Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Toxicol Pathol ; 52(2-3): 138-148, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38840532

ABSTRACT

In December 2021, the United States Food and Drug Administration (FDA) issued the final guidance for industry titled Pathology Peer Review in Nonclinical Toxicology Studies: Questions and Answers. The stated purpose of the FDA guidance is to provide information to sponsors, applicants, and nonclinical laboratory personnel regarding the management and conduct of histopathology peer review as part of nonclinical toxicology studies conducted in compliance with good laboratory practice (GLP) regulations. On behalf of and in collaboration with global societies of toxicologic pathology and the Society of Quality Assurance, the Scientific and Regulatory Policy Committee (SRPC) of the Society of Toxicologic Pathology (STP) initiated a review of this FDA guidance. The STP has previously published multiple papers related to the scientific conduct of a pathology peer review of nonclinical toxicology studies and appropriate documentation practices. The objectives of this review are to provide an in-depth analysis and summary interpretation of the FDA recommendations and share considerations for the conduct of pathology peer review in nonclinical toxicology studies that claim compliance to GLP regulations. In general, this working group is in agreement with the recommendations from the FDA guidance that has added clear expectations for pathology peer review preparation, conduct, and documentation.


Subject(s)
Pathology , Peer Review , Toxicology , United States Food and Drug Administration , United States , Toxicology/standards , Toxicology/legislation & jurisprudence , Toxicology/methods , Peer Review/standards , Pathology/standards , Guidelines as Topic , Animals , Toxicity Tests/standards , Toxicity Tests/methods
2.
Front Toxicol ; 6: 1377990, 2024.
Article in English | MEDLINE | ID: mdl-38845817

ABSTRACT

Industry representatives on the ICH S1B(R1) Expert Working Group (EWG) worked closely with colleagues from the Drug Regulatory Authorities to develop an addendum to the ICH S1B guideline on carcinogenicity studies that allows for a weight-of-evidence (WoE) carcinogenicity assessment in some cases, rather than conducting a 2-year rat carcinogenicity study. A subgroup of the EWG composed of regulators have published in this issue a detailed analysis of the Prospective Evaluation Study (PES) conducted under the auspices of the ICH S1B(R1) EWG. Based on the experience gained through the Prospective Evaluation Study (PES) process, industry members of the EWG have prepared the following commentary to aid sponsors in assessing the standard WoE factors, considering how novel investigative approaches may be used to support a WoE assessment, and preparing appropriate documentation of the WoE assessment for presentation to regulatory authorities. The commentary also reviews some of the implementation challenges sponsors must consider in developing a carcinogenicity assessment strategy. Finally, case examples drawn from previously marketed products are provided as a supplement to this commentary to provide additional examples of how WoE criteria may be applied. The information and opinions expressed in this commentary are aimed at increasing the quality of WoE assessments to ensure the successful implementation of this approach.

3.
Int J Toxicol ; 41(6): 431-441, 2022 12.
Article in English | MEDLINE | ID: mdl-36112859

ABSTRACT

The objective of the pharmaceutical industry is to develop new drugs that are safe for human use. In many cases, the accepted approach codified in guidance from regulatory authorities to assess the nonclinical safety profile of potential pharmaceuticals is to perform toxicity testing in two species. However, the use of a second species to establish the safety of new pharmaceuticals has been the subject of much scrutiny in recent years and the industry has been repeatedly challenged to reduce, refine, or replace some or all of the animals used to establish the safety of these pharmaceutical candidates. Specifically, the value of the dog in this testing paradigm has been questioned. Publications reviewing available data for marketed drugs suggest that for many drugs, the dog does not identify unique toxicities critical to human safety. The weakness of this approach, however, is that many of the cases where the dog (or any other species) has the greatest impact on drug development are cases for which development decisions based on safety concerns are not shared publicly. The European Federation of Pharmaceutical Industries and Associations (EFPIA) Preclinical Development Expert Group (PDEG) decided to share case studies collected from its membership and the literature to illustrate the value of the dog in drug development decision-making and clinical monitoring practices to protect the safety of trial subjects.


Subject(s)
Drug Industry , Toxicity Tests , Animals , Dogs , Drug Development , Drug Evaluation, Preclinical , Humans , Pharmaceutical Preparations
4.
J Toxicol Pathol ; 34(3 Suppl): 1S-182S, 2021.
Article in English | MEDLINE | ID: mdl-34712008

ABSTRACT

The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions Project (www.toxpath.org/inhand.asp) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP) and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in most tissues and organs from the nonhuman primate used in nonclinical safety studies. Some of the lesions are illustrated by color photomicrographs. The standardized nomenclature presented in this document is also available electronically on the internet (http://www.goreni.org/). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. Relevant infectious and parasitic lesions are included as well. A widely accepted and utilized international harmonization of nomenclature for lesions in laboratory animals will provide a common language among regulatory and scientific research organizations in different countries and increase and enrich international exchanges of information among toxicologists and pathologists.

5.
Toxicol Pathol ; 49(2): 232-234, 2021 02.
Article in English | MEDLINE | ID: mdl-33397211

ABSTRACT

The delivery of biotherapeutic molecules (antibodies, proteins, peptides) and nucleic acids via the respiratory route has presented challenges for regulatory approval, due in part to a lack of understanding of the expected pathology, mechanisms of toxicity, and immunogenicity induced by the inhalation route. Although the first inhaled biotherapeutic was approved some time ago (Dornase Alfa, Pulmozyme; Genetech, 1993), no other inhaled biotherapeutics have been marketed for the treatment of human disease other than the inhaled insulins (Exubera; Pfizer, 2006 and Afrezza; Mannkind Corporation, 2014). As a result, scientific knowledge within the toxicologic pathology community is fragmented with precious little publicly available data. Therefore, one of the aims of this special edition was to generate a collection of manuscripts that pathologists and toxicologists could refer in order to understand the pathology, mechanisms of toxicity, immunogenicity, and challenges associated with the development of inhaled biotherapeutics.


Subject(s)
Biological Products , Administration, Inhalation , Biological Products/administration & dosage , Humans
6.
Toxicol Pathol ; 46(8): 1013-1019, 2018 12.
Article in English | MEDLINE | ID: mdl-30157708

ABSTRACT

This article summarizes a continuing education presentation on immunogenicity that was part of a continuing education course entitled, "Clinical Pathology of Biotherapeutics." Immunogenicity of a biotherapeutic can have diverse impacts including altered systemic exposure and pharmacologic responses and, in a fraction of the cases, safety concerns including cross-reactive neutralization of endogenous proteins or sequela related to immune complex disease (ICD). In most cases, immune complexes are readily cleared from circulation; however, based on physiochemical properties, insoluble complexes form, activate complement, and deposit in tissues. Using published information and personal experience, a set of repeat-dose monkey toxicity studies with manifestations suggestive of ICD was reviewed to summarize the spectrum of clinical and pathology findings. The most common live-phase observation linked to ICD was an acute postdosing reaction following multiple dose administrations characterized by generalized collapse and attributed to acute complement activation. Less common live-phase observations were related to syndromes such as a consumptive coagulopathy or a protein losing nephropathy. The most common histologic change attributed to ICD was multi-organ vascular/perivascular inflammation followed by glomerulonephritis. The presentation concluded with a description of the challenges in assessing the relevance of immunogenicity-related reaction in monkey to human clinical use.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/immunology , Immune Complex Diseases/chemically induced , Immune Complex Diseases/immunology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Toxicology
7.
Regul Toxicol Pharmacol ; 96: 18-29, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29679677

ABSTRACT

Toxicogenomics held great promise as an approach to enable early detection of toxicities induced by xenobiotics; however, there remain questions regarding the impact of the discipline on pharmaceutical nonclinical safety assessment. To understand the current state of toxicogenomics in the sector, an industry group surveyed companies to determine the frequency of toxicogenomics use in in vivo studies at various stages of drug discovery and development and to assess how toxicogenomics use has evolved over time. Survey data were compiled during 2016 from thirteen pharmaceutical companies. Toxicogenomic analyses were infrequently conducted in the development phase and when performed were done to address specific mechanistic questions. Prior to development, toxicogenomics use was more frequent; however, there were significant differences in approaches among companies. Across all phases, gaining mechanistic insight was the most frequent reason cited for pursing toxicogenomics with few companies using toxicogenomics to predict toxicities. These data were consistent with the commentary submitted in response to survey questions asking companies to describe the evolution of their toxicogenomics strategy. Overall, these survey data indicate that toxicogenomics is not widely used as a predictive tool in the pharmaceutical industry but is used regularly by some companies and serves a broader role in mechanistic investigations and as a complement to other technologies.


Subject(s)
Drug Evaluation, Preclinical/adverse effects , Drug Industry , Toxicogenetics , Animals , Humans
8.
Toxicol Pathol ; 46(4): 460-472, 2018 06.
Article in English | MEDLINE | ID: mdl-29699458

ABSTRACT

Inhibitors of Bruton's tyrosine kinase (BTK) are under development as potential therapies for various autoimmune diseases. In repeat-dose toxicity studies, small-molecule BTK inhibitors (BTKi) have been reported to cause a constellation of histologic effects at the pancreatic endocrine-exocrine interface in male rats; however, similar findings were not reported in other species. Since the BTKi-induced pancreatic effect is morphologically similar to well-documented spontaneous changes (predominantly characterized by insular/peri-insular hemorrhage, pigment deposition, chronic inflammation, and fibrosis) that are known to vary by rat strain, we investigated potential strain-dependent differences in the pancreatic effects of a small-molecule BTKi, LY3337641. Following 13 weeks of LY3337641 treatment, Crl:CD(SD) rats were most sensitive, Crl:WI(Han) rats were of intermediate sensitivity, and Hsd:SD rats were least sensitive. These strain differences appear to be related to differences in rate of weight gain across strains and sexes; however, a definitive mechanism was not determined. This study demonstrated that BTKi-induced pancreatic effects were highly dependent on rat strain and correlated with differences in the incidence and severity of the spontaneous background change. When considered with the lack of pancreas effects in nonrat species, these changes in rats are unlikely predictive of similar changes in humans administered a BTK inhibitor.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Pancreas/drug effects , Protein Kinase Inhibitors/toxicity , Animals , Rats , Rats, Sprague-Dawley , Rats, Wistar
9.
Toxicol Pathol ; 45(7): 845-850, 2017 10.
Article in English | MEDLINE | ID: mdl-28969491

ABSTRACT

In animal studies, light microscopic examination remains an important tool in the detection and characterization of effects on the skeleton. In the case of both anticipated and unanticipated effects on bone histology, pathologists must carefully select terminology that accurately conveys skeletal morphology without overstating what information can be derived from a standard decalcified paraffin-embedded section. The International Harmonization of Nomenclature and Diagnostic Criteria (INHAND) project issued standardized nomenclature for toxicologic pathologists to use with respect to the skeleton. Consistent with general INHAND principles, the nomenclature emphasizes a descriptive approach to classifying lesions, rather than using terms that imply a specific pathogenesis or disease process. This article, which is based on a presentation at the Society of Toxicologic Pathology's 36th Annual Symposium, will discuss the role of anatomic pathology evaluation in the context of skeletal evaluation in toxicity studies. The integration of anatomic pathology data with quantitative bone end points is reviewed. An overview of the INHAND diagnostic scheme is provided and accompanied by case examples in which the INHAND terminology has been applied. In addition, this review summarizes key considerations for toxicologists and pathologists assigning adversity designations when considering bone end points.


Subject(s)
Bone Diseases/diagnosis , Bone and Bones/pathology , Pathology , Toxicology , Animals , Endpoint Determination , Histological Techniques , Humans , Microscopy , Terminology as Topic
10.
Regul Toxicol Pharmacol ; 88: 56-65, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28526658

ABSTRACT

Basaglar®/Abasaglar® (Lilly insulin glargine [LY IGlar]) is a long-acting human insulin analogue drug product granted marketing authorisation as a biosimilar to Lantus® (Sanofi insulin glargine [SA IGlar]) by the European Medicines Agency. We assessed the similarity of LY IGlar to the reference drug product, European Union-sourced SA IGlar (EU-SA IGlar), using nonclinical in vitro and in vivo studies. No biologically relevant differences were observed for receptor binding affinity at either the insulin or insulin-like growth factor-1 (IGF-1) receptors, or in assays of functional or de novo lipogenic activity. The mitogenic potential of LY IGlar and EU-SA IGlar was similar when tested in both insulin- and IGF-1 receptor dominant cell systems. Repeated subcutaneous daily dosing of rats for 4 weeks with 0, 0.3, 1.0, or 2.0 mg/kg LY IGlar and EU-SA IGlar produced mortalities and clinical signs consistent with severe hypoglycaemia. Glucodynamic profiles of LY IGlar and EU-SA IGlar in satellite animals showed comparable dose-related hypoglycaemia. Severe hypoglycaemia was associated with axonal degeneration of the sciatic nerve; the incidence and severity were low and did not differ between LY IGlar and EU-SA IGlar. These results demonstrated no biologically relevant differences in toxicity between LY IGlar and EU-SA IGlar.


Subject(s)
Biosimilar Pharmaceuticals/toxicity , Hypoglycemic Agents/toxicity , Insulin Glargine/toxicity , Animals , Biosimilar Pharmaceuticals/metabolism , Drug Approval , European Union , Humans , Hypoglycemia/chemically induced , Hypoglycemic Agents/metabolism , In Vitro Techniques , Insulin Glargine/metabolism , Rats , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism
11.
Toxicol Pathol ; 45(3): 402-415, 2017 04.
Article in English | MEDLINE | ID: mdl-28421968

ABSTRACT

Basal insulin peglispro (BIL) consists of insulin lispro with a 20-kDa polyethylene glycol (PEG) moiety covalently attached to lysine B28. Because chronic parenteral administration of PEGylated proteins to animals has sometimes resulted in PEG vacuolation of tissue macrophages, renal tubular cells, and choroid plexus ependymal cells, we investigated whether chronic subcutaneous (sc) injection of BIL in rats (52 weeks) and dogs (39 weeks) was associated with systemic toxicities or other changes, including vacuolation of tissue macrophages, renal tubular cells, and ependymal cells. Rats and dogs received daily sc injections of BIL (rats: 0.17, 0.45, or 1.15 mg/kg/d and dogs: 0.025, 0.10, or 0.20 mg/kg/d) and the reference compound, HUMULIN N® (neutral protamine Hagedorn [NPH] human insulin; rats: 0.15 mg/kg/d and dogs: 0.02-0.03 mg/kg/d). Animals were evaluated for standard end points including mortality, clinical signs, body weights, toxicokinetics, glucodynamics, clinical pathology, and morphological pathology. Nonadverse injection site lipohypertrophy occurred for all BIL and NPH doses but more frequently with BIL. No BIL-related hyperplasia or neoplasia was observed. There was no vacuolation of tissue macrophages, renal tubular cells, or ependymal cells attributable to PEG. These studies demonstrate BIL is not associated with tissue vacuolation attributable to PEG at 4- to 6-fold multiple of the median clinical exposure in patients with diabetes.


Subject(s)
Hypoglycemic Agents/toxicity , Insulin Lispro/analogs & derivatives , Polyethylene Glycols/toxicity , Animals , Body Weight/drug effects , Delayed-Action Preparations , Dogs , Dose-Response Relationship, Drug , Eating/drug effects , Ependyma/drug effects , Ependyma/pathology , Female , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Injections, Subcutaneous , Insulin Lispro/administration & dosage , Insulin Lispro/pharmacokinetics , Insulin Lispro/toxicity , Kidney Tubules/drug effects , Kidney Tubules/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Organ Specificity , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Rats, Sprague-Dawley , Species Specificity , Survival Analysis , Toxicity Tests, Chronic , Toxicokinetics
12.
Toxicol Pathol ; 43(8): 1093-102, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26269615

ABSTRACT

Glucagon-like peptide-1 (GLP-1) receptor agonist therapy has been implicated as a possible risk factor for acute pancreatitis in patients with type 2 diabetes. Dulaglutide is a long-acting GLP-1 receptor agonist in development for treatment of type 2 diabetes. The effects of dulaglutide were evaluated in male Zucker diabetic fatty (ZDF) rats to examine whether dulaglutide may induce or modulate pancreatitis. Rats were randomized to dose groups receiving twice-weekly subcutaneously administered dulaglutide 0.5, 1.5, and 5.0 mg/kg/dose (corresponding human plasma exposures following twice-weekly dosing are 3-, 8-, and 30-fold, respectively) for 13 weeks or to vehicle control. Following termination, serially trimmed sections of pancreases were stained with hematoxylin and eosin or co-stained with an epithelial marker and a marker of either proliferation or apoptosis. Efficacious reductions in glucose and hemoglobin A1c occurred at all dulaglutide doses. Lipase activity was unaffected, and there were modest increases in total and pancreatic amylase activities at all doses without individual microscopic inflammatory correlates. Microscopic dulaglutide-related pancreatic changes included increased interlobular ductal epithelium without ductal cell proliferation (≥0.5 mg/kg), increased acinar atrophy with/without inflammation (≥1.5 mg/kg), and increased incidence/severity of neutrophilic acinar pancreatic inflammation (5.0 mg/kg). In summary, dulaglutide treatment was associated with mild alterations in ductal epithelium and modest exacerbation of spontaneous lesions of the exocrine pancreas typically found in the ZDF rat model.


Subject(s)
Biomarkers/analysis , Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptides/analogs & derivatives , Hypoglycemic Agents/administration & dosage , Immunoglobulin Fc Fragments/administration & dosage , Pancreas/metabolism , Recombinant Fusion Proteins/administration & dosage , Animals , Blood Glucose/analysis , Blood Glucose/drug effects , Body Weight/drug effects , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/pharmacokinetics , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Injections, Subcutaneous , Male , Pancreas/pathology , Rats , Rats, Zucker , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
13.
Toxicol Pathol ; 43(7): 1004-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26059826

ABSTRACT

Clinical and nonclinical studies have implicated glucagon-like peptide-1 (GLP-1) receptor agonist therapy as a risk factor for acute pancreatitis in patients with type 2 diabetes. Therefore, it is critical to understand the effect that dulaglutide, an approved GLP-1 receptor agonist, has on the exocrine pancreas. Dulaglutide 8.15 mg/kg (approximately 500 times the maximum recommended human dose based on plasma exposure) was administered twice weekly for 12 months to cynomolgus monkeys. Serum amylase and lipase activities were measured and 6 sections of each pancreas were examined microscopically. Ductal epithelial cell proliferation was estimated using Ki67 labeling. Dulaglutide administration did not alter serum amylase or lipase activities measured at the end of treatment compared to control values. An extensive histologic evaluation of the pancreas revealed no changes in the acinar or endocrine portions and no evidence of pancreatitis, necrosis, or pancreatic intraepithelial neoplasia. An increase in goblet cells noted in 4 of the 19 treated monkeys was considered an effect of dulaglutide but was not associated with dilation, blockage, or accumulation of mucin in the pancreatic duct. There was no difference in cell proliferation in ductal epithelium between control and dulaglutide-treated monkeys. These data reveal that chronic dosing of nondiabetic primates with dulaglutide does not induce inflammatory or preneoplastic changes in exocrine pancreas.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptides/analogs & derivatives , Hypoglycemic Agents/toxicity , Immunoglobulin Fc Fragments/toxicity , Pancreas, Exocrine/drug effects , Recombinant Fusion Proteins/toxicity , Animals , Glucagon-Like Peptides/toxicity , Macaca fascicularis , Male , Pancreas, Exocrine/pathology
14.
Endocrinology ; 156(7): 2409-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25860028

ABSTRACT

Glucagon-like peptide-1 (GLP-1) receptor agonists, used for the treatment of type 2 diabetes, have caused hyperplasia/neoplasia of thyroid C cells in rodent carcinogenicity studies. Studies in monkeys have not identified an effect of GLP-1 receptor agonists on thyroid C cells; however, group sizes were small. Dulaglutide is a once-weekly, long-acting human GLP-1 receptor agonist recently approved in the United States and the European Union. The objective of this study was to determine whether dulaglutide altered C-cell mass in monkeys. Male cynomolgus monkeys (20 per group) were sc injected with dulaglutide 8.15 mg/kg (∼500-fold maximum human plasma exposure) or a vehicle control twice weekly for 52 weeks. Basal and calcium gluconate-stimulated serum calcitonin concentrations were obtained at 3, 6, 9, and 12 months. Thyroid glands were weighed, fixed, and sectioned at 500-µm intervals. C-cell volumes were measured using an automated image analysis. C-cell proliferation was estimated using Ki67/calcitonin colabeling and cell counting. Administration of dulaglutide 8.15 mg/kg twice weekly for 52 weeks did not increase serum calcitonin in monkeys or affect thyroid weight, histology, C-cell proliferation, or absolute/relative C-cell volume. This study represents a comprehensive evaluation of the monkey thyroid C cells after dosing with a GLP-1 receptor agonist, with a large group size, and measurement of multiple relevant parameters. The lack of effect of dulaglutide on C cells is consistent with other studies in monkeys using GLP-1 receptor agonists and suggests that nonhuman primates are less sensitive than rodents to the induction of proliferative changes in thyroid C cells by GLP-1 receptor agonists.


Subject(s)
Calcitonin/drug effects , Cell Proliferation/drug effects , Glucagon-Like Peptides/analogs & derivatives , Hypoglycemic Agents/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , Thyroid Gland/drug effects , Animals , Calcitonin/blood , Calcium Gluconate/pharmacology , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptides/pharmacology , Macaca fascicularis , Male , Organ Size/drug effects , Receptors, Glucagon/agonists , Thyroid Gland/metabolism , Thyroid Gland/pathology
15.
Endocrinology ; 156(7): 2417-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25860029

ABSTRACT

The tumorigenic potential of dulaglutide was evaluated in rats and transgenic mice. Rats were injected sc twice weekly for 93 weeks with dulaglutide 0, 0.05, 0.5, 1.5, or 5 mg/kg corresponding to 0, 0.5, 7, 20, and 58 times, respectively, the maximum recommended human dose based on plasma area under the curve. Transgenic mice were dosed sc twice weekly with dulaglutide 0, 0.3, 1, or 3 mg/kg for 26 weeks. Dulaglutide effects were limited to the thyroid C-cells. In rats, diffuse C-cell hyperplasia and adenomas were statistically increased at 0.5 mg/kg or greater (P ≤ .01 at 5 mg/kg), and C-cell carcinomas were numerically increased at 5 mg/kg. Focal C-cell hyperplasia was higher compared with controls in females given 0.5, 1.5, and 5 mg/kg. In transgenic mice, no dulaglutide-related C-cell hyperplasia or neoplasia was observed at any dose; however, minimal cytoplasmic hypertrophy of C cells was observed in all dulaglutide groups. Systemic exposures decreased over time in mice, possibly due to an antidrug antibody response. In a 52-week study designed to quantitate C-cell mass and plasma calcitonin responses, rats received twice-weekly sc injections of dulaglutide 0 or 5 mg/kg. Dulaglutide increased focal C-cell hyperplasia; however, quantitative increases in C-cell mass did not occur. Consistent with the lack of morphometric changes in C-cell mass, dulaglutide did not affect the incidence of diffuse C-cell hyperplasia or basal or calcium-stimulated plasma calcitonin, suggesting that diffuse increases in C-cell mass did not occur during the initial 52 weeks of the rat carcinogenicity study.


Subject(s)
Glucagon-Like Peptides/analogs & derivatives , Hypoglycemic Agents/toxicity , Immunoglobulin Fc Fragments/toxicity , Recombinant Fusion Proteins/toxicity , Thyroid Gland/drug effects , Thyroid Neoplasms/chemically induced , Animals , Calcitonin/blood , Calcitonin/drug effects , Carcinogenicity Tests , Carcinoma, Neuroendocrine , Female , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptides/toxicity , Hyperplasia , Male , Mice , Mice, Transgenic , Organ Size , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Receptors, Glucagon/agonists , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroid Neoplasms/pathology
17.
Toxicol Sci ; 125(2): 586-94, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22106037

ABSTRACT

LY2541546 is a humanized monoclonal antibody (IgG(4)) that has been optimized for neutralizing activity against sclerostin. In 5-week and 6-month nonclinical safety studies in rats, LY2541546 caused dose-dependent reversible decreases in platelet counts accompanied by accelerated platelet production, increased megakaryocytes, and altered megakaryocyte morphology. These treatment-related effects resulted in altered primary hemostasis as manifested by prolonged bleeding after phlebotomy or incidental toenail break. In some cases, the defects in hemostasis were sufficient to result in death of the affected rats. There was no evidence in rats of general bone marrow suppression or processes (e.g., disseminated intravascular coagulopathy) that may result in thrombocytopenia. Cynomolgus monkeys given LY2541546 for 5 weeks or 9 months had no changes in platelet count or megakaryocytes. In vitro cross-reactivity studies in rats, cynomolgus monkeys, and humans revealed LY2541546-bound rat but not cynomolgus monkey or human platelets and megakaryocytes. These data taken together demonstrated that the platelet and megakaryocyte effects in rats had a species-specific pathogenesis which likely involved LY2541546 binding of a rat-specific antigen on the surface of platelets and megakaryocytes resulting in the increased clearance of platelets and megakaryocyte hyperplasia. The species-specific nature of these reversible toxicological findings combined with the ease of clinical monitoring using standard hematology enabled the safe initiation of clinical studies in human volunteers.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Blood Platelets/drug effects , Bone Morphogenetic Proteins/immunology , Megakaryocytes/drug effects , Thrombocytopenia/chemically induced , Animals , Antibody Specificity , Blood Platelets/pathology , Cross Reactions , Dose-Response Relationship, Drug , Female , Hemostasis/drug effects , Humans , Hyperostosis/chemically induced , Macaca fascicularis , Male , Megakaryocytes/pathology , Platelet Count , Rats , Rats, Sprague-Dawley , Species Specificity , Thrombocytopenia/blood , Thrombocytopenia/pathology
18.
Toxicol Pathol ; 39(2): 429-34, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21297067

ABSTRACT

The Society of Toxicologic Pathology (STP) and American Society for Veterinary Clinical Pathology (ASCVP) convened a Clinical Pathology in Carcinogenicity Studies Working Group to recommend best practices for inclusion of clinical pathology testing in carcinogenicity studies. Regulatory guidance documents and literature were reviewed, and veterinary pathologists from North America, Japan, and Europe were surveyed regarding current practices, perceived value, and recommendations for clinical pathology testing in carcinogenicity studies. For two-year rodent carcinogenicity studies, the Working Group recommends that clinical pathology testing be limited to collection of blood smears at scheduled and unscheduled sacrifices to be examined only if indicated to aid in the diagnosis of possible hematopoietic neoplasia following histopathologic evaluation. Additional clinical pathology testing is most appropriately used to address specific issues from prior toxicity studies or known test article-related class effects. Inadequate data were available to make a recommendation concerning clinical pathology testing for alternative six-month carcinogenicity assays using genetically modified mice, although the Working Group suggests that it may be appropriate to use the same approach as for two-year carcinogenicity studies since the study goal is the same.


Subject(s)
Neoplasms/pathology , Pathology, Clinical/standards , Animals , Benchmarking , Carcinogenicity Tests , Drug-Related Side Effects and Adverse Reactions , Europe , Guidelines as Topic , Humans , Japan , Mice , Mice, Transgenic , North America
19.
Toxicol Pathol ; 38(4): 522-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20472697

ABSTRACT

An important safety consideration for developing new therapeutics is assessing the potential that the therapy will increase the risk of cancer. For biotherapeutics, traditional two-year rodent bioassays are often not scientifically applicable or feasible. This paper is a collaborative effort of industry toxicologists to review past and current practice regarding carcinogenicity assessments of biotherapeutics and to provide recommendations. Publicly available information on eighty marketed protein biotherapeutics was reviewed. In this review, no assessments related to carcinogenicity or tumor growth promotion were identified for fifty-one of the eighty molecules. For the twenty-nine biotherapeutics in which assessments related to carcinogenicity were identified, various experimental approaches were employed. This review also discusses several key principles to aid in the assessment of carcinogenic potential, including (1) careful consideration of mechanism of action to identify theoretical risks, (2) careful investigation of existing data for indications of proliferative or immunosuppressive potential, and (3) characterization of any proliferative or immunosuppressive signals detected. Traditional two-year carcinogenicity assays should not be considered as the default method for assessing the carcinogenicity potential of biotherapeutics. If experimentation is considered warranted, it should be hypothesis driven and may include a variety of experimental models. Ultimately, it is important that preclinical data provide useful guidance in product labeling.


Subject(s)
Biopharmaceutics/methods , Biotechnology/methods , Carcinogenicity Tests/methods , Drug Approval/methods , Animals , Drug-Related Side Effects and Adverse Reactions , Humans
SELECTION OF CITATIONS
SEARCH DETAIL