Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Cancer Ther ; 20(10): 1941-1955, 2021 10.
Article in English | MEDLINE | ID: mdl-34253590

ABSTRACT

B-cell maturation antigen (BCMA) is an attractive therapeutic target highly expressed on differentiated plasma cells in multiple myeloma and other B-cell malignancies. GSK2857916 (belantamab mafodotin, BLENREP) is a BCMA-targeting antibody-drug conjugate approved for the treatment of relapsed/refractory multiple myeloma. We report that GSK2857916 induces immunogenic cell death in BCMA-expressing cancer cells and promotes dendritic cell activation in vitro and in vivo GSK2857916 treatment enhances intratumor immune cell infiltration and activation, delays tumor growth, and promotes durable complete regressions in immune-competent mice bearing EL4 lymphoma tumors expressing human BCMA (EL4-hBCMA). Responding mice are immune to rechallenge with EL4 parental and EL4-hBCMA cells, suggesting engagement of an adaptive immune response, immunologic memory, and tumor antigen spreading, which are abrogated upon depletion of endogenous CD8+ T cells. Combinations with OX40/OX86, an immune agonist antibody, significantly enhance antitumor activity and increase durable complete responses, providing a strong rationale for clinical evaluation of GSK2857916 combinations with immunotherapies targeting adaptive immune responses, including T-cell-directed checkpoint modulators.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , B-Cell Maturation Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Immunoconjugates/pharmacology , Immunogenic Cell Death , Lymphoma/drug therapy , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Apoptosis , B-Cell Maturation Antigen/immunology , Cell Proliferation , Female , Humans , Lymphoma/immunology , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Toxicol Pathol ; 49(5): 996-1023, 2021 07.
Article in English | MEDLINE | ID: mdl-33576319

ABSTRACT

A host of novel renal biomarkers have been developed over the past few decades which have enhanced monitoring of renal disease and drug-induced kidney injury in both preclinical studies and in humans. Since chronic kidney disease (CKD) and acute kidney injury (AKI) share similar underlying mechanisms and the tubulointerstitial compartment has a functional role in the progression of CKD, urinary biomarkers of AKI may provide predictive information in chronic renal disease. Numerous studies have explored whether the recent AKI biomarkers could improve upon the standard clinical biomarkers, estimated glomerular filtration rate (eGFR), and urinary albumin to creatinine ratio, for predicting outcomes in CKD patients. This review is an introduction to alternative assays that can be utilized in chronic (>3 months duration) nonclinical safety studies to provide information on renal dysfunction and to demonstrate specific situations where these assays could be utilized in nonclinical drug development. Novel biomarkers such as symmetrical dimethyl arginine, dickkopf homolog 3, and cystatin C predict chronic renal injury in animals, act as surrogates for GFR, and may predict changes in GFR in patients over time, ultimately providing a bridge from preclinical to clinical renal monitoring.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Acute Kidney Injury/chemically induced , Acute Kidney Injury/diagnosis , Biomarkers , Creatinine , Glomerular Filtration Rate , Humans , Kidney , Renal Insufficiency, Chronic/chemically induced
3.
Toxicol Pathol ; 48(6): 766-783, 2020 08.
Article in English | MEDLINE | ID: mdl-32815469

ABSTRACT

We describe and characterize unilateral renal aplasia in a cynomolgus monkey (Macaca fascicularis) from a chronic toxicology study adding to the limited histopathology reports of congenital renal anomalies in macaques. In the current case, the affected kidney was macroscopically small and characterized microscopically by a thin cortex with an underdeveloped medulla and an absent papilla. The remnant medulla lacked a corticomedullary junction and contained only a few irregular collecting duct-like structures. The cortex had extensive interstitial mature collagen deposition with fibromuscular collar formation around Bowman's capsules. Due to parenchymal collapse, mature glomeruli were condensed together with occasional atrophic and sclerotic glomeruli. The majority of the cortical tubules were poorly differentiated with only small islands of fully developed cortical tubules present. Histochemical and immunohistochemical stains were utilized to demonstrate key diagnostic features of this congenital defect, to assist with differentiating it from renal dysplasia, and to provide potential mechanistic pathways. Immunostaining (S100, paired box gene 2 [PAX2], aquaporins) of the medulla was compatible with incomplete maturation associated with aplasia, while the immunostaining profile for the cortex (vimentin, calbindin, PAX2-positive cortical tubules, and smooth muscle actin-positive fibromuscular collars) was most compatible with dedifferentiation secondary to degenerative changes.


Subject(s)
Kidney Diseases , Monkey Diseases , Animals , Humans , Kidney/abnormalities , Kidney Diseases/veterinary , Kidney Glomerulus , Macaca fascicularis , Urogenital Abnormalities
4.
Toxicol Pathol ; 48(3): 481-493, 2020 04.
Article in English | MEDLINE | ID: mdl-31918642

ABSTRACT

Several chemicals and pharmaceuticals increase the incidence of hemangiosarcomas (HSAs) in mice, but the relevance to humans is uncertain. Recently, canine HSAs were identified as a powerful tool for investigating the pathogenesis of human HSAs. To characterize the cellular phenotype of canine HSAs, we evaluated immunoreactivity and/or messenger RNA (mRNA) expression of markers for hematopoietic stem cells (HSCs), endothelial cells (ECs), a tumor suppressor protein, and a myeloid marker in canine HSAs. Neoplastic canine cells expressed EC markers and a myeloid marker, but expressed HSC markers less consistently. The canine tumor expression results were then compared to previously published immunoreactivity results for these markers in human and mouse HSAs. There are 2 noteworthy differences across species: (1) most human HSAs had HSC marker expression, indicating that they were comprised of tumor cells that were less differentiated than those in canine and mouse tumors; and (2) human and canine HSAs expressed a late-stage EC maturation marker, whereas mouse HSAs were negative, suggesting that human and canine tumors may retain greater differentiation potential than mouse tumors. These results indicate that HSA development is variable across species and that caution is necessary when discussing translation of carcinogenic risk from animal models to humans.


Subject(s)
Biomarkers, Tumor/analysis , Dog Diseases/pathology , Hemangiosarcoma/pathology , Animals , Disease Models, Animal , Dogs , Endothelial Progenitor Cells/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Mice , Species Specificity
6.
Toxicol Pathol ; 47(7): 799-816, 2019 10.
Article in English | MEDLINE | ID: mdl-31280683

ABSTRACT

Chronic progressive nephropathy (CPN) is the most commonly encountered spontaneous background finding in laboratory rodents. Various theories on its pathogenesis have been proposed, but there is a paucity of data regarding specific mechanisms or physiologic pathways involved in early CPN development. The current CPN mechanism of action for tumorigenesis is largely based on its associated increase in tubular cell proliferation without regard to preceding subcellular degenerative changes. Combing through the published literature from multiple biology disciplines provided insight into the preceding cellular events. Mechanistic pathways involved in the progressive age-related decline in rodent kidney function and several key inflexion points have been identified. These critical pathway factors were then connected using data from renal models from multiple rodent strains, other species, and mechanistic work in humans to form a cohesive picture of pathways and protein interactions. Abundant data linked similar renal pathologies to local events involving hypoxia (hypoxia-inducible factor 1α), altered intrarenal renin-angiotensin system (RAS), oxidative stress (nitric oxide), and pro-inflammatory pathways (transforming growth factor ß), with positive feedback loops and downstream effectors amplifying the injury and promoting scarring. Intrarenal RAS alterations seem to be central to all these events and may be critical to CPN development and progression.


Subject(s)
Kidney Diseases/etiology , Renin-Angiotensin System/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Chronic Disease , Disease Progression , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Nitric Oxide/physiology , Polyarteritis Nodosa/etiology , Rats
7.
Sci Rep ; 9(1): 8420, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182754

ABSTRACT

Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Claudins/immunology , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/therapy , Stomach Neoplasms/therapy , Adenocarcinoma/therapy , Animals , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Humans , Immunoconjugates/blood , Mice , Pancreatic Neoplasms/blood , Rats , Stomach Neoplasms/blood
8.
Semin Nephrol ; 39(2): 190-201, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30827341

ABSTRACT

Drug-induced kidney toxicity is a significant contributor to acute kidney injury. Nephrotoxic drugs need to be identified during nonclinical testing to highlight potential risk translatable to the intended patient population. When nonclinical kidney toxicity signals arise, scientists and physicians affiliated with clinical trials need to be familiar with commonly encountered drug-induced perturbations in the kidney, terminology, and how these changes relate to clinical risk. Mechanistic and translational toxicologic studies beyond routine histopathology and clinical pathology approaches may be needed to elucidate the pathogenesis and human relevance to inform clinical risk assessment. Investigational studies may help elucidate specific sites of injury within the nephron, the presence of reactive metabolites, mechanisms of membrane transport or tissue distribution, potential drug-drug interactions, or the ability to recover function after drug withdrawal. Cutting-edge techniques such as in vitro alternative platforms, humanized animal models, translational imaging/microscopy or circulating/secretory biomarkers, omics platforms at the interface of genes, proteins, metabolites, or advanced molecular and biochemical approaches provide toxicologists and pathologists with a wide variety of potential experimental modalities to investigate mechanisms of kidney toxicity.


Subject(s)
Acute Kidney Injury/chemically induced , Nephrons/pathology , Acute Kidney Injury/diagnostic imaging , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Animals, Genetically Modified , Cell Line , Dogs , Drug Development , Gene Knockout Techniques , Humans , In Vitro Techniques , Kidney/diagnostic imaging , Kidney/metabolism , Kidney/pathology , Mice , Microvilli , Models, Animal , Nephrons/diagnostic imaging , Nephrons/metabolism , Primary Cell Culture , Rats , Zebrafish
9.
Toxicol Pathol ; 46(8): 904-917, 2018 12.
Article in English | MEDLINE | ID: mdl-30089413

ABSTRACT

Prevalence of immune-mediated glomerulonephritis has increased in preclinical toxicity studies, with more frequent use of biotherapeutic agents (especially antigenic humanized molecules) and antisense oligonucleotide (ASO) therapies. Immune complex disease affects a small number of study monkeys, often correlates with antidrug antibody (ADA) titers, and occurs at a dose that favors immune complex formation or impedes clearance. While preclinical glomerulonephritis often fails to correlate with evidence of glomerular or vascular injury in human clinical trials and is not considered predictive, additional animal investigative immunohistochemical work may be performed to substantiate evidence for immune complex pathogenesis. While ADA is most commonly encountered as a predisposing factor with biotherapeutic agents, complement activation may occur without circulating complexes, and other mechanisms of non-ADA immune-mediated glomerulonephritis have been observed including nonendogenous immune aggregates and immunoregulatory pharmacology. Although glomerulonephritis associated with oligonucleotide therapies has been noted occasionally in preclinical studies and more rarely with human patients, pathophysiologic mechanisms involved appear to be different between species and preclinical cases are not considered predictive for humans. ADA is not involved in oligonucleotide-associated cases, and complement fixation plays a more important role in monkeys. Recent screening of ASOs for proinflammatory activity appears to have decreased glomerulonephritis incidence preclinically.


Subject(s)
Biological Therapy/adverse effects , Glomerulonephritis/chemically induced , Oligonucleotides, Antisense/toxicity , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Immune Complex Diseases/chemically induced
10.
Toxicol Sci ; 161(1): 58-75, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28973697

ABSTRACT

Pharmaceuticals and chemicals produce hemangiosarcomas (HS) in mice, often by nongenotoxic, proliferative mechanisms. A mode-of-action (MOA) for hemangiosarcoma was proposed based on information presented at an international workshop (Cohen et al., Hemangiosarcoma in rodents: Mode-of-action evaluation and human relevance. Toxicol. Sci. 111, 4-18.). Five key elements of the MOA were articulated and included hypoxia, macrophage activation, increased angiogenic growth factors, dysregulated angiogenesis/erythropoiesis, and endothial cell proliferation. The goal of the current study was to add to the weight-of-evidence for the proposed MOA by assessing these key elements with 3 different compounds of varying potency for HS induction: fenretinide (high), troglitazone (intermediate), and elmiron (low). Multiple endpoints, including hypoxia (hyproxyprobe, transcriptomics), endothelial cell (EC) proliferation, and clinical and anatomic pathology, were assessed after 2, 4, and 13-weeks of treatment in B6C3F1 mice. All 3 compounds demonstrated strong evidence for dysregulated erythropoiesis (decrease in RBC and a failure to increase reticulocytes) and macrophage activation (4- to 11-fold increases); this pattern of hematological changes in mice might serve as an early biomarker to evaluate EC proliferation in suspected target organs for potential HS formation. Fenretinide demonstrated all 5 key elements, while troglitazone demonstrated 4 and elmiron demonstrated 3. Transcriptomics provided support for the 5 elements of the MOA, but was not any more sensitive than hypoxyprobe immunohistochemistry for detecting hypoxia. The overall transcriptional evidence for the key elements of the proposed MOA was also consistent with the potency of HS induction. These data, coupled with the previous work with 2-butoxyethanol and pregablin, increase the weight-of-evidence for the proposed MOA for HS formation.


Subject(s)
Fenretinide/toxicity , Hemangiosarcoma/chemically induced , Neovascularization, Pathologic/chemically induced , Pentosan Sulfuric Polyester/toxicity , Troglitazone/toxicity , Animals , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Hemangiosarcoma/metabolism , Hemangiosarcoma/pathology , Macrophage Activation/drug effects , Male , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Organ Specificity
11.
PLoS One ; 12(5): e0176768, 2017.
Article in English | MEDLINE | ID: mdl-28472135

ABSTRACT

A number of chemical compounds have been shown to induce liver tumors in mice but not in other species. While several mechanisms for this species-specific tumorigenicity have been proposed, no definitive mechanism has been established. We examined the effects of the nongenotoxic rodent hepatic carcinogen, WY-14,643, in male mice from a high liver tumor susceptible strain (C3H/HeJ), and from a low tumor susceptible strain (C57BL/6). WY-14,643, a PPARα activator induced widespread increases in the expression of some endogenous retroelements, namely members of LTR and LINE elements in both strains. The expression of a number of known retroviral defense genes was also elevated. We also demonstrated that basal immune-mediated viral defense was elevated in C57BL/6 mice (the resistant strain) and that WY-14,643 further activated those immuno-defense processes. We propose that the previously reported >100X activity of retroelements in mice drives mouse-specific tumorigenicity. We also propose that C57BL/6's competent immune to retroviral activation allows it to remove cells before the activation of these elements can result in significant chromosomal insertions and mutation. Finally, we showed that WY-14,643 treatment induced gene signatures of DNA recombination in the sensitive C3H/HeJ strain.


Subject(s)
Carcinogens/toxicity , Retroelements , Animals , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Pyrimidines/pharmacology
12.
Clin Cancer Res ; 23(7): 1760-1770, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27683177

ABSTRACT

Purpose: Adverse reactions reported in patients treated with antibody-calicheamicin conjugates such as gemtuzumab ozogamicin (Mylotarg) and inotuzumab ozogamicin include thrombocytopenia and sinusoidal obstruction syndrome (SOS). The objective of this experimental work was to investigate the mechanism for thrombocytopenia, characterize the liver injury, and identify potential safety biomarkers.Experimental Design: Cynomolgus monkeys were dosed intravenously at 6 mg/m2/dose once every 3 weeks with a nonbinding antibody-calicheamicin conjugate (PF-0259) containing the same linker-payload as gemtuzumab ozogamicin and inotuzumab ozogamicin. Monkeys were necropsied 48 hours after the first administration (day 3) or 3 weeks after the third administration (day 63).Results: PF-0259 induced acute thrombocytopenia (up to 86% platelet reduction) with nadirs on days 3 to 4. There was no indication of effects on megakaryocytes in bone marrow or activation of platelets in peripheral blood. Microscopic evaluation of liver from animals necropsied on day 3 demonstrated midzonal degeneration and loss of sinusoidal endothelial cells (SECs) associated with marked platelet accumulation in sinusoids. Liver histopathology on day 63 showed variable endothelial recovery and progression to a combination of sinusoidal capillarization and sinusoidal dilation/hepatocellular atrophy, consistent with early SOS. Among biomarkers evaluated, there were early and sustained increases in serum hyaluronic acid (HA) that correlated well with serum aspartate aminotransferase and liver microscopic changes, suggesting that HA may be a sensitive diagnostic marker of the liver microvascular injury.Conclusions: These data support the conclusion that target-independent damage to liver SECs may be responsible for acute thrombocytopenia (through platelet sequestration in liver sinusoids) and development of SOS. Clin Cancer Res; 23(7); 1760-70. ©2016 AACR.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Chemical and Drug Induced Liver Injury/pathology , Thrombocytopenia/pathology , Aminoglycosides/adverse effects , Aminoglycosides/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enediynes/administration & dosage , Enediynes/chemistry , Gemtuzumab , Humans , Hyaluronic Acid/blood , Immunoconjugates/administration & dosage , Immunoconjugates/adverse effects , Inotuzumab Ozogamicin , Liver/drug effects , Macaca fascicularis , Neoplasms/drug therapy , Neoplasms/pathology , Thrombocytopenia/chemically induced
13.
Toxicol Pathol ; 42(4): 784-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24670818

ABSTRACT

Several classes of drugs have been shown to cause drug-induced vascular injury (DIVI) in preclinical toxicity studies. Measurement of blood flow and vessel diameter in numerous vessels and across various tissues by ultrasound imaging has the potential to be a noninvasive translatable biomarker of DIVI. Our objective was to demonstrate the utility of high-frequency ultrasound imaging for measuring changes in vascular function by evaluating blood flow and vessel diameter in the superior mesenteric arteries (SMA) of rats treated with compounds that are known to cause DIVI and are known vasodilators in rat: fenoldopam, CI-1044, and SK&F 95654. Blood flow, vessel diameter, and other parameters were measured in the SMA at 4, 8, and 24 hr after dosing. Mild to moderate perivascular accumulations of mononuclear cells, neutrophils in tunica adventitia, and superficial tunica media as well as multifocal hemorrhage and necrosis in the tunica media were found in animals 24 hr after treatment with fenoldopam and SK&F 95654. Each compound caused marked increases in blood flow and shear stress as early as 4 hr after dosing. These results suggest that ultrasound imaging may constitute a functional correlate for the microscopic finding of DIVI in the rat.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Ultrasonography/methods , Vascular System Injuries/chemically induced , Vascular System Injuries/pathology , Animals , Azepines/adverse effects , Fenoldopam/adverse effects , Hemodynamics , Male , Mesenteric Arteries/diagnostic imaging , Mesenteric Arteries/drug effects , Niacinamide/adverse effects , Niacinamide/analogs & derivatives , Pyridazines/adverse effects , Pyridines/adverse effects , Rats , Rats, Sprague-Dawley , Vascular System Injuries/diagnostic imaging
14.
Toxicol Sci ; 128(1): 42-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22539613

ABSTRACT

The preceding article identified key components of pregabalin's mode of action on nongenotoxic hemangiosarcoma formation in mice, including increased serum bicarbonate leading to decreased respiratory rate, increased blood pH, increased venous oxygen saturation, increased vascular endothelial growth factor and basic fibroblast growth factor expression, increased hepatic vascular endothelial growth factor receptor 2 expression, and increased iron-laden macrophages. Increased platelet count and platelet activation were early, species-specific biomarkers in mice. Dysregulated erythropoiesis, macrophage activation, and elevations of tissue growth factors were consistent with the unified mode of action for nongenotoxic hemangiosarcoma recently proposed at an international hemangiosarcoma workshop (Cohen, S. M., Storer, R. D., Criswell, K. A., Doerrer, N. G., Dellarco, V. L., Pegg, D. G., Wojcinski, Z. W., Malarkey, D. E., Jacobs, A. C., Klaunig, J. E., et al. (2009). Hemangiosarcoma in rodents: Mode-of-action evaluation and human relevance. Toxicol. Sci. 111, 4-18). In this article, we present evidence that pregabalin induces hypoxia and increases endothelial cell (EC) proliferation in a species-specific manner. Dietary administration of pregabalin produced a significant 35% increase in an immunohistochemical stain for hypoxia (Hypoxyprobe) in livers from pregabalin-treated mice. Increased Hypoxyprobe staining was not observed in the liver, bone marrow, or spleen of rats, supporting the hypothesis that pregabalin produces local tissue hypoxia in a species-specific manner. Transcriptional analysis supports that rats, unlike mice, adapt to pregabalin-induced hypoxia. Using a dual-label method, increased EC proliferation was observed as early as 2 weeks in mouse liver and 12 weeks in bone marrow following pregabalin administration. These same assays showed decreased EC proliferation in hepatic ECs of rats, further supporting species specificity. Dietary supplementation with vitamin E, which is known to have antioxidant and antiangiogenic activity, inhibited pregabalin-induced increases in mouse hepatic EC proliferation, providing confirmatory evidence for the proposed mode of action and its species-specific response.


Subject(s)
Cell Hypoxia , Cell Proliferation/drug effects , Endothelium, Vascular/drug effects , Liver/drug effects , Vitamin E/administration & dosage , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Chromatography, High Pressure Liquid , Diet , Endothelium, Vascular/cytology , Female , Liver/cytology , Mice , Pregabalin , gamma-Aminobutyric Acid/toxicity
15.
Am J Pathol ; 179(1): 240-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21703406

ABSTRACT

Macrophage colony-stimulating factor (M-CSF) is a hematopoietic growth factor that is responsible for the survival and proliferation of monocytes and the differentiation of monocytes into macrophages, including Kupffer cells (KCs) in the liver. KCs play an important role in the clearance of several serum enzymes, including aspartate aminotransferase and creatine kinase, that are typically elevated as a result of liver or skeletal muscle injury. We used three distinct animal models to investigate the hypothesis that increases in the levels of serum enzymes can be the result of decreases in KCs in the apparent absence of hepatic or skeletal muscle injury. Specifically, neutralizing M-CSF activity via a novel human monoclonal antibody reduced the CD14(+)CD16(+) monocyte population, depleted KCs, and increased aspartate aminotransferase and creatine kinase serum enzyme levels in cynomolgus macaques. In addition, the treatment of rats with clodronate liposomes depleted KCs and led to increased serum enzyme levels, again without evidence of tissue injury. Finally, in the osteopetrotic (Csf1(op)/Csf1(op)) mice lacking functional M-CSF and having reduced levels of KCs, the levels of serum enzymes are higher than in wild-type littermates. Together, these findings support a mechanism for increases in serum enzyme levels through M-CSF regulation of tissue macrophage homeostasis without concomitant histopathological changes in either the hepatic or skeletal system.


Subject(s)
Aspartate Aminotransferases/blood , Creatine Kinase/blood , Kupffer Cells/pathology , Liver/enzymology , Muscle, Skeletal/enzymology , Osteopetrosis/pathology , Animals , Antibodies, Monoclonal/pharmacology , Bone Density Conservation Agents/pharmacology , Clodronic Acid/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Liver/injuries , Liver/pathology , Macaca fascicularis , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/physiology , Male , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Osteopetrosis/metabolism , Rats , Rats, Sprague-Dawley , Receptors, IgG/metabolism
16.
Birth Defects Res B Dev Reprod Toxicol ; 89(6): 474-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21058326

ABSTRACT

BACKGROUND: The effects of histamine H1 antagonist chlorcyclizine on rat palate development were characterized following in utero exposure. METHODS: To identify the optimum dose for inducing cleft palate, pregnant rats were administered 30, 60, or 90 mg/kg chlorcyclizine on Gestation Days 11 to 14. Fetal palate gene expression was also assessed after 90 mg/kg chlorcyclizine at 8, 15 and 30 hours post-dose on Gestation Day 14 using microarray and qRT-PCR. RESULTS: Rats in the 60- and 90-mg/kg groups exhibited adverse clinical signs and body weight loss. Rats in the 90-mg/kg group also demonstrated increases in late resorptions and decreases in fetal weight. Effects in the low-dose group were limited to decreases in body weight gain. Fetal assessment on Gestation Day 21 revealed that findings were limited to the 60- and 90-mg/kg groups, and included cleft palate (80% of litters for both groups), high arched palate, small nose, micrognathia, high domed head, digits shortened/absent and small limb. The fetal incidence of cleft palate was higher at 90 mg/kg, thus this dose was selected to assess palate gene expression. The altered genes associated with chlorcyclizine-induced cleft palate included Wnt5a, Bmp2, Bmp4, Fgf10, Fgfr2, Msx1, and Insig1 but the magnitude of the change was relatively small (1.5- to 2-fold). CONCLUSIONS: Expression of several genes involved in palate, limb and digit development was altered in the fetal palate following in utero exposure to chlorcyclizine. The subtle perturbation and interplay of these genes may have profound effects on the dynamics of fetal palate development.


Subject(s)
Cleft Palate/chemically induced , Embryo, Mammalian/drug effects , Histamine H1 Antagonists/toxicity , Palate/drug effects , Piperazines/toxicity , Animals , Biomarkers/metabolism , Cleft Palate/genetics , Cleft Palate/pathology , Embryo, Mammalian/abnormalities , Female , Fetal Resorption/chemically induced , Fetal Weight/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Maternal Exposure , Microarray Analysis , Palate/abnormalities , Palate/metabolism , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Weight Gain
17.
Nat Biotechnol ; 28(5): 446-54, 2010 May.
Article in English | MEDLINE | ID: mdl-20458314

ABSTRACT

Application of any new biomarker to support safety-related decisions during regulated phases of drug development requires provision of a substantial data set that critically assesses analytical and biological performance of that biomarker. Such an approach enables stakeholders from industry and regulatory bodies to objectively evaluate whether superior standards of performance have been met and whether specific claims of fit-for-purpose use are supported. It is therefore important during the biomarker evaluation process that stakeholders seek agreement on which critical experiments are needed to test that a biomarker meets specific performance claims, how new biomarker and traditional comparators will be measured and how the resulting data will be merged, analyzed and interpreted.


Subject(s)
Biomarkers , Drug Discovery , Pharmaceutical Preparations , Animals , Drug Discovery/legislation & jurisprudence , Drug Discovery/methods , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/standards
18.
Toxicol Sci ; 113(1): 254-66, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812364

ABSTRACT

To understand the molecular mechanisms underlying compound-induced hemangiosarcomas in mice, and therefore, their human relevance, a systems biology approach was undertaken using transcriptomics and Causal Network Modeling from mice treated with 2-butoxyethanol (2-BE). 2-BE is a hemolytic agent that induces hemangiosarcomas in mice. We hypothesized that the hemolysis induced by 2-BE would result in local tissue hypoxia, a well-documented trigger for endothelial cell proliferation leading to hemangiosarcoma. Gene expression data from bone marrow (BM), liver, and spleen of mice exposed to a single dose (4 h) or seven daily doses of 2-BE were used to develop a mechanistic model of hemangiosarcoma. The resulting mechanistic model confirms previous work proposing that 2-BE induces macrophage activation and inflammation in the liver. In addition, the model supports local tissue hypoxia in the liver and spleen, coupled with increased erythropoeitin signaling and erythropoiesis in the spleen and BM, and suppression of mechanisms that contribute to genomic stability, events that could be contributing factors to hemangiosarcoma formation. Finally, an immunohistochemistry method (Hypoxyprobe) demonstrated that tissue hypoxia was present in the spleen and BM. Together, the results of this study identify molecular mechanisms that initiate hemangiosarcoma, a key step in understanding safety concerns that can impact drug decision processes, and identified hypoxia as a possible contributing factor for 2-BE-induced hemangiosarcoma in mice.


Subject(s)
Bone Marrow/metabolism , Cell Transformation, Neoplastic/metabolism , Hemangiosarcoma/metabolism , Liver/metabolism , Models, Biological , Signal Transduction , Spleen/metabolism , Systems Biology , Animals , Bone Marrow/pathology , Cell Cycle , Cell Differentiation , Cell Hypoxia , Cell Proliferation , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Erythropoiesis , Erythropoietin/metabolism , Ethylene Glycols , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genomic Instability , Hemangiosarcoma/chemically induced , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Hematopoietic Stem Cells/metabolism , Hemolysis , Hepatitis/metabolism , Hepatitis/pathology , Immunohistochemistry , Liver/pathology , Macrophage Activation , Male , Mice , Spleen/pathology , Time Factors
19.
Mol Pharmacol ; 74(2): 320-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18413659

ABSTRACT

Organic anion-transporting polypeptides (OATP) 1B1 and 1B3 are widely acknowledged as important and rate-limiting to the hepatic uptake of many drugs in clinical use. Accordingly, to better understand the in vivo relevance of OATP1B transporters, targeted disruption of murine Slco1b2 gene was carried out. It is noteworthy that Slco1b2(-/-) mice were fertile, developed normally, and exhibited no overt phenotypic abnormalities. We confirmed the loss of Oatp1b2 expression in liver using real-time polymerase chain reaction, Western Blot analysis, and immunohistochemistry. Expression of Oatp1a4 and Oatp2b1 but not Oatp1a1 was greater in female Slco1b2(-/-) mice, but expression of other non-OATP transporters did not significantly differ between wild-type and Slco1b2(-/-) male mice. Total bilirubin level was elevated by 2-fold in the Slco1b2(-/-) mice despite the fact that liver enzymes ALT and AST were normal. Pharmacological characterization was carried out using two prototypical substrates of human OATP1B1 and -1B3, rifampin and pravastatin. After a single intravenous dose of rifampin (1 mg/kg), a 1.7-fold increase in plasma area under the concentration-time curve (AUC) was observed, whereas the liver-to-plasma ratio was reduced by 5-fold, and nearly 8-fold when assessed at steady-state conditions after 24 h of continuous subcutaneous infusion in Slco1b2(-/-) mice. Likewise, continuous subcutaneous infusion at low (8 microg/h) or high (32 microg/h) dose rates of pravastatin resulted in a 4-fold lower liver-plasma ratio in the in Slco1b2(-/-) mice. This is the first report of altered drug disposition profile in the Slco1b2 knockout mice and suggests the utility of this model for understanding the in vivo role of hepatic OATP transporters in drug disposition.


Subject(s)
Gene Targeting/methods , Organic Anion Transporters, Sodium-Independent/metabolism , Pravastatin/metabolism , Rifampin/metabolism , Animals , Female , Injections, Intravenous , Liver-Specific Organic Anion Transporter 1 , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Organic Anion Transporters, Sodium-Independent/deficiency , Organic Anion Transporters, Sodium-Independent/genetics , Pravastatin/pharmacokinetics , Rifampin/administration & dosage , Rifampin/pharmacokinetics , Substrate Specificity/genetics
20.
Toxicol Pathol ; 35(5): 728-34, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17763287

ABSTRACT

Hepatocellular vacuolation can be a diagnostic challenge since cytoplasmic accumulations of various substances (lipid, water, phospholipids, glycogen, and plasma) can have a similar morphology. Cytoplasmic accumulation of phospholipids following administration of cationic amphiphilic drugs (CAD) can be particularly difficult to differentiate from nonphosphorylated lipid accumulations at the light microscopic level. Histochemical methods (Sudan Black, Oil Red-O, Nile Blue, etc.) can be used to identify both nonphosphorylated and/or phosphorylated lipid accumulations, but these techniques require non-paraffin-embedded tissue and are only moderately sensitive. Thus, electron microscopy is often utilized to achieve a definitive diagnosis based upon the characteristic morphologic features of phospholipid accumulations; however, this is a low throughput and labor intense procedure. In this report, we describe the use of immunohistochemical staining for LAMP-2 (a lysosome-associated protein) and adipophilin (a protein that forms the membrane around non-lysosomal lipid droplets) to differentiate phospholipidosis and lipidosis, respectively in the livers of rats. This staining procedure can be performed on formalin-fixed paraffin embedded tissues, is more sensitive than histochemistry, and easier to perform than ultrastructural evaluation.


Subject(s)
Lipidoses/diagnosis , Liver/ultrastructure , Lysosomal-Associated Membrane Protein 2/analysis , Peptides/analysis , Phospholipids/metabolism , Vacuoles/drug effects , Animals , Cytoplasm/metabolism , Diagnosis, Differential , Female , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Membrane Proteins , Perilipin-2 , Rats , Rats, Sprague-Dawley , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL