrije Universiteit Brussel (VUB), SKIN Research Group, Department of Dermatology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - J M Mandelin
- Department of Dermatology, Helsinki University Central Hospital, Helsinki, Finland
| | - M C Schmidt
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Aarhus, Denmark
| | - J P Thyssen
- Department of Dermatology and Venereology, Bispebjerg Hospital, Copenhagen, Denmark
| | - C Vestergaard
- Department of Dermatology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Hisada S, Tsubota K, Inoue K, Yamada H, Ikeda T, Sistare FD. Survey of tumorigenic sensitivity in 6-month rasH2-Tg mice studies compared with 2-year rodent assays. J Toxicol Pathol 2022; 35:53-73. [PMID: 35221496 PMCID: PMC8828610 DOI: 10.1293/tox.2021-0031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/07/2021] [Indexed: 12/04/2022] Open
Abstract
The pharmacokinetic endpoint of a 25-fold increase in human exposure is one of the specified criteria for high-dose selection for 2-year carcinogenicity studies in rodents according to ICH S1C(R2). However, this criterion is not universally accepted for 6-month carcinogenicity tests in rasH2-Tg mice. To evaluate an appropriate multiple for rasH2-Tg mice, we evaluated data for 53 compounds across five categories of rasH2-Tg mouse-positive [(1) genotoxic and (2) non-genotoxic] carcinogens and rasH2-Tg mouse-negative [(3) non-genotoxic carcinogens with clear or uncertain human relevance; (4) non-genotoxic rodent-specific carcinogens; and (5) non-carcinogens], and surveyed their tumorigenic activities and high doses in rasH2-Tg mice and 2-year rodent models. Our survey indicated that area under the curve (AUC) margins (AMs) or body surface area-adjusted dose ratios (DRs) of tumorigenesis in rasH2-Tg mice to the maximum recommended human dose (MRHD) were 0.05- to 5.2-fold in 6 category (1) compounds with small differences between models and 0.2- to 47-fold in 7 category (2) including three 2-year rat study-negative compounds. Among all 53 compounds, including 40 compounds of the rasH2-Tg mouse-negative category (3), (4), and (5), no histopathologic risk factors for rodent neoplasia were induced only at doses above 50-fold AM or DR in rasH2-Tg mice except for two compounds, which induced hyperplasia and had no relationship with the tumors observed in the rasH2-Tg mouse or 2-year rodent studies. From the results of these surveys, we confirmed that exceeding a high dose level of 50-fold AM in rasH2-Tg mouse carcinogenicity studies does not appear to be of value.
Collapse
Affiliation(s)
- Shigeru Hisada
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- ASKA Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-chome, Fujisawa-shi, Kanagawa 251-8555, Japan
| | - Kenjiro Tsubota
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba-shi, Ibaraki
305-8585, Japan
| | - Kenji Inoue
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- Maruho Co., Ltd., 93 Chudoji Awatacho, Shimogyo-ku, Kyoto
600-8815, Japan
| | - Hisaharu Yamada
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku,
Saitama 331-9530, Japan
| | - Takanori Ikeda
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- MSD K.K., 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667,
Japan
| | - Frank D. Sistare
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA
19486-0004, USA
- Current Address: 315 Meadowmont Lane, Chapel Hill, NC 27517,
USA
| |
Collapse
|
4
|
MicroRNA-495 alleviates ulcerative interstitial cystitis via inactivating the JAK-STAT signaling pathway by inhibiting JAK3. Int Urogynecol J 2021; 32:1253-1263. [PMID: 33416962 DOI: 10.1007/s00192-020-04593-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION AND HYPOTHESIS As a notable chronic disorder, the incidence of interstitial cystitis (IC) has been documented to have increased among the female population with activity in microRNA-495 (miR-495) implicated in this disease. The current study was aimed at elucidating the effects associated with miR-495 on the inflammatory response and bladder fibrosis in rats with ulcerative IC via the JAK-STAT pathway by targeting JAK3. METHODS Ulcerative IC rat models were established. The targeting relationship between JAK3 and miR-495 was evaluated using luciferase reporter assay. After gain- or loss-of-function assays, mast-cell infiltration was assessed using toluidine blue staining, bladder fibrosis using Masson staining, and NO content using nitrate reductase method. JAK3 protein expression was detected by immunohistochemistry, JAK3, STAT1, STAT3, TGFβ-1, Col-I, Col-III, JAK1, JAK2, p-STAT1, and p-STAT3 expression by RT-qPCR and Western blot analysis, and serum IL-6, IL-8, IL-10, IL-17, and TNF-α levels in rats by ELISA. RESULTS Following transfection of overexpressed miR-495 or siRNA-JAK3, a diminished degree of mast-cell infiltration, number of mast cells, bladder fibrosis, NO content, JAK3-positive expression, mRNA expression of JAK3, STAT1, STAT3, TGFβ-1, Col-I, Col-III, protein expression of JAK1, JAK2, JAK3, p-STAT1, p-STAT3, and expression of IL-6, IL-8, IL-10, IL-17, and TNF-α were identified. CONCLUSIONS Collectively, our key findings provide evidence supporting the notion that the overexpression of miR-495 ameliorates inflammatory response and bladder fibrosis in ulcerative IC rat models via inactivation of the JAK-STAT signaling pathway by inhibiting JAK3.
Collapse
|
5
|
Widyastuti R, Prastowo S, Sumarsono SH, Lubis A, Hartady T, Syamsunarno MRAA, Sudiman J. Deleterious effect of short-term gavage of an ethanol extract of cogon grass ( Imperata cylindrica L.) roots on testis and epididymal sperm quality. Vet World 2020; 13:1311-1318. [PMID: 32848305 PMCID: PMC7429374 DOI: 10.14202/vetworld.2020.1311-1318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/19/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND AIM Cogon grass (Imperata cylindrica L.) (CGG) is a herbal medicine that could be developed into a male antifertility agent. The present study aims to determine the effect of an ethanol extract of CGG roots on mice testicular activity, reproductive hormone levels, and epididymal sperm quality. MATERIALS AND METHODS This study was designed as completely randomized with three different doses, such as an ethanol extract of CGG roots at 0 (control), 90, and 115 mg/kg body weight. In total, 21 male DDY mice strain were treated with the CGG extract (by gavage) for 14 days, followed by an evaluation of reproductive organs, epididymal sperm quality, testis histology, histomorphometry, and reproductive hormone assays. All quantitative data were analyzed by analysis of variance, followed by Tukey's post hoc test at α=0.05. RESULTS The results showed that the administration of the CGG root ethanol extract disrupted the testis interstitial area and seminiferous tubules, resulting in decreased epididymal sperm quality as well as serum testosterone levels in a dose-dependent pattern. CONCLUSION Oral administration of a CGG root ethanol extract induced testicular damage, decreased epididymal sperm quality, and impaired testosterone secretion.
Collapse
Affiliation(s)
- Rini Widyastuti
- Laboratory of Animal Reproduction and Artificial Insemination, Department of Animal Production, Faculty of Animal Husbandry, Universitas Padjadjaran, Jl. Raya Bandung - Sumedang km. 21, West Java, Indonesia
- Central Laboratory, Universitas Padjadjaran, Jl. Raya Bandung - Sumedang km. 21, Jatinangor Sumedang, West Java, Indonesia
| | - Sigit Prastowo
- Department of Animal Science, Faculty of Agriculture, Universitas Sebelas Maret, Surakarta, Indonesia
- Centre for Biotechnology and Biodiversity Research and Development, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Sony H. Sumarsono
- Physiology, Developmental Biology and Biomedical Science Research Group, School of Life Science and Technology, Bandung Institute of Technology, Bandung, West Java, Indonesia
| | - Alkaustariyah Lubis
- Under Graduate Medical Study Program, Faculty of Medicine, Universitas Padjajaran, Jl. Raya Bandung - Sumedang km. 21, Jatinangor Sumedang, West Java, Indonesia
| | - Tyagita Hartady
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung - Sumedang km. 21, Jatinangor Sumedang, West Java, Indonesia
| | - Mas R. A. A. Syamsunarno
- Central Laboratory, Universitas Padjadjaran, Jl. Raya Bandung - Sumedang km. 21, Jatinangor Sumedang, West Java, Indonesia
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Jl. Raya Bandung - Sumedang km. 21, Jatinangor Sumedang, West Java, Indonesia
| | - Jaqueline Sudiman
- Department of Obstetrics and Gynecology, Faculty of Medicine, Udayana University, Jl. PB. Sudirman, Denpasar, Bali, Indonesia
| |
Collapse
|
6
|
Van Cott A, Frericks M, Hastings C, Honarvar N, Flick B, Fabian E, van Ravenzwaay B. Uterine adenocarcinoma in the rat induced by afidopyropen. An analysis of the lesion's induction, progression and its relevance to humans. Regul Toxicol Pharmacol 2018; 95:29-51. [DOI: 10.1016/j.yrtph.2018.02.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/29/2018] [Accepted: 02/28/2018] [Indexed: 11/16/2022]
|
7
|
Collinge M, Ball DJ, Bowman CJ, Nilson AL, Radi ZA, Vogel WM. Immunologic effects of chronic administration of tofacitinib, a Janus kinase inhibitor, in cynomolgus monkeys and rats - Comparison of juvenile and adult responses. Regul Toxicol Pharmacol 2018; 94:306-322. [PMID: 29454012 DOI: 10.1016/j.yrtph.2018.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 01/25/2023]
Abstract
Tofacitinib, an oral Janus kinase (JAK) inhibitor for treatment of rheumatoid arthritis, targets JAK1, JAK3, and to a lesser extent JAK2 and TYK2. JAK1/3 inhibition impairs gamma common chain cytokine receptor signaling, important in lymphocyte development, homeostasis and function. Adult and juvenile cynomolgus monkey and rat studies were conducted and the impact of tofacitinib on immune parameters (lymphoid tissues and lymphocyte subsets) and function (T-dependent antibody response (TDAR), mitogen-induced T cell proliferation) assessed. Tofacitinib administration decreased circulating T cells and NK cells in juvenile and adult animals of both species. B cell decreases were observed only in rats. These changes and decreased lymphoid tissue cellularity are consistent with the expected pharmacology of tofacitinib. No differences were observed between juvenile and adult animals, either in terms of doses at which effects were observed or differential effects on immune endpoints. Lymphomas were observed in three adult monkeys. Tofacitinib impaired the primary TDAR in juvenile monkeys, although a recall response was generated. Complete or partial reversal of the effects on the immune system was observed.
Collapse
Affiliation(s)
- Mark Collinge
- Pfizer Worldwide Research and Development, Drug Safety R&D, Eastern Point Road, Groton, CT 06340, USA.
| | - Douglas J Ball
- Pfizer Worldwide Research and Development, Drug Safety R&D, Eastern Point Road, Groton, CT 06340, USA
| | - Christopher J Bowman
- Pfizer Worldwide Research and Development, Drug Safety R&D, Eastern Point Road, Groton, CT 06340, USA
| | - Andrea L Nilson
- Pfizer Worldwide Research and Development, Drug Safety R&D, Eastern Point Road, Groton, CT 06340, USA
| | - Zaher A Radi
- Pfizer Worldwide Research and Development, Drug Safety R&D, One Portland Street, Cambridge, MA 02139, USA
| | - W Mark Vogel
- Pfizer Worldwide Research and Development, Drug Safety R&D, One Portland Street, Cambridge, MA 02139, USA
| |
Collapse
|
8
|
Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov 2017; 17:78. [PMID: 29282366 PMCID: PMC6168198 DOI: 10.1038/nrd.2017.267] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This corrects the article DOI: 10.1038/nrd.2017.201.
Collapse
|
9
|
JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov 2017; 16:843-862. [PMID: 29104284 DOI: 10.1038/nrd.2017.201] [Citation(s) in RCA: 698] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of cytokines as key drivers of immune-mediated diseases has spurred efforts to target their associated signalling pathways. Janus kinases (JAKs) are essential signalling mediators downstream of many pro-inflammatory cytokines, and small-molecule inhibitors of JAKs (jakinibs) have gained traction as safe and efficacious options for the treatment of inflammation-driven pathologies such as rheumatoid arthritis, psoriasis and inflammatory bowel disease. Building on the clinical success of first-generation jakinibs, second-generation compounds that claim to be more selective are currently undergoing development and proceeding to clinical trials. However, important questions remain about the advantages and limitations of improved JAK selectivity, optimal routes and dosing regimens and how best to identify patients who will benefit from jakinibs. This Review discusses the biology of jakinibs from a translational perspective, focusing on recent insights from clinical trials, the development of novel agents and the use of jakinibs in a spectrum of immune and inflammatory diseases.
Collapse
|