Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
J Med Chem ; 64(4): 2024-2045, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33538587

ABSTRACT

We identified a set of thiosemicarbazone (TSC) metal ion chelators that reactivate specific zinc-deficient p53 mutants using a mechanism called zinc metallochaperones (ZMCs) that restore zinc binding by shuttling zinc into cells. We defined biophysical and cellular assays necessary for structure-activity relationship studies using this mechanism. We investigated an alternative class of zinc scaffolds that differ from TSCs by substitution of the thiocarbamoyl moiety with benzothiazolyl, benzoxazolyl, and benzimidazolyl hydrazones. Members of this series bound zinc with similar affinity and functioned to reactivate mutant p53 comparable to the TSCs. Acute toxicity and efficacy assays in rodents demonstrated C1 to be significantly less toxic than the TSCs while demonstrating equivalent growth inhibition. We identified C85 as a ZMC with diminished copper binding that functions as a chemotherapy and radiation sensitizer. We conclude that the benzothiazolyl, benzoxazolyl, and benzimidazolyl hydrazones can function as ZMCs to reactivate mutant p53 in vitro and in vivo.


Subject(s)
Benzothiazoles/therapeutic use , Benzoxazoles/therapeutic use , Chelating Agents/therapeutic use , Hydrazones/therapeutic use , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacology , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacology , Cell Line, Tumor , Chelating Agents/chemical synthesis , Chelating Agents/pharmacology , Humans , Hydrazones/chemical synthesis , Hydrazones/pharmacology , Mice, Nude , Molecular Structure , Neoplasms/drug therapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Structure-Activity Relationship , Tumor Suppressor Protein p53/drug effects , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 18(8): 1355-1365, 2019 08.
Article in English | MEDLINE | ID: mdl-31196889

ABSTRACT

Chemotherapy and radiation are more effective in wild-type (WT) p53 tumors due to p53 activation. This is one rationale for developing drugs that reactivate mutant p53 to synergize with chemotherapy and radiation. Zinc metallochaperones (ZMC) are a new class of mutant p53 reactivators that restore WT structure and function to zinc-deficient p53 mutants. We hypothesized that the thiosemicarbazone, ZMC1, would synergize with chemotherapy and radiation. Surprisingly, this was not found. We explored the mechanism of this and found the reactive oxygen species (ROS) activity of ZMC1 negates the signal on p53 that is generated with chemotherapy and radiation. We hypothesized that a zinc scaffold generating less ROS would synergize with chemotherapy and radiation. The ROS effect of ZMC1 is generated by its chelation of redox active copper. ZMC1 copper binding (K Cu) studies reveal its affinity for copper is approximately 108 greater than Zn2+ We identified an alternative zinc scaffold (nitrilotriacetic acid) and synthesized derivatives to improve cell permeability. These compounds bind zinc in the same range as ZMC1 but bound copper much less avidly (106- to 107-fold lower) and induced less ROS. These compounds were synergistic with chemotherapy and radiation by inducing p53 signaling events on mutant p53. We explored other combinations with ZMC1 based on its mechanism of action and demonstrate that ZMC1 is synergistic with MDM2 antagonists, BCL2 antagonists, and molecules that deplete cellular reducing agents. We have identified an optimal Cu2+:Zn2+ binding ratio to facilitate development of ZMCs as chemotherapy and radiation sensitizers. Although ZMC1 is not synergistic with chemotherapy and radiation, it is synergistic with a number of other targeted agents.


Subject(s)
Copper/metabolism , Metallochaperones/metabolism , Mutation , Transcriptional Activation/drug effects , Transcriptional Activation/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line , Combined Modality Therapy , Humans , Metallochaperones/genetics , Mice , Protein Binding , Pyridines/pharmacology , Radiation , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
4.
NPJ Breast Cancer ; 5: 14, 2019.
Article in English | MEDLINE | ID: mdl-30993195

ABSTRACT

Triple negative breast cancer (TNBC) is an aggressive subset for which effective therapeutic approaches are needed. A significant proportion of TNBC patients harbor either germline or somatic mutations in BRCA1, or epigenetic silencing of BRCA1, which renders them deficient in DNA repair. Virtually all BRCA1 deficient breast cancers harbor mutations in TP53 suggesting that inactivation of p53 is a requirement for tumor progression in the setting of BRCA1 deficiency. Due to this dependency, we hypothesized that restoring wild type p53 function in BRCA1 deficient breast cancer would be therapeutic. The majority of TP53 mutations are missense, which generate a defective protein that potentially can be targeted with small molecules. Zinc metallochaperones (ZMCs) are a new class of anti-cancer drugs that specifically reactivate zinc-deficient mutant p53 by restoring zinc binding. Using ZMC1 in human breast cancer cell lines expressing the zinc deficient p53R175H, we demonstrate that loss of BRCA1 sensitizes cells to mutant p53 reactivation. Using murine breast cancer models with Brca1 deficiency, we demonstrate that ZMC1 significantly improves survival of mice bearing tumors harboring the zinc-deficient Trp53 R172H allele but not the Trp53 -/- allele. We synthesized a new formulation of ZMC1 (Zn-1), in which the drug is made in complex with zinc to improve zinc delivery, and demonstrate that Zn-1 has increased efficacy. Furthermore, we show that ZMC1 plus olaparib is a highly effective combination for p53R172H tumor growth inhibition. In conclusion, we have validated preclinically a new therapeutic approach for BRCA1 deficient breast cancer through reactivation of mutant p53.

5.
Clin Cancer Res ; 24(18): 4505-4517, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29914895

ABSTRACT

Purpose: Zinc metallochaperones (ZMC) are a new class of anticancer drugs that reactivate zinc-deficient mutant p53 by raising and buffering intracellular zinc levels sufficiently to restore zinc binding. In vitro pharmacodynamics of ZMCs indicate that p53-mutant activity is ON by 4-6 hours and is OFF by 24. We sought to understand the mechanism of this regulation and to translate these findings preclinically. We further sought to innovate the formulation of ZMCs to improve efficacy.Experimental Design: We performed in vitro mechanistic studies to determine the role of cellular zinc homeostatic mechanisms in the transient pharmacodynamics of ZMCs. We conducted preclinical pharmacokinetic, pharmacodynamic, and efficacy studies using a genetically engineered murine pancreatic cancer model (KPC) to translate these mechanistic findings and investigate a novel ZMC formulation.Results:In vitro, cellular zinc homeostatic mechanisms that restore zinc to its physiologic levels function as the OFF switch in ZMC pharmacodynamics. In vivo pharmacokinetic studies indicate that ZMCs have a short half-life (< 30 minutes), which is sufficient to significantly improve survival in mice expressing a zinc-deficient allele (p53R172H) while having no effect in mice expressing a non-zinc-deficient allele (p53R270H). We synthesized a novel formulation of the drug in complex with zinc and demonstrate this significantly improves survival over ZMC1.Conclusions: Cellular zinc homeostatic mechanisms function as an OFF switch in ZMC pharmacodynamics, indicating that a brief period of p53-mutant reactivation is sufficient for on-target efficacy. ZMCs synthesized in complex with zinc are an improved formulation. Clin Cancer Res; 24(18); 4505-17. ©2018 AACR.


Subject(s)
Metallochaperones/pharmacology , Pancreatic Neoplasms/drug therapy , Tumor Suppressor Protein p53/genetics , Zinc/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Disease Models, Animal , Humans , Metallochaperones/chemistry , Metallochaperones/pharmacokinetics , Mice , Mutant Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Zinc/deficiency
6.
Target Oncol ; 12(4): 449-462, 2017 08.
Article in English | MEDLINE | ID: mdl-28589491

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) represents one of the most lethal cancers worldwide due to therapy resistance and disease recurrence. Tumor relapse following treatment could be driven by the persistence of liver cancer stem-like cells (CSCs). The protein BMI1 is a member of the polycomb epigenetic factors governing cellular self-renewal, proliferation, and stemness maintenance. BMI1 expression also correlates with poor patient survival in various cancer types. OBJECTIVE: We aimed to elucidate the extent to which BMI1 can be used as a potential therapeutic target for CSC eradication in HCC. METHODS: We have recently participated in characterizing the first known pharmacological small molecule inhibitor of BMI1. Here, we synthesized a panel of novel BMI1 inhibitors and examined their ability to alter cellular growth and eliminate cancer progenitor/stem-like cells in HCC with different p53 backgrounds. RESULTS: Among various molecules examined, RU-A1 particularly downregulated BMI1 expression, impaired cell viability, reduced cell migration, and sensitized HCC cells to 5-fluorouracil (5-FU) in vitro. Notably, long-term analysis of HCC survival showed that, unlike chemotherapy, RU-A1 effectively reduced CSC content, even as monotherapy. BMI1 inhibition with RU-A1 diminished the number of stem-like cells in vitro more efficiently than the model compound C-209, as demonstrated by clonogenic assays and impairment of CSC marker expression. Furthermore, xenograft assays in zebrafish showed that RU-A1 abrogated tumor growth in vivo. CONCLUSIONS: This study demonstrates the ability to identify agents with the propensity for targeting CSCs in HCC that could be explored as novel treatments in the clinical setting.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Polycomb Repressive Complex 1/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , HEK293 Cells , Hep G2 Cells , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/genetics , Small Molecule Libraries/chemistry , Xenograft Model Antitumor Assays , Zebrafish
7.
Dermatoendocrinol ; 9(1): e1300213, 2017.
Article in English | MEDLINE | ID: mdl-28458767

ABSTRACT

Supplementation by the general public with vitamin D at doses above the Tolerable Upper Level of Intake (UL) is becoming quite common. The objective of the current analysis was to characterize the effect of vitamin D supplementation at doses up to 15,000 IU/d in a community-based program on vitamin D status, calcium homeostasis as well as on kidney, liver and immune function. We evaluated data collected for 3,882 participants in a community program for whom there were blood measurements at program entry and at follow-up within 6-18 months between 2013 and 2015. Participants were supplemented with a wide range of vitamin D doses (1,000 - 15,000 IU/d) aimed at achieving serum 25-hydroxyvitamin D [25(OH)D] levels of at least 100 nmol/L. Serum 25(OH)D concentrations up to 300 nmol/L were achieved without perturbation of calcium homeostasis or incidence of toxicity. Hypercalcemia and hypercalciuria were not related to an increase in 25(OH)D concentrations nor vitamin D dose. To achieve serum 25(OH)D levels >100 nmol/L on average, required vitamin D intakes of 6,000 IU/d for normal Body Mass Index (BMI), 7,000 IU/d for overweight and 8,000 IU/d for obese. Doses of vitamin D in excess of 6,000 IU/d were required to achieve serum 25(OH)D concentrations above 100 nmol/L, especially in individuals who were overweight or obese without any evidence of toxicity. Serum 25(OH)D concentrations up to 300 nmol/L were found to be safe.

8.
Mol Pharmacol ; 91(6): 567-575, 2017 06.
Article in English | MEDLINE | ID: mdl-28320780

ABSTRACT

Small-molecule restoration of wild-type structure and function to mutant p53 (so-called mutant reactivation) is a highly sought-after goal in cancer drug development. We previously discovered that small-molecule zinc chelators called zinc metallochaperones (ZMCs) reactivate mutant p53 by restoring zinc binding to zinc-deficient p53 mutants. The lead compound identified from the NCI-60 human tumor cell lines screen, NSC319726 (ZMC1), belongs to the thiosemicarbazone (TSC) class of metal ion chelators that bind iron, copper, magnesium, zinc, and other transition metals. Here, we have investigated the other TSCs, NSC319725 and NSC328784, identified in the same screen, as well as the more well studied TSC, 3-AP (Triapine), to determine whether they function as ZMCs. We measured the zinc Kd zinc ionophore activity, ability to restore zinc to purified p53 DNA binding domain (DBD), and ability to restore site-specific DNA binding to purified R175H-DBD in vitro. We tested all four TSCs in a number of cell-based assays to examine mutant p53 reactivation and the generation of reactive oxygen species (ROS). We found that NSC319725 and NSC328784 behave similarly to ZMC1 in both biophysical and cell-based assays and are heretofore named ZMC2 (NSC319725) and ZMC3 (NSC328784). 3-AP generates a ROS signal similar to ZMC1-3, but it fails to function as a ZMC both in vitro and in cells and ultimately does not reactivate p53. These findings indicate that not all TSCs function as ZMCs, and much of their activity can be predicted by their affinity for zinc.


Subject(s)
Growth Inhibitors/metabolism , Metallochaperones/metabolism , Mutation/physiology , Thiosemicarbazones/metabolism , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Growth Inhibitors/pharmacology , Humans , Mutation/drug effects , Tumor Suppressor Protein p53/genetics
11.
Mol Pharmacol ; 87(5): 825-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25710967

ABSTRACT

p53 is a Zn(2+)-dependent tumor suppressor inactivated in >50% of human cancers. The most common mutation, R175H, inactivates p53 by reducing its affinity for the essential zinc ion, leaving the mutant protein unable to bind the metal in the low [Zn(2+)]free environment of the cell. The exploratory cancer drug zinc metallochaperone-1 (ZMC1) was previously demonstrated to reactivate this and other Zn(2+)-binding mutants by binding Zn(2+) and buffering it to a level such that Zn(2+) can repopulate the defective binding site, but how it accomplishes this in the context of living cells and organisms is unclear. In this study, we demonstrated that ZMC1 increases intracellular [Zn(2+)]free by functioning as a Zn(2+) ionophore, binding Zn(2+) in the extracellular environment, diffusing across the plasma membrane, and releasing it intracellularly. It raises intracellular [Zn(2+)]free in cancer (TOV112D) and noncancer human embryonic kidney cell line 293 to 15.8 and 18.1 nM, respectively, with half-times of 2-3 minutes. These [Zn(2+)]free levels are predicted to result in ∼90% saturation of p53-R175H, thus accounting for its observed reactivation. This mechanism is supported by the X-ray crystal structure of the [Zn(ZMC1)2] complex, which demonstrates structural and chemical features consistent with those of known metal ionophores. These findings provide a physical mechanism linking zinc metallochaperone-1 in both in vitro and in vivo activities and define the remaining critical parameter necessary for developing synthetic metallochaperones for clinical use.


Subject(s)
Biological Transport/physiology , Carrier Proteins/metabolism , Ionophores/metabolism , Metallochaperones/metabolism , Tumor Suppressor Protein p53/metabolism , Zinc/metabolism , Binding Sites , Cell Line , Cell Membrane/metabolism , HEK293 Cells , Humans , Mutation/genetics , Protein Conformation , Tumor Suppressor Protein p53/genetics
13.
Oncotarget ; 5(19): 8879-92, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25294809

ABSTRACT

UNLABELLED: NSC319726 (ZMC1) is a small molecule that reactivates mutant p53 by restoration of WT structure/function to the most common p53 missense mutant (p53-R175H). We investigated the mechanism by which ZMC1 reactivates p53-R175H and provide evidence that ZMC1: 1) restores WT structure by functioning as a zinc-metallochaperone, providing an optimal concentration of zinc to facilitate proper folding; and 2) increases cellular reactive oxygen species that transactivate the newly conformed p53-R175H (via post-translational modifications), inducing an apoptotic program. We not only demonstrate that this zinc metallochaperone function is possessed by other zinc-binding small molecules, but that it can reactivate other p53 mutants with impaired zinc binding. This represents a novel mechanism for an anti-cancer drug and a new pathway to drug mutant p53. SIGNIFICANCE: We have elucidated a novel mechanism to restore wild-type structure/function to mutant p53 using small molecules functioning as zinc-metallochaperones. The pharmacologic delivery of a metal ion to restore proper folding of a mutant protein is unique to medicinal chemistry and represents a new pathway to drug mutant p53.


Subject(s)
Antineoplastic Agents/pharmacology , Metallochaperones/pharmacology , Pyridines/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , HCT116 Cells , Humans , MCF-7 Cells , Metallochaperones/metabolism , Mutation , Protein Binding , Protein Folding/drug effects , Pyridines/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Zinc/metabolism
14.
Bioorg Med Chem Lett ; 24(16): 3886-90, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25037917

ABSTRACT

The incidence of cognitive disorders such as Alzheimer's disease continues to increase unabated. While cures for such diseases have eluded investigators, progress is being made on alleviating certain symptoms of these diseases. Mouse knockouts of the proline transporter (PROT), a high affinity Na(+)/Cl(-)-dependent transporter, indicated its potential as a novel therapeutic target for cognition improvement. Herein we report our investigation into a novel class of PROT inhibitors.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Cognition Disorders/drug therapy , Small Molecule Libraries/pharmacology , Amino Acid Transport Systems, Neutral/deficiency , Amino Acid Transport Systems, Neutral/metabolism , Animals , Biological Transport/drug effects , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Mice , Mice, Knockout , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship
15.
Neuropathol Appl Neurobiol ; 39(5): 458-84, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23336971

ABSTRACT

Vitamin D and its metabolites have pleomorphic roles in both nervous system health and disease. Animal models have been paramount in contributing to our knowledge and understanding of the consequences of vitamin D deficiency on brain development and its implications for adult psychiatric and neurological diseases. The conflation of in vitro, ex vivo, and animal model data provide compelling evidence that vitamin D has a crucial role in proliferation, differentiation, neurotrophism, neuroprotection, neurotransmission, and neuroplasticity. Vitamin D exerts its biological function not only by influencing cellular processes directly, but also by influencing gene expression through vitamin D response elements. This review highlights the epidemiological, neuropathological, experimental and molecular genetic evidence implicating vitamin D as a candidate in influencing susceptibility to a number of psychiatric and neurological diseases. The strength of evidence varies for schizophrenia, autism, Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease, and is especially strong for multiple sclerosis.


Subject(s)
Nervous System Diseases/metabolism , Nervous System/metabolism , Vitamin D/metabolism , Humans , Nervous System/physiopathology , Nervous System Diseases/physiopathology , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/physiopathology
16.
Vasc Endovascular Surg ; 46(2): 187-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22308215

ABSTRACT

Central venous catheters are commonly utilized to gain vascular access for varied clinical indications. Successful catheter placement requires not only technical expertise but also awareness of the potential complications. In this article, we report a malposition of a central venous catheter in a hemodialysis patient. We review the complications of this catheter misplacement, as well as complications resulting from other central venous catheter malpositions.


Subject(s)
Catheterization, Central Venous/adverse effects , Catheters, Indwelling/adverse effects , Renal Dialysis , Thorax/blood supply , Catheterization, Central Venous/instrumentation , Device Removal , Female , Humans , Middle Aged , Radiography, Interventional , Tomography, X-Ray Computed , Veins
18.
J Med Chem ; 53(24): 8650-62, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21090716

ABSTRACT

Sphingosine 1-phosphate lyase (S1PL) has been characterized as a novel target for the treatment of autoimmune disorders using genetic and pharmacological methods. Medicinal chemistry efforts targeting S1PL by direct in vivo evaluation of synthetic analogues of 2-acetyl-4(5)-(1(R),2(S),3(R),4-tetrahydroxybutyl)-imidazole (THI, 1) led to the discovery of 2 (LX2931) and 4 (LX2932). The immunological phenotypes observed in S1PL deficient mice were recapitulated by oral administration of 2 or 4. Oral dosing of 2 or 4 yielded a dose-dependent decrease in circulating lymphocyte numbers in multiple species and showed a therapeutic effect in rodent models of rheumatoid arthritis (RA). Phase I clinical trials indicated that 2, the first clinically studied inhibitor of S1PL, produced a dose-dependent and reversible reduction of circulating lymphocytes and was well tolerated at dose levels of up to 180 mg daily. Phase II evaluation of 2 in patients with active rheumatoid arthritis is currently underway.


Subject(s)
Aldehyde-Lyases/antagonists & inhibitors , Antirheumatic Agents/chemical synthesis , Imidazoles/chemical synthesis , Isoxazoles/chemical synthesis , Oximes/chemical synthesis , Aldehyde-Lyases/genetics , Animals , Antirheumatic Agents/pharmacokinetics , Antirheumatic Agents/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Blood Pressure/drug effects , Cell Movement , Dogs , Heart Rate/drug effects , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Isoxazoles/pharmacokinetics , Isoxazoles/pharmacology , Lymphocytes/drug effects , Lymphocytes/physiology , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Oximes/pharmacokinetics , Oximes/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship
19.
Neurology ; 74(23): 1852-9, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20427749

ABSTRACT

OBJECTIVE: Low vitamin D status has been associated with multiple sclerosis (MS) prevalence and risk, but the therapeutic potential of vitamin D in established MS has not been explored. Our aim was to assess the tolerability of high-dose oral vitamin D and its impact on biochemical, immunologic, and clinical outcomes in patients with MS prospectively. METHODS: An open-label randomized prospective controlled 52-week trial matched patients with MS for demographic and disease characteristics, with randomization to treatment or control groups. Treatment patients received escalating vitamin D doses up to 40,000 IU/day over 28 weeks to raise serum 25-hydroxyvitamin D [25(OH)D] rapidly and assess tolerability, followed by 10,000 IU/day (12 weeks), and further downtitrated to 0 IU/day. Calcium (1,200 mg/day) was given throughout the trial. Primary endpoints were mean change in serum calcium at each vitamin D dose and a comparison of serum calcium between groups. Secondary endpoints included 25(OH)D and other biochemical measures, immunologic biomarkers, relapse events, and Expanded Disability Status Scale (EDSS) score. RESULTS: Forty-nine patients (25 treatment, 24 control) were enrolled [mean age 40.5 years, EDSS 1.34, and 25(OH)D 78 nmol/L]. All calcium-related measures within and between groups were normal. Despite a mean peak 25(OH)D of 413 nmol/L, no significant adverse events occurred. Although there may have been confounding variables in clinical outcomes, treatment group patients appeared to have fewer relapse events and a persistent reduction in T-cell proliferation compared to controls. CONCLUSIONS: High-dose vitamin D (approximately 10,000 IU/day) in multiple sclerosis is safe, with evidence of immunomodulatory effects. CLASSIFICATION OF EVIDENCE: This trial provides Class II evidence that high-dose vitamin D use for 52 weeks in patients with multiple sclerosis does not significantly increase serum calcium levels when compared to patients not on high-dose supplementation. The trial, however, lacked statistical precision and the design requirements to adequately assess changes in clinical disease measures (relapses and Expanded Disability Status Scale scores), providing only Class level IV evidence for these outcomes.


Subject(s)
Calcium/administration & dosage , Multiple Sclerosis/diet therapy , Multiple Sclerosis/metabolism , Vitamin D/administration & dosage , Vitamins/administration & dosage , Adolescent , Adult , Age Factors , Calcium/urine , Case-Control Studies , Cell Proliferation/drug effects , Cytokines/blood , Dose-Response Relationship, Drug , Female , Humans , Male , Matrix Metalloproteinase 9/metabolism , Middle Aged , Prospective Studies , Statistics, Nonparametric , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamins/metabolism , Young Adult
20.
Pharmacogenomics J ; 10(5): 385-95, 2010 Oct.
Article in English | MEDLINE | ID: mdl-19997081

ABSTRACT

Biomarkers that can be measured in preclinical models in a high-throughput, reproducible manner offer the potential to increase the speed and efficacy of drug development. Development of therapeutic agents for many conditions is hampered by the limited number of validated preclinical biomarkers available to gauge pharmacoefficacy and disease progression, but the validation process for preclinical biomarkers has received limited attention. This report defines a five-step preclinical biomarker validation process and applies the process to a case study of diabetic retinopathy. By showing that a gene expression panel is highly reproducible, coincides with disease manifestation, accurately classifies individual animals and identifies animals treated with a known therapeutic agent, a biomarker panel can be considered validated. This particular biomarker panel consisting of 14 genes (C1inh, C1s, Carhsp1, Chi3l1, Gat3, Gbp2, Hspb1, Icam1, Jak3, Kcne2, Lama5, Lgals3, Nppa, Timp1) can be used in diabetic retinopathy pharmacotherapeutic research, and the biomarker development process outlined here is applicable to drug development efforts for other diseases.


Subject(s)
Biomarkers, Pharmacological/analysis , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Animals , Databases, Genetic , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/genetics , Endpoint Determination , Gene Expression/drug effects , Gene Expression Profiling , Genetic Markers/genetics , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Insulin/administration & dosage , Insulin/therapeutic use , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...