1
|
Berridge B, Pierson J, Pettit S, Stockbridge N. Challenging the status quo: a framework for mechanistic and human-relevant cardiovascular safety screening. FRONTIERS IN TOXICOLOGY 2024; 6:1352783. [PMID: 38590785 PMCID: PMC10999590 DOI: 10.3389/ftox.2024.1352783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Traditional approaches to preclinical drug safety assessment have generally protected human patients from unintended adverse effects. However, these assessments typically occur too late to make changes in the formulation or in phase 1 and beyond, are highly dependent on animal studies and have the potential to lead to the termination of useful drugs due to liabilities in animals that are not applicable in patients. Collectively, these elements come at great detriment to both patients and the drug development sector. This phenomenon is particularly problematic in the area of cardiovascular safety assessment where preclinical attrition is high. We believe that a more efficient and translational approach can be defined. A multi-tiered assessment that leverages our understanding of human cardiovascular biology, applies human cell-based in vitro characterizations of cardiovascular responses to insult, and incorporates computational models of pharmacokinetic relationships would enable earlier and more translational identification of human-relevant liabilities. While this will take time to develop, the ultimate goal would be to implement such assays both in the lead selection phase as well as through regulatory phases.
Collapse
Affiliation(s)
| | - Jennifer Pierson
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Syril Pettit
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Norman Stockbridge
- US Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, United States
| |
Collapse
|
2
|
Sevim C, Tsatsakis A, Taghizadehghalehjoughi A, Ozkaraca M, Kara M, Genc S, Mendil AS, Yeni Y, Nikolouzakis TK, Ozcagli E. Investigation of the miRNA levels changes to acceptable daily intake dose pesticide mixture exposure on rat mesentery and pancreas. CHEMOSPHERE 2024; 349:140712. [PMID: 38036224 DOI: 10.1016/j.chemosphere.2023.140712] [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: 09/07/2023] [Revised: 11/12/2023] [Accepted: 11/12/2023] [Indexed: 12/02/2023]
Abstract
Consumers are constantly exposed to a variety of chemical mixtures as part of their everyday activities and lifestyle. Food, water and commercial products are only some examples of the possible ways people get exposed to these mixtures. However, following federal and local guidelines for risk assessment related to chemical exposure, risk analysis focuses on a single substance exposure scenario and not on a mixture, as in real life. Realizing the pronounced gap of this methodology, the real-life risk simulation scenario approach tries to address this problem by investigating the possible effect of long-term exposure to chemical mixtures closely resembling the actual circumstances of modern life. As part of this effort, this study aimed to identify the cumulative effects of pesticides belonging to different classes and commonly used commercial products on long-term exposure with realistic doses. Sprague Dawley rats were given a pesticide mix of active ingredients and formulation chemicals in a daily acceptable dose (ADI) and 10xADI for 90 days. Following thorough everyday documentation of possible side-effects, after 90 days all animals were sacrificed and their organs were examined. Exposure to pesticides particularly affects the miRNA levels at that point will provide us with more information about whether they can be potential biomarkers.
Collapse
Affiliation(s)
- Cigdem Sevim
- Deparment of Medical Pharmacology, Faculty of Medicine, Kastamonu University , 37200, Kastamonu, Turkey.
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, Crete University, 71003, Heraklion, Greece.
| | - Ali Taghizadehghalehjoughi
- Deparment of Medical Pharmacology, Faculty of Medicine, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey.
| | - Mustafa Ozkaraca
- Deparment of Pathology, Faculty of Veterinary, Cumhuriyet University , 58070, Sivas, Turkey.
| | - Mehtap Kara
- Deparment of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University , 34116, Istanbul, Turkey.
| | - Sidika Genc
- Deparment of Medical Pharmacology, Faculty of Medicine, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey.
| | - Ali Sefa Mendil
- Deparment of Pathology, Faculty of Veterinary, Erciyes University , 38280, Kayseri, Turkey.
| | - Yesim Yeni
- Deparment of Medical Pharmacology, Faculty of Medicine, Turgut Özal University, 44210, Malatya, Turkey.
| | | | - Eren Ozcagli
- Deparment of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University , 34116, Istanbul, Turkey.
| |
Collapse
|
3
|
Abstract
Vascular injury can be induced by different classes of drug candidates, and it can affect the mesenteric vasculature. Sampling of the mesenteric vessels in the rat is crucial for proper assessment of potential adverse or pharmacologic effects of drugs in nonclinical rodent studies. To date, several sampling and processing techniques for the histopathologic evaluation of the mesenteric artery in rodents have been described and used in studies with candidate drugs that may affect the vascular system. However, most of those techniques require a significant amount of time and effort. A less labor-intensive, time-consuming, and expensive technique that allows examination of the mesentery vasculature with abundant longitudinal and cross sections of the vessels when examined microscopically was developed and presented here.
Collapse
|
4
|
Frazier KS, Engelhardt JA, Fant P, Guionaud S, Henry SP, Leach MW, Louden C, Scicchitano MS, Weaver JL, Zabka TS. Scientific and Regulatory Policy Committee Points-to-consider Paper*. Toxicol Pathol 2015; 43:915-34. [DOI: 10.1177/0192623315570340] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Drug-induced vascular injury (DIVI) is a recurrent challenge in the development of novel pharmaceutical agents. Although DIVI in laboratory animal species has been well characterized for vasoactive small molecules, there is little available information regarding DIVI associated with biotherapeutics such as peptides/proteins or antibodies. Because of the uncertainty about whether DIVI in preclinical studies is predictive of effects in humans and the lack of robust biomarkers of DIVI, preclinical DIVI findings can cause considerable delays in or even halt development of promising new drugs. This review discusses standard terminology, characteristics, and mechanisms of DIVI associated with biotherapeutics. Guidance and points to consider for the toxicologist and pathologist facing preclinical cases of biotherapeutic-related DIVI are outlined, and examples of regulatory feedback for each of the mechanistic types of DIVI are included to provide insight into risk assessment.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael W. Leach
- Pfizer—Drug Safety Research and Development, Andover, Massachusetts, USA
| | | | | | | | | |
Collapse
|
5
|
Elmore SA, Cora MC, Gruebbel MM, Hayes SA, Hoane JS, Koizumi H, Peters R, Rosol TJ, Singh BP, Szabo KA. Proceedings of the 2014 National Toxicology Program Satellite Symposium. Toxicol Pathol 2014; 43:10-40. [PMID: 25385331 DOI: 10.1177/0192623314555526] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The 2014 annual National Toxicology Program (NTP) Satellite Symposium, entitled "Pathology Potpourri" was held in Washington, D.C., in advance of the Society of Toxicologic Pathology's 33rd annual meeting. The goal of this annual NTP Symposium is to present current diagnostic pathology or nomenclature issues to the toxicologic pathology community. This article presents summaries of the speakers' presentations, including diagnostic or nomenclature issues that were presented, along with select images that were used for audience voting and discussion. Some lesions and topics covered during the symposium included a pulmonary mucinous adenocarcinoma in a male B6C3F1 mouse; plexiform vasculopathy in Wistar Han (Crl:WI[Han]) rats; staging of the estrous cycle in rats and mice; peri-islet fibrosis, hemorrhage, lobular atrophy and inflammation in male Sprague-Dawley (SD) rats; retinal dysplasia in Crl:WI[Han] rats and B6C3F1 mice; multicentric lymphoma with intravascular microemboli and tumor lysis syndrome, and 2 cases of myopathy and vascular anomaly in Tg.rasH2 mice; benign thymomas in Crl:WI[Han] rats; angiomatous lesions in the mesenteric lymph nodes of Crl:WI[Han] rats; an unusual foveal lesion in a cynomolgous monkey; and finally a series of nomenclatures challenges from the endocrine International Harmonization of Nomenclature and Diagnostic Criteria (INHAND) Organ Working Group (OWG).
Collapse
Affiliation(s)
- Susan A Elmore
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Michelle C Cora
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Margarita M Gruebbel
- Experimental Pathology Laboratories, Inc., Research Triangle Park, North Carolina, USA
| | - Schantel A Hayes
- Charles River Laboratories, Pathology Associates, Durham, North Carolina, USA
| | - Jessica S Hoane
- Charles River Laboratories, Pathology Associates, Durham, North Carolina, USA
| | | | - Rachel Peters
- Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| | | | - Bhanu P Singh
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Kathleen A Szabo
- Charles River Laboratories, Pathology Associates, Durham, North Carolina, USA
| |
Collapse
|
6
|
Mikaelian I, Cameron M, Dalmas DA, Enerson BE, Gonzalez RJ, Guionaud S, Hoffmann PK, King NMP, Lawton MP, Scicchitano MS, Smith HW, Thomas RA, Weaver JL, Zabka TS. Nonclinical Safety Biomarkers of Drug-induced Vascular Injury. Toxicol Pathol 2014; 42:635-57. [DOI: 10.1177/0192623314525686] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Better biomarkers are needed to identify, characterize, and/or monitor drug-induced vascular injury (DIVI) in nonclinical species and patients. The Predictive Safety Testing Consortium (PSTC), a precompetitive collaboration of pharmaceutical companies and the U.S. Food and Drug Administration (FDA), formed the Vascular Injury Working Group (VIWG) to develop and qualify translatable biomarkers of DIVI. The VIWG focused its research on acute DIVI because early detection for clinical and nonclinical safety monitoring is desirable. The VIWG developed a strategy based on the premise that biomarkers of DIVI in rat would be translatable to humans due to the morphologic similarity of vascular injury between species regardless of mechanism. The histomorphologic lexicon for DIVI in rat defines degenerative and adaptive findings of the vascular endothelium and smooth muscles, and characterizes inflammatory components. We describe the mechanisms of these changes and their associations with candidate biomarkers for which advanced analytical method validation was completed. Further development is recommended for circulating microRNAs, endothelial microparticles, and imaging techniques. Recommendations for sample collection and processing, analytical methods, and confirmation of target localization using immunohistochemistry and in situ hybridization are described. The methods described are anticipated to aid in the identification and qualification of translational biomarkers for DIVI.
Collapse
Affiliation(s)
- Igor Mikaelian
- Hoffmann-La Roche Inc, Nutley, New Jersey, USA
- Abbvie, Worcester, Massachusetts, USA
| | | | | | | | - Raymond J. Gonzalez
- Merck Research Laboratories, Merck and Co, Inc, West Point, Pennsylvania, USA
| | - Silvia Guionaud
- Shire, Hampshire International Business Park, Basingstoke, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Bendjama K, Guionaud S, Aras G, Arber N, Badimon L, Bamberger U, Bratfalean D, Brott D, David M, Doessegger L, Firat H, Gallas JF, Gautier JC, Hoffmann P, Kraus S, Padro T, Saadoun D, Szczesny P, Thomann P, Vilahur G, Lawton M, Cacoub P. Translation Strategy for the Qualification of Drug-induced Vascular Injury Biomarkers. Toxicol Pathol 2014; 42:658-71. [DOI: 10.1177/0192623314527644] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Drug-induced vascular injury (DIVI) is a common preclinical toxicity usually characterized by hemorrhage, vascular endothelial and smooth muscle damage, and inflammation. DIVI findings can cause delays or termination of drug candidates due to low safety margins. The situation is complicated by the absence of sensitive, noninvasive biomarkers for monitoring vascular injury and the uncertain relevance to humans. The Safer And Faster Evidence-based Translation (SAFE-T) consortium is a public–private partnership funded within the European Commission’s Innovative Medicines Initiative (IMI) aiming to accelerate drug development by qualifying biomarkers for drug-induced organ injuries, including DIVI. The group is using patients with vascular diseases that have key histomorphologic features (endothelial damage, smooth muscle damage, and inflammation) in common with those observed in DIVI, and has selected candidate biomarkers associated with these features. Studied populations include healthy volunteers, patients with spontaneous vasculitides and other vascular disorders. Initial results from studies with healthy volunteers and patients with vasculitides show that a panel of biomarkers can successfully discriminate the population groups. The SAFE-T group plans to seek endorsement from health authorities (European Medicines Agency and Food and Drug Administration) to qualify the biomarkers for use in regulatory decision-making processes.
Collapse
Affiliation(s)
| | | | | | - Nadir Arber
- Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Lina Badimon
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Uwe Bamberger
- Boehringer Ingelheim Pharma GmbH & Co. KG Nonclinical Drug Safety Biberach/Riss, Germany
| | | | - David Brott
- AstraZeneca Pharmaceuticals, Translational Patient Safety and Enabling Sciences, Wilmington, Delaware, USA
| | - Maayan David
- Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | | | | | | | - Peter Hoffmann
- Novartis Pharmaceuticals Corporation, Preclinical safety, East Hanover, New Jersey, USA
| | - Sarah Kraus
- Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Teresa Padro
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - David Saadoun
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France; Département Hospitalo-Universitaire I2B, UPMC Univ Paris 06, F-75005, Paris, France; INSERM UMR 7211, F-75005, Paris, France; INSERM, UMR_S 959, F-75013, Paris, France; CNRS, UMR 7211, F-75005, Paris, France
| | | | | | - Gemma Vilahur
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Michael Lawton
- Pfizer Worldwide Research & Development, Drug Safety Research & Development, Groton, Connecticut, USA
| | - Patrice Cacoub
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, F-75013, Paris, France; Département Hospitalo-Universitaire I2B, UPMC Univ Paris 06, F-75005, Paris, France; INSERM UMR 7211, F-75005, Paris, France; INSERM, UMR_S 959, F-75013, Paris, France; CNRS, UMR 7211, F-75005, Paris, France
| |
Collapse
|
8
|
Tobin GAM, Zhang J, Goodwin D, Stewart S, Xu L, Knapton A, González C, Bancos S, Zhang L, Lawton MP, Enerson BE, Weaver JL. The role of eNOS phosphorylation in causing drug-induced vascular injury. Toxicol Pathol 2014; 42:709-24. [PMID: 24705881 DOI: 10.1177/0192623314522885] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Previously we found that regulation of eNOS is an important part of the pathogenic process of Drug-induced vascular injury (DIVI) for PDE4i. The aims of the current study were to examine the phosphorylation of eNOS in mesentery versus aorta at known regulatory sites across DIVI-inducing drug classes and to compare changes across species. We found that phosphorylation at S615 in rats was elevated 35-fold 2 hr after the last dose of CI-1044 in mesentery versus 3-fold in aorta. Immunoprecipitation studies revealed that many of the upstream regulators of eNOS activation were associated with eNOS in 1 or more signalosome complexes. Next rats were treated with drugs from 4 other classes known to cause DIVI. Each drug was given alone and in combination with SIN-1 (NO donor) or L-NAME (eNOS inhibitor), and the level of eNOS phosphorylation in mesentery and aorta tissue was correlated with the extent of vascular injury and measured serum nitrite. Drugs or combinations produced altered serum nitrite levels as well as vascular injury score in the mesentery. The results suggested that phosphorylation of S615 may be associated with DIVI activity. Studies with the species-specific A2A adenosine agonist CI-947 in rats versus primates showed a similar pattern.
Collapse
Affiliation(s)
- Grainne A McMahon Tobin
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jun Zhang
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - David Goodwin
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sharron Stewart
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lin Xu
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Alan Knapton
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Carlos González
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Simona Bancos
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Leshuai Zhang
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA Department of Anatomy & Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Michael P Lawton
- Drug Safety Research and Development, Pfizer Inc, Groton, Connecticut, USA
| | - Bradley E Enerson
- Drug Safety Research and Development, Pfizer Inc, Groton, Connecticut, USA
| | - James L Weaver
- Division of Applied Regulatory Science, CDER, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
9
|
Ramot Y, Brauner R, Kang K, Heymach JV, Furtado S, Nyska A. Quantitative evaluation of drug-induced microvascular constriction in mice kidney using a novel tool for 3D geometrical analysis of ex vivo organ vasculature. Toxicol Pathol 2014; 42:774-83. [PMID: 24670814 DOI: 10.1177/0192623314525685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The analysis of organ vasculature, and more specifically organ microvasculature, carries special importance for toxicological sciences, and especially for evaluation of drug-induced vascular toxicity. This field presents a special challenge in nonclinical drug safety assessments since there are currently no reliable microvascular toxicity biomarkers. Therefore, we aimed to systematically investigate the use of microvascular 3D geometrical analysis of corrosion casts for evaluation of drug-induced vascular toxicity, utilizing a novel image investigation tool that allows full 3D-quantified geometrical analysis of the entire vascular tree structure. Vascular casts of kidneys from control and low- and high-dose ephedrine/caffeine-treated mice were scanned by a micro CT, and images were processed and analyzed using the Vasculomics™ platform. All evaluations were performed on the kidney cortex. Treatment resulted in a significant and dose-related reduction in overall microvessel density throughout the kidney cortex. This effect was most pronounced for vessels with diameters between 25 µm and 35 µm, and affected mostly vessels located in the superficial part of the kidney cortex. The use of 3D analysis tools in drug-induced vascular toxicity studies allows for very high resolution and characterization of drug effects on the microvasculature and can be used as a valuable tool in drug safety assessments.
Collapse
Affiliation(s)
- Yuval Ramot
- Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Raul Brauner
- Bio-Tree Systems Inc., Framingham, Massachusetts, USA
| | - Kongbin Kang
- Bio-Tree Systems Inc., Framingham, Massachusetts, USA
| | - John V Heymach
- Bio-Tree Systems Inc., Framingham, Massachusetts, USA Thoracic, Head and Neck Oncology Department, MD Anderson Cancer Center, Houston, Texas, USA
| | - Stacia Furtado
- Bio-Tree Systems Inc., Framingham, Massachusetts, USA Brown University, Providence, Rhode Island, USA
| | - Abraham Nyska
- Consultant in Toxicologic Pathology, Sackler School of Medicine, Tel Aviv University, Timrat, 36576, Israel
| |
Collapse
|
10
|
Heuser A, Mecklenburg L, Ockert D, Kohler M, Kemkowski J. Selective inhibition of PDE4 in Wistar rats can lead to dilatation in testis, efferent ducts, and epididymis and subsequent formation of sperm granulomas. Toxicol Pathol 2012. [PMID: 23197197 DOI: 10.1177/0192623312463783] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Testicular tubular dilatation and degeneration and epididymal sperm granulomas were frequently seen in 4-week toxicity studies using different phosphodiesterase-4 (PDE4) inhibitors in Wistar rats, including the prototypic PDE4 inhibitor BYK169171. To investigate the pathogenesis of testicular and epididymal lesions, a time course study with BYK169171 was conducted with sequential necropsies after 7, 14, 21, and 28 days of treatment. After 7 days, a dilatation of efferent ducts and of the initial segment of the epididymis and a subacute interstitial inflammation were seen followed by a diffuse dilatation of seminiferous tubules in the testis. Dilatation and inflammation were most pronounced after 14 days. Single animals also exhibited vascular necrosis in the inflamed interstitium. Although dilatation decreased later in the study, the incidence and severity of tubular degeneration increased from 14 days onward. Sperm granulomas developed in efferent ducts and in the caput and cauda of the epididymis after 14 days. Our results demonstrate a clear time course of PDE4 inhibition-induced lesions, with dilatation preceding sperm granuloma formation. We conclude that the most likely mechanism of toxicity is a disturbance of fluid homeostasis in efferent and epididymal ducts resulting in abnormal luminal fluid and sperm contents, epithelial damage at specific sites of the excurrent duct system, sperm leakage, and granuloma formation.
Collapse
Affiliation(s)
- Anke Heuser
- Institute for Pharmacology and Preclinical Drug Safety (IPAS), Nycomed GmbH (Nycomed: A Takeda Company), Barsbüttel, Germany.
| | | | | | | | | |
Collapse
|
11
|
Atkins CM, Cepero ML, Kang Y, Liebl DJ, Dietrich WD. Effects of early rolipram treatment on histopathological outcome after controlled cortical impact injury in mice. Neurosci Lett 2012; 532:1-6. [PMID: 23103712 DOI: 10.1016/j.neulet.2012.10.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/12/2012] [Accepted: 10/16/2012] [Indexed: 11/19/2022]
Abstract
Traumatic brain injury (TBI) pathology includes contusions, cavitation, cell death, all of which can be exacerbated by inflammation. We hypothesized that an anti-inflammatory drug, rolipram, may reduce pathology after TBI, since in several CNS injury models rolipram reduces inflammation and improves cell survival and functional recovery. Adult male C57BL/6 mice received a craniotomy over the right parietotemporal cortex. Vertically directed controlled cortical impact (CCI) injury was delivered. Naïve controls were used for comparison. At 30 min post-surgery, animals were treated with vehicle or rolipram (1 mg/kg), and then once per day for 3 days. On day 3, the brains were systematically sectioned and stained to visualize the resulting pathology using hematoxylin and eosin (H&E) staining and NeuN immunocytochemistry. Total parietotemporal cortical contusion and cavity volume were significantly increased in rolipram-treated as compared to vehicle-treated CCI animals. Contusion areas at specific bregma levels indicated a significant effect of drug across bregma levels. Neuronal cell loss in the dentate hilus and area CA3 of the hippocampus were similar between vehicle and rolipram-treated animals. Although rolipram is well known to reduce pathology and inflammation in several other CNS injury models, the pathology resulting from CCI was worsened with rolipram at this particular dose and administration schedule. These studies suggest that consideration of the unique characteristics of TBI pathology is important in the extrapolation of promising therapeutic interventions from other CNS injury models.
Collapse
Affiliation(s)
- Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136-1060, USA.
| | | | | | | | | |
Collapse
|
12
|
Atkins CM, Kang Y, Furones C, Truettner JS, Alonso OF, Dietrich WD. Postinjury treatment with rolipram increases hemorrhage after traumatic brain injury. J Neurosci Res 2012; 90:1861-71. [PMID: 22535545 DOI: 10.1002/jnr.23069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 03/01/2012] [Accepted: 03/22/2012] [Indexed: 11/11/2022]
Abstract
The pathology caused by traumatic brain injury (TBI) is exacerbated by the inflammatory response of the injured brain. Two proinflammatory cytokines that contribute to inflammation after TBI are tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). From previous studies using the parasagittal fluid-percussion brain injury model, we reported that the anti-inflammatory drug rolipram, a phosphodiesterase 4 inhibitor, reduced TNF-α and IL-1β levels and improved histopathological outcome when administered 30 min prior to injury. We now report that treatment with (±)-rolipram given 30 min after injury significantly reduced TNF-α levels in the cortex and hippocampus. However, postinjury administration of (±)-rolipram significantly increased cortical contusion volume and increased atrophy of the cortex compared with vehicle-treated animals at 10 days postinjury. Thus, despite the reduction in proinflammatory cytokine levels, histopathological outcome was worsened with post-TBI (±)-rolipram treatment. Further histological analysis of (±)-rolipram-treated TBI animals revealed significant hemorrhage in the contused brain. Given the well-known role of (±)-rolipram of increasing vasodilation, it is likely that (±)-rolipram worsened outcome after fluid-percussion brain injury by causing increased bleeding.
Collapse
Affiliation(s)
- C M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Potential candidate genomic biomarkers of drug induced vascular injury in the rat. Toxicol Appl Pharmacol 2011; 257:284-300. [DOI: 10.1016/j.taap.2011.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/15/2011] [Accepted: 09/16/2011] [Indexed: 01/14/2023]
|
14
|
Turteltaub KW, Davis MA, Burns-Naas LA, Lawton MP, Clark AM, Reynolds JA. Identification and Elucidation of the Biology of Adverse Events: The Challenges of Safety Assessment and Translational Medicine. Clin Cancer Res 2011; 17:6641-5. [DOI: 10.1158/1078-0432.ccr-11-1106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Zhang J, Hanig JP, De Felice AF. Biomarkers of endothelial cell activation: candidate markers for drug-induced vasculitis in patients or drug-induced vascular injury in animals. Vascul Pharmacol 2011; 56:14-25. [PMID: 21968053 DOI: 10.1016/j.vph.2011.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 08/26/2011] [Accepted: 09/12/2011] [Indexed: 01/08/2023]
Abstract
There is a pressing need for vascular biomarkers for studies of drug-induced vasculitis in patients and drug-induced vascular injury (DIVI) in animals. We previously reviewed a variety of candidate biomarkers of endothelial cell (EC) activation (Zhang et al., 2010). Now we update information on EC activation biomarkers from animal data on DIVI and clinical data of vasculitic patients, particularly patients with primary antineutrophil cytoplasmic autoantibody (ANCA)-associated small vessel vasculitis (primary AAVs), including Wegener's granulomatosis, microscopic polyangiitis, Churg-Strauss syndrome and necrotizing crescentic glomerulonephritis. Drug-associated ANCA-positive small vessel vasculitis (drug-AAVs) can closely resemble primary AAVs, suggesting the large overlap between primary idiopathic systemic vasculitis and drug-induced vasculitis. AAVs in patients and DIVI in animals vary considerably; however, there is close resemblance between AAVs and DIVI in some respects: (1) the immunopathogenetic mechanisms (activation of primed neutrophils, ECs and T cells by ANCA in patients and activation of ECs, mast cells, and macrophages by drugs in animals); (2) the morphologic changes (fibrinoid necrosis of the vessel wall and neutrophilic infiltration); (3) the preferable sites (small arteries, arterioles, capillaries and venules); and (4) elevation of vascular biomarkers suggestive of an endothelial origin. The present review discusses soluble and cell component biomarkers and provides a rationale for the potential utility of EC activation biomarkers in nonclinical and clinical studies during new drug development. Further investigation, however, is needed to assess their potential utility.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Drug Safety Research, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, MD 20993, USA.
| | | | | |
Collapse
|
16
|
Sheth CM, Enerson BE, Peters D, Lawton MP, Weaver JL. Effects of Modulating In Vivo Nitric Oxide Production on the Incidence and Severity of PDE4 Inhibitor–Induced Vascular Injury in Sprague-Dawley Rats. Toxicol Sci 2011; 122:7-15. [DOI: 10.1093/toxsci/kfr082] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
17
|
|
18
|
Zhang J, Defelice AF, Hanig JP, Colatsky T. Biomarkers of endothelial cell activation serve as potential surrogate markers for drug-induced vascular injury. Toxicol Pathol 2010; 38:856-71. [PMID: 20716788 DOI: 10.1177/0192623310378866] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Drug-induced vascular injury (DIVI) is a nonclinical finding that often confounds the toxicological evaluation of investigational drugs, but there is an absence of qualified biomarkers that can be used to detect and monitor its appearance in animals and patients during drug development and clinical use. It is well known that endothelial cell (EC) activation plays a key role in the expression and evolution of DIVI, and the various immunological and inflammatory factors involved in its expression may serve as potential biomarker candidates. Activated ECs change their morphology and gene expression, generating endothelial adhesion molecules, pro-coagulant molecules, cytokines, chemokines, vasodilators, nitric oxide, and acute-phase reactants. This review provides a brief historical background of EC activation and the search for biomarkers of early EC activation for monitoring DIVI. At present, no biomarkers of EC activation have been qualified to predict DIVI in the nonclinical or clinical context, and a robust pathologic foundation for their use is still lacking. We propose three categories of EC activation biomarkers: recommended surrogate markers, potentially useful markers, and emerging candidate markers. This review alerts pharmaceutical companies, research institutions, and regulatory agencies to the continuing need for reliable biomarkers of EC activation in drug development.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| | | | | | | |
Collapse
|
19
|
Chapman RW, House A, Richard J, Prelusky D, Lamca J, Wang P, Lundell D, Wu P, Ting PC, Lee JF, Aslanian R, Phillips JE. Pharmacology of a potent and selective inhibitor of PDE4 for inhaled administration. Eur J Pharmacol 2010; 643:274-81. [PMID: 20621091 DOI: 10.1016/j.ejphar.2010.06.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 06/07/2010] [Accepted: 06/24/2010] [Indexed: 12/16/2022]
Abstract
A strategy to overcome the side effect liabilities of oral PDE4 inhibitors has been to deliver the drugs by inhalation. In this report, we identify 1-[[5-(1(S)-aminoethly)-2-[8-methoxy-2-(triflurormethyl)-5-quinolinyl]-4-oxazolyl] carbonyl]-4(R)-[(cyclopropylcarbonyl)amino]-L-proline, ethyl ester xinafoate salt, (COMPOUND 1) as a potent and selective inhibitor of PDE4 with biological and pharmacokinetic properties suitable for delivery by the inhaled route. COMPOUND 1 potently inhibits human PDE4 (IC(50)=70pM) with little or no activity against other PDEs. It is highly potent against PDE4B and PDE4D which are important isoforms of PDE4 controlling inflammation and airway functions. In an allergen-challenged Brown Norway rat model of asthma, COMPOUND 1 inhibited the late phase influx of inflammatory cells and reductions in lung function following its administration by the intratracheal or nose-only routes of administration. Important differences were seen between intratracheal COMPOUND 1 and our previously published results with the oral PDE4 inhibitor roflumilast (Celly et al., 2005), as COMPOUND 1 rapidly (within 1h) reversed the decline in lung function when it was given therapeutically to rats already challenged with antigen. COMPOUND 1 was weakly active by the oral route which is a finding consistent with results showing this compound has poor oral bioavailability in animals. Positive interactions between COMPOUND 1 and albuterol, and COMPOUND 1 and mometasone furoate were seen on the improvement in lung functions in allergen-challenged rats. These results identify COMPOUND 1 as a potent and selective inhibitor of PDE4 with properties suitable for delivery by inhalation.
Collapse
Affiliation(s)
- Richard W Chapman
- Department of Bone, Respiratory, Immunology and Dermatology, Merck Research Laboratories, Kenilworth, New Jersey 07033, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Weaver JL, Zhang J, Knapton A, Miller T, Espandiari P, Smith R, Gu YZ, Snyder RD. Early events in vascular injury in the rat induced by the phosphodiesterase IV inhibitor SCH 351591. Toxicol Pathol 2010; 38:738-44. [PMID: 20585143 DOI: 10.1177/0192623310374331] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment with drugs from multiple classes induces vascular injury with medial necrosis, hemorrhage, endothelial damage, and inflammation. Previous research has suggested early events might be occurring well in advance of the full lesions that appear forty-eight to seventy-two hours after dosing with SCH 351591, a PDE IV inhibitor. This study was performed to study early events in detail. Rats were dosed with 20 mg/kg of drug by gavage and sacrificed at times between fifteen and 240 minutes after dosing. Tissues were collected for histopathological analysis and gene expression studies. Serum was collected for biomarker analysis. The data from biomarker analysis showed a three-part response with an early phase that was maximal at fifteen to thirty minutes, a second phase from forty-five to 180 minutes, and the third phase that was starting to rise at four hours. The first phase included increases in lymphocytes, serum histamine, and serum nitrite. The second phase shows continued elevation of serum nitrite. The third phase was marked by an increase in serum GRO/CINC-1. At fifteen minutes, histopathology showed activation of mast cells, but not degranulation. Increases in endothelial activation and perivascular inflammatory cells were first apparent at thirty minutes and increased through 240 minutes.
Collapse
Affiliation(s)
- James L Weaver
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Losco PE, Poulet FM, Kaminska-McNamara GZ, Klein MF. Myocardial and Reproductive System Toxicity of SCH 351591, a Selective Phosphodiesterase-4 Inhibitor, in CD-1 Mice. Toxicol Pathol 2010; 38:568-82. [DOI: 10.1177/0192623310370463] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This report describes the findings of preclinical testing of SCH 351591, a selective phosphodiesterase 4 inhibitor, in CD-1 mice over a wide range of doses, in which the heart and reproductive organs of both sexes demonstrated toxic effects. Repeat-dose toxicity studies assessed 5, 15, 50, 100, 200, 400, and 800 mg/kg/day, orally by gavage, for one or three months. Findings included higher testes and ovary weights and lower uterus weights (≥200 mg/kg), small ovaries/uterus (≥400 mg/kg), and histopathologic changes of large corpora lutea and ovarian atrophy at 200 and 800 mg/kg, respectively. In addition, chronic myocardial inflammation of the heart base occurred at 100 mg/kg. Vaginal staging of the estrous cycle revealed persistent diestrus. There was no histopathologic correlate or morphometric change to explain higher testes weights. A pilot fertility and early embryonic developmental toxicity study assessing doses of 100, 200, 400, and 800 mg/kg/day produced complementary results. Females had prolonged or abnormal estrous cycles, fewer successful pregnancies, increased ovarian corpora lutea, and decreased size of live litters owing to fetal resorptions. Male fertility was not affected. However, males had a 25% increase in testes weights at all doses. The pharmacology of specific PDE4 isoenzymes may explain both the reproductive and cardiac findings.
Collapse
Affiliation(s)
| | | | | | - Max F. Klein
- Schering Plough Research Institute, Lafayette, New Jersey, USA
| |
Collapse
|
22
|
Design of phosphodiesterase 4D (PDE4D) allosteric modulators for enhancing cognition with improved safety. Nat Biotechnol 2010; 28:63-70. [DOI: 10.1038/nbt.1598] [Citation(s) in RCA: 301] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 12/04/2009] [Indexed: 12/25/2022]
|
23
|
Weaver JL, Snyder R, Knapton A, Herman EH, Honchel R, Miller T, Espandiari P, Smith R, Gu YZ, Goodsaid FM, Rosenblum IY, Sistare FD, Zhang J, Hanig J. Biomarkers in Peripheral Blood Associated with Vascular Injury in Sprague-Dawley Rats Treated with the Phosphodiesterase IV Inhibitors SCH 351591 or SCH 534385. Toxicol Pathol 2008; 36:840-9. [DOI: 10.1177/0192623308322310] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Drug-associated vascular injury can be caused by phosphodiesterase (PDE) IV inhibitors and drugs from several other classes. The pathogenesis is poorly understood, but it appears to include vascular and innate immunological components. This research was undertaken to identify changes in peripheral blood associated with vascular injury caused by PDE IV inhibitors. We evaluated twelve proteins, serum nitrite, and leukocyte populations in peripheral blood of rats treated with experimental PDE IV inhibitors. We found that these compounds produced histological microvascular injury in a dose- and time-dependent manner. Measurement of these serum proteins showed changes in eight of the twelve examined. Changes were seen in the levels of: tissue inhibitor of metalloproteinase-1, α1-acid glycoprotein, GRO/CINC-1, vascular endothelial growth factor, C-reactive protein, haptoglobin, thrombomodulin, and interleukin-6. No changes were seen in levels of tumor necrosis factor-α, hepatocyte growth factor, nerve growth factor, and granulocyte-monocyte colony stimulating factor. Serum levels of nitrite were also increased. Circulating granulocyte numbers were increased, and lymphocyte numbers were decreased. The changes in these parameters showed both a dose- and time-dependent association with histopathologic changes. These biomarkers could provide an additional tool for the nonclinical and clinical evaluation of investigational compounds.
Collapse
Affiliation(s)
- James L. Weaver
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ronald Snyder
- Schering-Plough Research Institute, Summit, New Jersey, USA
| | - Alan Knapton
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Eugene H. Herman
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ronald Honchel
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Terry Miller
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Parvaneh Espandiari
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Roger Smith
- Schering-Plough Research Institute, Summit, New Jersey, USA
| | - Yi-Zhong Gu
- Schering-Plough Research Institute, Summit, New Jersey, USA
| | - Federico M. Goodsaid
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | | | | | - Jun Zhang
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Joseph Hanig
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|