Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 263
Filter
1.
Eur J Cancer ; 160: 72-79, 2022 01.
Article in English | MEDLINE | ID: mdl-34785111

ABSTRACT

BACKGROUND: The outcome of infants with KMT2A-germline acute lymphoblastic leukaemia (ALL) is superior to that of infants with KMT2A-rearranged ALL but has been inferior to non-infant ALL patients. Here, we describe the outcome and prognostic factors for 167 infants with KMT2A-germline ALL enrolled in the Interfant-06 study. METHODS: Univariate analysis on prognostic factors (age, white blood cell count at diagnosis, prednisolone response and CD10 expression) was performed on KMT2A-germline infants in complete remission at the end of induction (EOI; n = 163). Bone marrow minimal residual disease (MRD) was measured in 73 patients by real-time quantitative polymerase chain reaction at various time points (EOI, n = 68; end of consolidation, n = 56; and before OCTADAD, n = 57). MRD results were classified as negative, intermediate (<5∗10-4), and high (≥5∗10-4). RESULTS: The 6-year event-free and overall survival was 73.9% (standard error [SE] = 3.6) and 87.2% (SE = 2.7). Relapses occurred early, within 36 months from diagnosis in 28 of 31 (90%) infants. Treatment-related mortality was 3.6%. Age <6 months was a favourable prognostic factor with a 6-year disease-free survival (DFS) of 91% (SE = 9.0) compared with 71.7% (SE = 4.2) in infants >6 months of age (P = 0.04). Patients with high EOI MRD ≥5 × 10-4 had a worse outcome (6-year DFS 61.4% [SE = 12.4], n = 16), compared with patients with undetectable EOI MRD (6-year DFS 87.9% [SE = 6.6], n = 28) or intermediate EOI MRD <5 × 10-4 (6-year DFS 76.4% [SE = 11.3], n = 24; P = 0.02). CONCLUSION: We conclude that young age at diagnosis and low EOI MRD seem favourable prognostic factors in infants with KMT2A-germline ALL and should be considered for risk stratification in future clinical trials.


Subject(s)
Neoplasm, Residual/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Female , Germ Cells , Humans , Infant , Male , Neoplasm, Residual/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Survival Analysis , Treatment Outcome
3.
Horm Res Paediatr ; 91(6): 357-372, 2019.
Article in English | MEDLINE | ID: mdl-31319416

ABSTRACT

This update, written by authors designated by multiple pediatric endocrinology societies (see List of Participating Societies) from around the globe, concisely addresses topics related to changes in GnRHa usage in children and adolescents over the last decade. Topics related to the use of GnRHa in precocious puberty include diagnostic criteria, globally available formulations, considerations of benefit of treatment, monitoring of therapy, adverse events, and long-term outcome data. Additional sections review use in transgender individuals and other pediatric endocrine related conditions. Although there have been many significant changes in GnRHa usage, there is a definite paucity of evidence-based publications to support them. Therefore, this paper is explicitly not intended to evaluate what is recommended in terms of the best use of GnRHa, based on evidence and expert opinion, but rather to describe how these drugs are used, irrespective of any qualitative evaluation. Thus, this paper should be considered a narrative review on GnRHa utilization in precocious puberty and other clinical situations. These changes are reviewed not only to point out deficiencies in the literature but also to stimulate future studies and publications in this area.


Subject(s)
Gonadotropin-Releasing Hormone/therapeutic use , Puberty, Precocious , Adolescent , Child , Female , Humans , Male , Puberty, Precocious/diagnosis , Puberty, Precocious/drug therapy , Puberty, Precocious/pathology , Puberty, Precocious/physiopathology
4.
Int J Lab Hematol ; 40(3): 343-351, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29500862

ABSTRACT

INTRODUCTION: Minimal residual disease (MRD) in B lymphoblastic leukemia has been demonstrated to be a powerful predictor of clinical outcome in numerous studies in both children and adults. In this study, we evaluated 86 pediatric patients with both diagnostic and remission flow cytometry studies and compared expression of CD81, CD58, CD19, CD34, CD20, and CD38 in the detection of MRD. METHODS: We evaluated 86 patients with B lymphoblastic leukemia who had both diagnostic studies and remission studies for the presence of MRD using multicolor flow cytometry. We established our detection limit for identifying abnormal lymphoblasts using serial dilutions. We also compared flow cytometry findings with molecular MRD detection in a subset of patients. RESULTS: We found that we can resolve differences between hematogones and lymphoblasts in 85 of 86 cases using a combination of CD45, CD19, CD34, CD10, CD20, CD38, CD58, and CD81. Our detection limit using flow cytometry is 0.002% for detecting a population of abnormal B lymphoblasts. Comparison with MRD assessment by molecular methods showed a high concordance rate with flow cytometry findings. CONCLUSIONS: Our study highlights importance of using multiple markers to detect MRD in B lymphoblastic leukemia. Our findings indicate that including both CD58 and CD81 markers in addition to CD19, CD34, CD20, CD38, and CD10 are helpful in MRD detection by flow cytometry.


Subject(s)
CD58 Antigens/blood , Neoplasm, Residual/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Tetraspanin 28/blood , Adolescent , Antigens, CD/blood , Biomarkers, Tumor/blood , CD58 Antigens/physiology , Child , Child, Preschool , Female , Flow Cytometry/methods , Humans , Male
5.
Pediatr Blood Cancer ; 65(5): e26952, 2018 05.
Article in English | MEDLINE | ID: mdl-29319209

ABSTRACT

BACKGROUND: Pediatric patients receiving induction chemotherapy for newly diagnosed acute lymphoblastic leukemia (ALL) are at high risk of developing life-threatening infections. We investigated whether uniform antibacterial guidelines, including mandatory antibacterial prophylaxis in afebrile patients during induction, decreases the incidence of microbiologically documented bacteremia. METHODS: Between 2012 and 2015, 230 patients with newly diagnosed ALL (aged 1-21) were enrolled on Dana-Farber Cancer Institute ALL Consortium Protocol 11-001 (DFCI 11-001). Induction therapy, regardless of risk group, included vincristine, prednisone, doxorubicin, methotrexate, and PEG-asparaginase. Afebrile patients received fluoroquinolone prophylaxis at the initiation of induction and those presenting with fever received broad-spectrum antibiotics; antibiotics were continued until blood count recovery. Rates of documented bacteremias and fungal infections on DFCI 11-001 were compared to those on the predecessor protocol (DFCI 05-001), which included the same induction phase without antibiotic prophylaxis guidelines. RESULTS: Sixty-six (28.7%) patients received fluoroquinolone prophylaxis, the remaining patients received broad-spectrum antibiotics. Twenty-four (36.4%) patients on prophylaxis developed fever and seven (10.6%) developed bacteremia. The overall rate of infection during induction on DFCI 11-001 was lower than on DFCl 05-001 (14.3% vs. 26.3%, P < 0.0001) due to a decreased rate of bacteremia (10.9% vs. 24.4%, P < 0.0001). The rate of fungal infections (4.8% vs. 3.6%) and induction death (0.9% vs. 2%) was not significantly different. CONCLUSION: For children with newly diagnosed ALL, uniform antibiotic administration until blood count recovery, including fluoroquinolone prophylaxis for afebrile patients, reduced the incidence of bacteremia during the induction phase. Larger, randomized studies should be performed to confirm these findings.


Subject(s)
Antibiotic Prophylaxis , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bacteremia/prevention & control , Induction Chemotherapy/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Asparaginase/administration & dosage , Bacteremia/chemically induced , Bacteremia/microbiology , Child , Child, Preschool , Doxorubicin/administration & dosage , Female , Follow-Up Studies , Humans , Infant , Male , Methotrexate/administration & dosage , Polyethylene Glycols/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prednisone/administration & dosage , Prognosis , Survival Rate , Vincristine/administration & dosage , Young Adult
7.
Oncogene ; 36(49): 6762-6773, 2017 12 07.
Article in English | MEDLINE | ID: mdl-28806397

ABSTRACT

Upon different types of stress, the gene encoding the mitosis-promoting phosphatase Cdc25C is transcriptionally repressed by p53, contributing to p53's enforcement of a G2 cell cycle arrest. In addition, Cdc25C protein stability is also decreased following DNA damage. Mdm2, another p53 target gene, encodes a ubiquitin ligase that negatively regulates p53 levels by ubiquitination. Ablation of Mdm2 by siRNA led to an increase in p53 protein and repression of Cdc25C gene expression. However, Cdc25C protein levels were actually increased following Mdm2 depletion. Mdm2 is shown to negatively regulate Cdc25C protein levels by reducing its half-life independently of the presence of p53. Further, Mdm2 physically interacts with Cdc25C and promotes its degradation through the proteasome in a ubiquitin-independent manner. Either Mdm2 overexpression or Cdc25C downregulation delays cell cycle progression through the G2/M phase. Thus, the repression of the Cdc25C promoter by p53, together with p53-dependent induction of Mdm2 and subsequent degradation of Cdc25C, could provide a dual mechanism by which p53 can enforce and maintain a G2/M cell cycle arrest.


Subject(s)
G2 Phase Cell Cycle Checkpoints/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , cdc25 Phosphatases/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line , Cell Line, Tumor , Cells, Cultured , Down-Regulation/drug effects , Doxorubicin/pharmacology , Gene Expression Regulation/drug effects , HCT116 Cells , Humans , Immunoblotting , Mice, Knockout , Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism , cdc25 Phosphatases/metabolism
9.
Vox Sang ; 110(1): 79-89, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26084882

ABSTRACT

BACKGROUND AND OBJECTIVES: Epigenetic modifications tightly regulate the gene expression and cellular function of haematopoietic stem cells. Histone deacetylase inhibitors (HDACIs) alter the gene expression profile of cord blood (CB) CD34(+) cells by controlling the genes involved in chromatin modification, thereby influencing the self-renewal, maintenance and expansion of haematopoietic stem and progenitor cells (HSPCs). MATERIALS AND METHODS: The class I and II HDACIs, valproic acid and scriptaid, were utilized to expand CB-CD34(+) cells ex vivo. The gene profiling was performed on HSPC using Illumina microarray, GeneGO MetaCore(™) and Ingenuity pathway analyses. The molecular analyses were performed using Q-PCR and Western blotting. RESULTS: Each HDACI treatment of CB-CD34(+) cells created unique epigenetic and molecular signatures that governed chromatin modification required for cellular and functional behaviour of stem cells. GeneGO MetaCore(™) and Ingenuity pathway analyses established the molecular understanding of epigenetically regulated HSPCs in the presence of scriptaid and VPA that revealed different network(s) of potential regulators during erythropoiesis. VPA induced transcriptional activation of the glucocorticoid receptor (GCR) and an increase in the intracellular signalling of signal transducers and activators of transcription (STAT) required during stress erythropoiesis. Canonical Wnt signalling and many epigenetically regulated chromatin remodellers were significantly influenced so as to establish maintenance and regulation of HSPC. CONCLUSION: Treatment with Individual HDACIs has demonstrated significantly unique epigenetic and molecular signatures of CB-HSPC. This study identifies potential key regulators of HSPC and gives insights into the clinically important processes of HSPC expansion and haematopoietic lineage development for transplantation purposes.


Subject(s)
Antigens, CD34/genetics , Chromatin/genetics , Epigenesis, Genetic , Hematopoietic Stem Cells/drug effects , Histone Deacetylase Inhibitors/pharmacology , Transcriptome , Antigens, CD34/metabolism , Cells, Cultured , Chromatin/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Valproic Acid/pharmacology
11.
Pharmacogenomics J ; 16(6): 530-535, 2016 11.
Article in English | MEDLINE | ID: mdl-26345518

ABSTRACT

Anthracyclines are efficient chemotherapy agents. However, their use is limited by anthracycline-induced cardiotoxicity (CT). We investigated the influence of polymorphisms in doxorubicin metabolic and functional pathways on late-onset CT as estimated by echocardiography in 251 childhood acute lymphoblastic leukemia (cALL) patients. Association analyses revealed a modulating effect of two variants: A-1629 T in ABCC5, an ATP-binding cassette transporter, and G894T in the NOS3 endothelial nitric oxide synthase gene. Individuals with the ABCC5 TT-1629 genotype had an average of 8-12% reduction of ejection (EF) and shortening fractions (SF; EF: P<0.0001, and SF: P=0.001, respectively). A protective effect of the NOS3 TT894 genotype on EF was seen in high-risk patients (P=0.02), especially in those who did not receive dexrazoxane (P=0.002). Analysis of an additional cohort of 44 cALL patients replicated the ABCC5 association but was underpowered for NOS3. In summary, we identified two biomarkers that may contribute to cALL anthracycline CT risk stratification.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Heart Diseases/genetics , Multidrug Resistance-Associated Proteins/genetics , Nitric Oxide Synthase Type III/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Cardiotonic Agents/therapeutic use , Cardiotoxicity , Child , Child, Preschool , Dexrazoxane/therapeutic use , Female , Genetic Predisposition to Disease , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/prevention & control , Heterozygote , Homozygote , Humans , Infant , Male , Multidrug Resistance-Associated Proteins/metabolism , Myocardial Contraction , Nitric Oxide Synthase Type III/metabolism , Pharmacogenetics , Phenotype , Protective Factors , Risk Assessment , Risk Factors , Stroke Volume , Time Factors , Treatment Outcome , Ventricular Function, Left , Young Adult
12.
Leukemia ; 29(3): 526-34, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25079173

ABSTRACT

On the basis of the data suggesting that adolescents and young adult patients with acute lymphoblastic leukemia (ALL) have improved outcomes when treated on pediatric protocols, we assessed the feasibility of treating adult patients aged 18-50 years with ALL with the DFCI Pediatric ALL Consortium regimen utilizing a 30-week course of pharmacokinetically dose-adjusted E. coli L-asparaginase during consolidation. Between 2002 and 2008, 92 eligible patients aged 18-50 years were enrolled at 13 participating centers. Seventy-eight patients (85%) achieved a complete remission (CR) after 1 month of intensive induction therapy. With a median follow-up of 4.5 years, the 4-year disease-free survival (DFS) for the patients achieving a CR was 69% (95% confidence interval (CI) 56-78%) and the 4-year overall survival (OS) for all eligible patients was 67% (95% CI 56-76%). The 4-year DFS for the 64 patients who achieved a CR and were Philadelphia chromosome negative (Ph-) was 71% (95% CI 58-81%), and for all 74 Ph- patients the 4-year OS was 70% (95% CI 58-79%). We conclude that a pediatric-like treatment strategy for young adults with de novo ALL is feasible, associated with tolerable toxicity, and results in improved outcomes compared with historical regimens in young adult patients with ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Asparaginase/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Cytarabine/administration & dosage , Doxorubicin/administration & dosage , Drug Administration Schedule , Female , Humans , Karyotyping , Male , Methotrexate/administration & dosage , Middle Aged , Precision Medicine , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prednisone/administration & dosage , Remission Induction , Survival Analysis , Treatment Outcome , Vincristine/administration & dosage
13.
Leukemia ; 28(9): 1819-27, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24552990

ABSTRACT

Treatment resistance in T-cell acute lymphoblastic leukemia (T-ALL) is associated with phosphatase and tensin homolog (PTEN) deletions and resultant phosphatidylinositol 3'-kinase (PI3K)-AKT pathway activation, as well as MYC overexpression, and these pathways repress mitochondrial apoptosis in established T-lymphoblasts through poorly defined mechanisms. Normal T-cell progenitors are hypersensitive to mitochondrial apoptosis, a phenotype that is dependent on the expression of proapoptotic BIM. In a conditional zebrafish model, MYC downregulation induced BIM expression in T-lymphoblasts, an effect that was blunted by expression of constitutively active AKT. In human T-ALL cell lines and treatment-resistant patient samples, treatment with MYC or PI3K-AKT pathway inhibitors each induced BIM upregulation and apoptosis, indicating that BIM is repressed downstream of MYC and PI3K-AKT in high-risk T-ALL. Restoring BIM function in human T-ALL cells using a stapled peptide mimetic of the BIM BH3 domain had therapeutic activity, indicating that BIM repression is required for T-ALL viability. In the zebrafish model, where MYC downregulation induces T-ALL regression via mitochondrial apoptosis, T-ALL persisted despite MYC downregulation in 10% of bim wild-type zebrafish, 18% of bim heterozygotes and in 33% of bim homozygous mutants (P=0.017). We conclude that downregulation of BIM represents a key survival signal downstream of oncogenic MYC and PI3K-AKT signaling in treatment-resistant T-ALL.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Membrane Proteins/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-myc/physiology , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Bcl-2-Like Protein 11 , Cell Line, Tumor , Humans , Imidazoles/therapeutic use , Membrane Proteins/antagonists & inhibitors , MicroRNAs/physiology , Phosphatidylinositol 3-Kinases/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Quinolines/therapeutic use , Signal Transduction/physiology , Zebrafish
14.
Horm Res Paediatr ; 80(4): 242-51, 2013.
Article in English | MEDLINE | ID: mdl-24021297

ABSTRACT

BACKGROUND: Several models have been developed to predict growth response to growth hormone (GH) based on auxological and biochemical parameters for children with non-GH-deficient, idiopathic short stature (ISS). OBJECTIVE: To demonstrate if an individualized, formula-based, target-driven GH regimen for children with ISS would lead to a height (Ht) gain to -1.3 SDS during the first 24 months of treatment of this 4-year study, with less variability than with standard weight-based dosing. METHODS: A 4-year, open-label, multi-center, randomized, two-arm study comparing formula-based dosing of Genotropin® GH from 0.18 to 0.7 mg/kg/week versus standard FDA-approved ISS dosing of Genotropin® (0.37 mg/kg/week). Subjects (n = 316, 89 females) were prepubertal, 3-14 years of age, bone age 3-10 years (m) and 3-9 years (f), naive to GH treatment, Ht SDS -3 to -2.25, Ht velocity <25th percentile for bone age, and peak GH >10 ng/ml. RESULTS: The majority (83%) of subjects had Ht SDS within the normal range by 2 years. All subjects displayed catch-up growth consistent with other studies of GH treatment of ISS. CONCLUSION: The formula-based therapy did not meet the primary endpoint achieving targeted gain with lower variability. No new safety concerns were found.


Subject(s)
Dwarfism, Pituitary/drug therapy , Dwarfism, Pituitary/physiopathology , Human Growth Hormone/administration & dosage , Child , Child, Preschool , Dwarfism, Pituitary/pathology , Female , Human Growth Hormone/adverse effects , Humans , Male , Time Factors
15.
Horm Res Paediatr ; 79(5): 257-70, 2013.
Article in English | MEDLINE | ID: mdl-23735882

ABSTRACT

The goal of growth hormone (GH) treatment in a short child is to attain a fast catch-up growth toward the target height (TH) standard deviation score (SDS), followed by a maintenance phase, a proper pubertal height gain, and an adult height close to TH. The short-term response variable of GH treatment, first-year height velocity (HV) (cm/year or change in height SDS), can either be compared with GH response charts for diagnosis, age and gender, or with predicted HV based on prediction models. Three types of prediction models have been described: the Kabi International Growth Hormone Study models, the Gothenburg models and the Cologne model. With these models, 50-80% of the variance could be explained. When used prospectively, individualized dosing reduces the variation in growth response in comparison with a fixed dose per body weight. Insulin-like growth factor-I-based dose titration also led to a decrease in the variation. It is uncertain whether adding biochemical, genetic or proteomic markers may improve the accuracy of the prediction. Prediction models may lead to a more evidence-based approach to determine the GH dose regimen and may reduce the drug costs for GH treatment. There is a need for user-friendly software programs to make prediction models easily available in the clinic.


Subject(s)
Growth Disorders/drug therapy , Human Growth Hormone/therapeutic use , Models, Biological , Adolescent , Adult , Age Factors , Biomarkers/metabolism , Child , Child, Preschool , Female , Growth Disorders/physiopathology , Humans , Insulin-Like Growth Factor I/metabolism , Male , Sex Factors
17.
Vet Pathol ; 48(3): 772-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21383116

ABSTRACT

The pharmaceutical industry and regulatory agency toxicology testing paradigms in the United States currently appear successful, in part because of the continuously increasing life expectancy and the declining age-adjusted cancer rates in the United States. Although drugs likely have a minimal impact on the population statistics for cancer rates, pharmaceutical pathologists and toxicologists must focus on the individual risk for pharmaceutical carcinogenesis. As our understanding of carcinogenesis increases exponentially, and after hundreds if not thousands of rodent cancer tests, significant improvement in the precision of human pharmaceutical carcinogenesis hazard identification should now be possible and would enable a reduction in the substantial false-negative and false positive-rates reported herein. The appropriate use of acute, subchronic, chronic, and special toxicology tests to identify the major associated cancer risk factors, specifically, hormonal modulation, immunosuppression, genetic toxicity, and chronic toxicity, can be recognized through this review of pharmaceutical carcinogens. Significant opportunities exist for improving the effectiveness and efficiency of the current cancer risk assessment paradigm.


Subject(s)
Biological Assay/methods , Carcinogenicity Tests/methods , Drug-Related Side Effects and Adverse Reactions , Animals , Humans , Risk Factors , Rodentia
18.
Langmuir ; 26(12): 9899-904, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20491496

ABSTRACT

The effects of three acidic hexapeptides on in vitro hydroxyapatite growth were characterized by pH-stat kinetic studies, adsorption isotherms, and molecular modeling. The three peptides, pSDEpSDE, SDESDE, and DDDDDD, are equal-length model compounds for the acidic sequences in osteopontin, a protein that inhibits mineral formation in both calcified and noncalcified tissues. Growth rates from 1.67 mM calcium and 1.00 mM phosphate solution were measured at pH 7.4 and 37 degrees C in 150 mM NaCl. pSDEpSDE was a strong growth inhibitor when preadsorbed onto hydroxyapatite (HA) seeds from > or = 0.67 mM solutions, concentrations where adsorption isotherms showed relatively complete surface coverage. The nonphosphorylated SDESDE control showed no growth inhibition. Although it adsorbed to almost the same extent as pSDEpSDE, it rapidly desorbed under the pH-stat growth conditions while pSDEpSDE did not. DDDDDD exhibited weak inhibition as its concentration was increased and similar adsorption/desorption behavior to pSDEpSDE. Molecular modeling yielded binding energy trends based on simple adsorption of peptides on the [100] surface that were consistent with observed inhibition, but not for the [001] surface. The relatively unfavorable binding energies for peptides on the [001] surface suggest that their absorption will be primarily on the [100] face. The kinetic and adsorption data are consistent with phosphorylation of osteopontin acting to control mineral formation.


Subject(s)
Durapatite/antagonists & inhibitors , Osteopontin/chemistry , Peptide Fragments/pharmacology , Adsorption , Animals , Calcification, Physiologic , Durapatite/chemistry , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Phosphorylation
19.
Vet Pathol ; 47(2): 358-67, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124009

ABSTRACT

Proteasome inhibitor therapeutics (PITs) have the potential to cause peripheral neuropathy. In a mouse model of PIT-induced peripheral neuropathy, the authors demonstrated that ubiquitin-positive multifocal protein aggregates with nuclear displacement appear in dorsal root ganglion cells of animals that subsequently develop nerve injuries. This peripheral-nerve effect in nonclinical models has generally been recognized as the correlate of grade 3 neuropathy in clinical testing. In differentiated PC12 cells, the authors demonstrated perturbations correlative with the development of neuropathy in vivo, including ubiquitinated protein aggregate (UPA) formation and/or nuclear displacement associated with the degree of proteasome inhibition. They compared 7 proteasome inhibitors of 3 chemical scaffolds (peptide boronate, peptide epoxyketone, and lactacystin analog) to determine if PIT-induced peripheral neuropathy is modulated by inhibition of the proteasome (ie, a mechanism-based effect) or due to effects independent of proteasome inhibition (ie, an off target or chemical-structure-based effect). The appearance of UPAs was assayed at IC(90) +/- 5% (90% inhibition concentration +/- 5%) for 20S proteasome inhibition. Results show that each of the investigated proteasome inhibitors induced identical proteasome-inhibitor-specific ubiquitin-positive immunostaining and nuclear displacement in PC12 cells. Other agents--such as paclitaxel, cisplatin, and thalidomide, which cause neuropathy by other mechanisms--did not cause UPAs or nuclear displacement, demonstrating that the effect was specific to proteasome inhibitors. In conclusion, PIT-induced neuronal cell UPA formation and nuclear displacement are mechanism based and independent of the proteasome inhibitor scaffold. These data indicate that attempts to modulate the neuropathy associated with PIT may not benefit from changing scaffolds.


Subject(s)
Peripheral Nervous System Diseases/chemically induced , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Ubiquitin/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Proliferation , Cell Survival , Dose-Response Relationship, Drug , Immunohistochemistry , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Oligopeptides/pharmacology , PC12 Cells , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/metabolism , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Rats , Ubiquitin/antagonists & inhibitors
20.
Toxicol Pathol ; 38(1): 148-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20075108

ABSTRACT

A daunting, unmet medical need exists for effective oncology chemotherapies, with cancer deaths in 2009 to exceed 560,000 in the United States alone. Because of the rapid demise of the majority of cancer patients with metastatic disease, oncology drug development must follow a much different paradigm than therapeutic candidates for less onerous diseases. The majority of drug candidates in development today are targeted at cancer therapy. Many of these candidate chemotherapeutic agents are active against novel targets, often presenting unique toxicological profiles. Since many of these novel targets are not unique to cancer cells, therapeutic margins may not exist. Decision making, in this event, is among the most challenging that any pharmaceutical toxicologist/pathologist or regulator will face. Nonclinical development scientists must compress timelines to present therapeutic options for cancer patients who have failed conventional therapy. In support of this goal, the U. S. Food and Drug Administration has created an oncology-specific paradigm for nonclinical testing and has introduced strategies to accelerate development and approval of successful candidates. Pharmaceutical toxicology testing strategies must not only satisfy regulation as the minimal expectation, but also attempt to reduce the current high attrition rates for oncologic candidates. A successful toxicology testing strategy represents the substance of this treatise.


Subject(s)
Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Toxicology/methods , Animals , Databases, Factual , Drug Discovery , Humans , Investigational New Drug Application , Maximum Tolerated Dose , Neoplasm Staging , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...