Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Blood Cells Mol Dis ; 63: 1-8, 2017 03.
Article in English | MEDLINE | ID: mdl-27888688

ABSTRACT

An oral therapeutic which reduces duration of cytopenias and is active following accidental radiation exposures is an unmet need in radiation countermeasures. Alpha methylhydrocinnamate (ST7) prolongs STAT-5 phosphorylation, reduces growth-factor dependency of multi-lineage cell lines, and stimulates erythropoiesis. Here, ST7 and its isomers were studied for their effects on myeloid progenitors and hematopoietic stem cells (HSCs) following radiation, in nonhuman primates, and murine irradiation models. Addition of ST7 or ST7-S increased CFU-GM production by 1.7-fold (p<0.001), reduced neutrophil apoptosis comparable to G-CSF, and enhanced HSC survival post-radiation by 2-fold, (p=0.028). ST7 and ST7-S administered in normal baboons increased ANC and platelet counts by 50-400%. In sub-lethally-irradiated mice, ANC nadir remained >200/mm3 and neutropenia recovered in 6days with ST7 treatment and 18days in controls (p<0.05). In lethally-irradiated mice, marrow pathology at 15days was hypocellular (10% cellularity) in controls, but normal (55-75% cellularity) with complete neutrophil maturation with ST7-S treatment. Following lethal irradiation, ST7, given orally for 4days, reduced mortality, with 30% survival in ST7-animals vs 8% in controls, (p<0.05). Collectively, the studies indicate that ST7 and ST7-S enhance myeloid recovery post-radiation and merit further evaluation to accelerate hematologic recovery in conditions of radiation-related and other marrow hypoplasias.


Subject(s)
Myeloid Cells/drug effects , Neutrophils/drug effects , Phenylpropionates/therapeutic use , Recovery of Function/drug effects , Whole-Body Irradiation/adverse effects , Animals , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Mice , Myeloid Cells/radiation effects , Neutrophils/radiation effects , Papio , Phenylpropionates/pharmacology , Radiation Exposure/adverse effects , Survival Rate , Whole-Body Irradiation/mortality
2.
PLoS One ; 10(12): e0144660, 2015.
Article in English | MEDLINE | ID: mdl-26713848

ABSTRACT

High-level fetal (γ) globin expression ameliorates clinical severity of the beta (ß) hemoglobinopathies, and safe, orally-bioavailable γ-globin inducing agents would benefit many patients. We adapted a LCR-γ-globin promoter-GFP reporter assay to a high-throughput robotic system to evaluate five diverse chemical libraries for this activity. Multiple structurally- and functionally-diverse compounds were identified which activate the γ-globin gene promoter at nanomolar concentrations, including some therapeutics approved for other conditions. Three candidates with established safety profiles were further evaluated in erythroid progenitors, anemic baboons and transgenic mice, with significant induction of γ-globin expression observed in vivo. A lead candidate, Benserazide, emerged which demonstrated > 20-fold induction of γ-globin mRNA expression in anemic baboons and increased F-cell proportions by 3.5-fold in transgenic mice. Benserazide has been used chronically to inhibit amino acid decarboxylase to enhance plasma levels of L-dopa. These studies confirm the utility of high-throughput screening and identify previously unrecognized fetal globin inducing candidates which can be developed expediently for treatment of hemoglobinopathies.


Subject(s)
Anemia/genetics , Fetal Hemoglobin/genetics , High-Throughput Screening Assays , Papio , Transcriptional Activation/drug effects , gamma-Globins/genetics , Animals , Benserazide/adverse effects , Benserazide/pharmacology , Drug Evaluation, Preclinical , Erythroid Precursor Cells/drug effects , Loratadine/adverse effects , Loratadine/analogs & derivatives , Loratadine/pharmacology , Mice , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Br J Haematol ; 161(4): 587-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23530969

ABSTRACT

ß-thalassaemia intermedia (BTI) syndromes cause haemolytic anaemia, ineffective erythropoiesis, and widespread complications. Higher fetal globin expression within genotypes reduces globin imbalance and ameliorates anaemia. Sodium 2,2 dimethylbutyrate (HQK-1001), an orally bioavailable short-chain fatty acid derivative, induces γ-globin expression experimentally and is well-tolerated in normal subjects. Accordingly, a randomized, blinded, placebo-controlled, Phase I/II trial was performed in 21 adult BTI patients (14 with HbE/ß(0) thalassaemia and seven with ß(+)/ß(0) thalassaemia intermedia, to determine effective doses for fetal globin induction, safety, and tolerability. HQK-1001 or placebo were administered once daily for 8 weeks at four dose levels (10, 20, 30, or 40 mg/kg per day), and subjects were monitored for laboratory and clinical events. Pharmacokinetic profiles demonstrated a t(1/2) of 10-12 h. Adverse events with HQK-1001 treatment were not significantly different from placebo treatment. The 20 mg/kg treatment doses increased median HbF above baseline levels by 6·6% and 4·4 g/l (P < 0·01) in 8/9 subjects; total haemoglobin (Hb) increased by a mean of 11 g/l in 4/9 subjects. These findings identified a safe oral therapeutic which induces fetal globin in BTI. Further investigation of HQK-1001 with longer dosing to definitively evaluate its haematological potential appears warranted.


Subject(s)
Butyrates/pharmacology , Butyrates/therapeutic use , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics , Administration, Oral , Adolescent , Adult , Butyrates/administration & dosage , Butyrates/adverse effects , Female , Fetal Hemoglobin/metabolism , Humans , Male , Middle Aged , Splenectomy , Treatment Outcome , Young Adult , beta-Thalassemia/surgery
5.
Biores Open Access ; 1(4): 192-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23514803

ABSTRACT

In addition to being a part of the metabolic fatty acid fuel cycle, butyrate is also capable of inducing growth arrest in a variety of normal cell types and senescence-like phenotypes in gynecological cancer cells, inhibiting DNA synthesis and cell growth in colonic tumor cell lines, suppressing hTERT mRNA expression and telomerase activity in human prostate cancer cells, and inducing stem cell differentiation and apoptosis by DNA fragmentation. It regulates gene expression by inhibiting histone deacetylases (HDACs), enhances memory recovery and formation in mice, stimulates neurogenesis in the ischemic brain, promotes osteoblast formation, selectively blocks cell replication in transformed cells (compared to healthy cells), and can prevent and treat diet-induced obesity and insulin resistance in mouse models of obesity, as well as stimulate fetal hemoglobin expression in individuals with hematologic diseases such as the thalassemias and sickle-cell disease, in addition to a multitude of other biochemical effects in vivo. However, efforts to exploit the potential of butyrate in the clinical treatment of cancer and other medical disorders are thwarted by its poor pharmacological properties (short half-life and first-pass hepatic clearance) and the multigram doses needed to achieve therapeutic concentrations in vivo. Herein, we review some of the methods used to overcome these difficulties with an emphasis on HDAC inhibition.

6.
Blood Cells Mol Dis ; 47(2): 107-16, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21641240

ABSTRACT

The ß-hemoglobinopathies and thalassemias are serious genetic blood disorders affecting the ß-globin chain of hemoglobin A (α(2)ß(Α)(2)). Their clinical severity can be reduced by enhancing expression of fetal hemoglobin (γ-globin), producing HbF (α(2)γ(2,)). In studies reported here, γ-globin induction by 23 novel, structurally-unrelated compounds, which had been predicted through molecular modeling and in silico screening of a 13,000 chemical library, was evaluated in vitro in erythroid progenitors cultured from normal subjects and ß-thalassemia patients, and in vivo in transgenic mice or anemic baboons. Four predicted candidates were found to have high potency, with 4- to 8-fold induction of HbF. Two of these compounds have pharmacokinetic profiles favorable for clinical application. These studies thus effectively identified high potency γ-globin inducing candidate therapeutics and validated the utility of in silico molecular modeling.


Subject(s)
Anemia/drug therapy , Biological Products/administration & dosage , Drug Design , Erythroid Precursor Cells/drug effects , Fetal Hemoglobin/biosynthesis , Small Molecule Libraries/administration & dosage , beta-Thalassemia/drug therapy , gamma-Globins/biosynthesis , Administration, Oral , Anemia/genetics , Anemia/metabolism , Animals , Biological Products/chemistry , Biological Products/therapeutic use , Cells, Cultured , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/genetics , Gene Expression , Humans , Injections, Intravenous , Mice , Mice, Transgenic , Models, Molecular , Papio , Phlebotomy , Polymerase Chain Reaction , RNA, Messenger/analysis , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , beta-Globins/deficiency , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , gamma-Globins/genetics
7.
J Clin Pharmacol ; 51(8): 1186-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21422239

ABSTRACT

Pharmacologic induction of fetal globin synthesis is an accepted therapeutic strategy for treatment of the beta hemoglobinopathies and thalassemias, as even small increases in hemoglobin F (HbF) levels reduce clinical severity in sickle cell disease (SCD) and reduce anemia in beta thalassemia. Prior generation short chain fatty acid therapeutics, arginine butyrate (AB), and phenylbutyrate, increased fetal and total hemoglobin levels in patients, but were limited by high doses or intravenous (IV) infusion. A fetal globin-inducing therapeutic with convenient oral dosing would be an advance for these classic molecular diseases. Healthy adult human subjects were treated with a novel short chain fatty acids (SCFA) derivative, sodium 2,2 dimethylbutyrate (SDMB), or placebo, with 1 of 4 single dose levels (2, 5, 10, and 20 mg/kg) or daily doses (5, 10, or 15 mg/kg) over 14 days, and monitored for adverse clinical and laboratory events, drug levels, reticulocytes, and HbF assays. SDMB was well-tolerated with no clinically significant adverse events related to study medication. The terminal half-life ranged from 9 to 15 hours. Increases in mean absolute reticulocytes were observed at all dose levels in the 14-day study. The favorable pharmacokinetics (PK) profiles and safety findings indicate that SDMB warrants further investigation for treatment of anemic subjects with beta hemoglobinopathies.


Subject(s)
Butyrates/adverse effects , Butyrates/pharmacokinetics , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Hematinics/adverse effects , Hematinics/pharmacokinetics , Administration, Oral , Adolescent , Adult , Biological Availability , Blood Cell Count , Butyrates/administration & dosage , Butyrates/analysis , Dose-Response Relationship, Drug , Double-Blind Method , Drugs, Investigational/administration & dosage , Drugs, Investigational/analysis , Female , Fetal Hemoglobin/analysis , Food-Drug Interactions , Half-Life , Hematinics/administration & dosage , Hematinics/analysis , Humans , Male , Metabolic Clearance Rate , Plasma/chemistry , Reticulocytes/drug effects , Urine/chemistry , Young Adult
8.
Eur J Haematol ; 82(6): 466-76, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19220418

ABSTRACT

OBJECTIVES: The erythroid Kruppel-like factor (EKLF) is an essential transcription factor for beta-type globin gene switching, and specifically activates transcription of the adult beta-globin gene promoter. We sought to determine if EKLF is also required for activation of the gamma-globin gene by short-chain fatty acid (SCFA) derivatives, which are now entering clinical trials. METHODS: The functional and physical interaction of EKLF and co-regulatory molecules with the endogenous human globin gene promoters was studied in primary human erythroid progenitors and cell lines, using chromatin immunoprecipitation (ChIP) assays and genetic manipulation of the levels of EKLF and co-regulators. RESULTS AND CONCLUSIONS: Knockdown of EKLF prevents SCFA-induced expression of the gamma-globin promoter in a stably expressed microLCRbeta(pr)R(luc) (A)gamma(pr)F(luc) cassette, and prevents induction of the endogenous gamma-globin gene in primary human erythroid progenitors. EKLF is actively recruited to endogenous gamma-globin gene promoters after exposure of primary human erythroid progenitors, and murine hematopoietic cell lines, to SCFA derivatives. The core ATPase BRG1 subunit of the human SWI/WNF complex, a ubiquitous multimeric complex that regulates gene expression by remodeling nucleosomal structure, is also required for gamma-globin gene induction by SCFA derivatives. BRG1 is actively recruited to the endogenous gamma-globin promoter of primary human erythroid progenitors by exposure to SCFA derivatives, and this recruitment is dependent upon the presence of EKLF. These findings demonstrate that EKLF, and the co-activator BRG1, previously demonstrated to be required for definitive or adult erythropoietic patterns of globin gene expression, are co-opted by SCFA derivatives to activate the fetal globin genes.


Subject(s)
Fatty Acids, Volatile/physiology , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic , Transcriptional Activation , gamma-Globins/genetics , Animals , Cells, Cultured , DNA Helicases/physiology , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin , Humans , Kruppel-Like Transcription Factors/physiology , Mice , Nuclear Proteins/physiology , Protein Transport , Transcription Factors/physiology
9.
Blood ; 108(9): 3179-86, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16849648

ABSTRACT

High-level induction of fetal (gamma) globin gene expression for therapy of beta-hemoglobinopathies likely requires local chromatin modification and dissociation of repressor complexes for gamma-globin promoter activation. A novel gamma-globin-inducing short-chain fatty acid derivative (SCFAD), RB7, which was identified through computational modeling, produced a 6-fold induction in a reporter assay that detects only strong inducers of the gamma-globin gene promoter and in cultured human erythroid progenitors. To elucidate the molecular mechanisms used by high-potency SCFADs, chromatin immunoprecipitation (ChIP) assays performed at the human gamma- and beta-globin gene promoters in GM979 cells and in erythroid progenitors demonstrate that RB7 and butyrate induce dissociation of HDAC3 (but not HDAC1 or HDAC2) and its adaptor protein NCoR, specifically from the gamma-globin gene promoter. A coincident and proportional recruitment of RNA polymerase II to the gamma-globin gene promoter was observed with exposure to these gamma-globin inducers. Knockdown of HDAC3 by siRNA induced transcription of the gamma-globin gene promoter, demonstrating that displacement of HDAC3 from the gamma-globin gene promoter by the SCFAD is sufficient to induce gamma-globin gene expression. These studies demonstrate new dynamic alterations in transcriptional regulatory complexes associated with SCFAD-induced activation of the gamma-globin gene and provide a specific molecular target for potential therapeutic intervention.


Subject(s)
Fatty Acids, Volatile/pharmacology , Gene Expression Regulation/drug effects , Globins/genetics , Histone Deacetylases/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Adult , DNA Primers , Globins/drug effects , Histone Deacetylases/drug effects , Humans , K562 Cells , Nuclear Proteins/drug effects , Nuclear Receptor Co-Repressor 1 , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Messenger/genetics , Repressor Proteins/drug effects , Transfection
10.
Chem Biol Drug Des ; 67(5): 318-28, 2006 May.
Article in English | MEDLINE | ID: mdl-16784456

ABSTRACT

Pharmacologic reinduction of the developmentally silenced fetal (gamma) globin genes has been achieved in hemoglobinopathy patients using short chain fatty acid derivatives, with therapeutic effects. However, higher-potency inducers than are available in currently identified short chain fatty acid derivatives are desirable for long-term use. Using several short-chain fatty acids with established gamma-globin induction activity, a pharmacophore template was constructed with the TFIT module of the flo software and used to select several new candidate compounds, three of which exhibited significant activity in a gamma-globin gene reporter transcriptional assay which detects only strong inducers. The data were used to construct a new pharmacophore and a 'pseudo' receptor around it. Six hundred and thirty low-molecular weight compounds were docked into this receptor model. Of 26 compounds selected and tested in functional assays, two compounds showed activity >500% over control levels and two had activity 200% over control range, significantly more active than previously identified short chain fatty acid derivative fetal globin gene inducers. Three compounds had less activity; the remainder showed moderate activity. These findings demonstrate the feasibility of using iterative construction of pharmacophores, pseudo-binding site modeling, and virtual screening to identify small molecules with the ability to induce transcription of specific target genes, for potential therapeutics.


Subject(s)
Drug Design , Fatty Acids/pharmacology , Fetal Hemoglobin/genetics , Models, Molecular , Transcriptional Activation , Animals , Binding Sites , Butyrates/chemistry , Butyrates/pharmacology , Cell Line, Tumor , Leukemia, Erythroblastic, Acute/metabolism , Luciferases , Mice , Molecular Structure , Phenylbutyrates/chemistry , Phenylbutyrates/pharmacology
11.
Ann N Y Acad Sci ; 1054: 257-65, 2005.
Article in English | MEDLINE | ID: mdl-16339673

ABSTRACT

Accelerated apoptosis of erythroid progenitors in beta-thalassemia is a significant barrier to definitive therapy because the beneficial effects of fetal globin-inducing agents on globin chain balance may not be inducible in cells in which programmed cell death is established early. Accordingly, our objectives have been to identify methods to decrease cellular apoptosis and to identify orally tolerable fetal globin gene inducers. A pilot clinical trial was conducted to determine whether combined use of a fetal globin gene inducer (butyrate) and rhu-erythropoietin (EPO), the hematopoietic growth factor that prolongs erythroid cell survival and stimulates erythroid proliferation, would produce additive hematologic responses in any thalassemia subjects. Butyrate and EPO were administered in 10 patients. Novel fetal globin gene inducers that also stimulate erythroid proliferation were evaluated for pharmacokinetic profiles. Patients with beta+-thalassemia had relatively low levels of endogenous EPO (<145 mU/mL) and had additive responses to administered EPO and butyrate. Patients with at least one beta 0-globin mutation had higher baseline HbF levels (>60%) and EPO levels (>160 mU/mL), and three-fourths of these subjects responded to the fetal globin gene inducer alone. A few select fetal globin-inducing short-chain fatty acid derivatives that stimulated cell proliferation also had favorable pharmacokinetics. These studies identify a significant subset of thalassemia patients who appear to require exogenous EPO to respond optimally to any HbF inducer, as well as new therapeutic candidates that act on both cellular and molecular pathologies of beta-thalassemia. Both approaches now offer excellent potential for tolerable, definitive treatment of beta-thalassemia.


Subject(s)
Butyrates/therapeutic use , Erythroid Cells/drug effects , Erythropoietin/therapeutic use , Fetal Hemoglobin/biosynthesis , Gene Expression/drug effects , beta-Thalassemia/drug therapy , Animals , Apoptosis/drug effects , Blood Transfusion , Butyrates/administration & dosage , Cells, Cultured/drug effects , Combined Modality Therapy , Drug Evaluation, Preclinical , Drug Therapy, Combination , Erythroid Cells/metabolism , Erythropoietin/administration & dosage , Fatty Acids, Volatile/pharmacokinetics , Fatty Acids, Volatile/pharmacology , Fetal Hemoglobin/genetics , Humans , Papio , Pilot Projects , Recombinant Proteins , Treatment Outcome , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/therapy
12.
Blood Cells Mol Dis ; 35(2): 217-26, 2005.
Article in English | MEDLINE | ID: mdl-16099182

ABSTRACT

Accelerated apoptosis of erythroid progenitors is a characteristic of beta-thalassemia which presents a significant barrier to definitive therapeutic approaches utilizing induction of endogenous fetal globin gene expression. gamma-globin gene expression may not be inducible in, or may not be able to rescue, erythroid cells in which programmed cell death is initiated early in erythroblast development. In this report, short-chain fatty acid derivatives (SCFADs) which induce fetal globin gene expression were tested for their ability to promote proliferation and survival of erythroid progenitors cultured from beta-thalassemic subjects, and of cytokine-dependent erythroid cell lines. Certain SCFADs promoted thalassemic Bfu-e growth and cytokine-independent growth and survival of erythroid cell lines. A 40-80% increase in erythroid Bfu-e colony number was observed in cultures established with any of five mitogenic SCFADs, compared to control or butyrate-treated cultures from the same subjects. Immunoblot analysis demonstrated that these same SCFADs also regulated the expression of specific protein inhibitors of apoptosis. Anti-apoptotic ratios of the proteins Bcl-xL/Bcl-xS in thalassemic Bfu-e were increased by 30-120% with exposure to the SCFDs, compared to the ratios in the same cells cultured under control conditions. Similar anti-apoptotic increases in Mcl-1L/Mcl-1S ratios were induced by the SCFADs. These findings suggest that select fetal globin-inducing SCFADs which enhance proliferation of beta-thalassemia progenitors may enhance survival of these progenitors by altering levels of Bcl-family protein members. This combination of effects should enhance erythroid cell survival in the beta-thalassemia syndromes, allowing fetal globin gene expression to be induced more effectively than currently available, growth-suppressing, fetal globin-inducing agents, such as the butyrates or chemotherapeutic agents.


Subject(s)
Erythroid Precursor Cells/pathology , Fatty Acids, Volatile/pharmacology , bcl-X Protein/genetics , beta-Thalassemia/blood , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/pharmacology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Gene Expression Regulation/drug effects , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/analysis , Neoplasm Proteins/drug effects , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/drug effects , bcl-X Protein/analysis , beta-Thalassemia/genetics
13.
Leuk Res ; 28(11): 1125-36, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15380335

ABSTRACT

Six patients with bcr-abl positive AML or chronic myelogenous leukemia in blast crisis (CML-BC) were treated with the IMP-dehydrogenase (IMPDH) inhibitor, Tiazofurin, in a Phase-II trial. Tiazofurin was given by IV infusion (2200-2700 mg/m2 per day) for up to 10 days. Leukemia blasts rapidly disappeared from the circulation of patients during treatment, while mature myeloid cells in the marrow increased in number. Although these hematologic responses were transient, persisting less than 3-4 weeks, our findings confirm that Tiazofurin has anti-leukemia activity. This drug warrants further study in combination regimens with other chemotherapeutic agents for the treatment of bcr-abl positive AML and CML-BC.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Genes, abl , IMP Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Ribavirin/analogs & derivatives , Ribavirin/therapeutic use , Adult , Aged , Female , Humans , Male , Middle Aged , Treatment Outcome
14.
Leuk Res ; 28(11): 1137-43, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15380336

ABSTRACT

Inosine-5'-monophosphate-dehydrogenase (IMPDH) regulates the de novo synthesis of guanine ribonucleotides (GNT). IMPDH activity varies inversely with intracellular [GNT] and is linked to cellular proliferation. K562 leukemia cell growth was studied relative to IMPDH expression and activity following culture of the cells with Tiazofurin, an IMPDH inhibitor. Tiazofurin depressed IMPDH activity and [GTP] in K562 cells, and also increased IMPDH mRNA expression. Following exposure to Tiazofurin, K562 cell proliferation, entry into cycle, and sensitivity to cycle-active cytotoxic agents were increased. These findings indicate that the efficacy of standard chemotherapy in bcr-abl positive leukemias might be enhanced if combined sequentially with Tiazofurin.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Ribavirin/analogs & derivatives , Ribavirin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , In Vitro Techniques , K562 Cells , RNA, Messenger/genetics
15.
Int J Cancer ; 110(5): 741-50, 2004 Jul 10.
Article in English | MEDLINE | ID: mdl-15146564

ABSTRACT

Combinatorial Ig libraries with phage display allow in vitro generation of human Ig fragments without the need to maintain hybridomas in ongoing cell culture or to select circulating Ig from human serum. Identifying tumor-associated antigens on the surface of intact tumor cells, as opposed to purified proteins, presents a challenge due to the difficulty of preserving complex 3-D epitopic sites on the cell surface, the variable expression of antigens on different malignant cell types and the stereotactic interference of closely associated proteins on the intact membrane surface limiting accessibility to antigenic sites. A combinatorial Ig library of 10(10) clones was generated from the cDNA of PBMCs derived from patients with breast adenocarcinoma. Following subtractive panning, the library was enriched for Ig (Fab fragment) binding to intact adenocarcinoma cells and the resultant Fabs were screened against a cDNA expression library, itself generated from breast cancer cells. Using this approach, we isolated clones from the cDNA library expressing gC1q-R, a glycoprotein comprising the major structure of C1, the first component of the complement system. gC1q-R is a 33 kDa glycoprotein expressed not only on the cell surface but also intracellularly, with motifs that target it to mitochondria and complete homology with HABP and human HeLa cell protein p32, which is copurified with pre-mRNA SF2. Sequencing of the gene encoding tumor-associated gC1q-R did not reveal any consistent tumor-specific mutations. However, histochemical staining with anti-gC1q-R MAb demonstrated marked differential expression of gC1q-R in thyroid, colon, pancreatic, gastric, esophageal and lung adenocarcinomas compared to their nonmalignant histologic counterparts. In contrast, differential expression was not seen in endometrial, renal and prostate carcinomas. Despite high expression in breast carcinoma, gC1q-R was also expressed in nonmalignant breast tissue. Although the precise relation of gC1q-R to carcinogenesis remains unclear, our finding of tumor overexpression and the known multivalent binding of gC1q-R to not only C1q itself but also a variety of circulating plasma proteins as well as its involvement in cell-to-cell interactions suggest that gC1q-R may have a role in tumor metastases and potentially serve in molecule-specific targeting of malignant cells.


Subject(s)
Adenocarcinoma/metabolism , Complement C1q/biosynthesis , Hyaluronan Receptors/biosynthesis , Membrane Glycoproteins , Receptors, Complement/biosynthesis , Base Sequence , Blotting, Northern , Breast Neoplasms/metabolism , Carrier Proteins , Cell Line, Tumor , Cell Membrane/metabolism , DNA, Complementary/metabolism , Epitopes , Gene Library , Humans , Immunohistochemistry , Mitochondrial Proteins , Models, Biological , Molecular Sequence Data , Neoplasm Metastasis , Peptide Library , Protein Binding , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid
16.
Blood ; 100(13): 4640-8, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12393583

ABSTRACT

Orally bioactive compounds that induce gamma globin gene expression at tolerable doses are needed for optimal treatment of the beta-hemoglobinopathies. Short-chain fatty acids (SCFAs) of 2 to 6 carbons in length induce gamma globin expression in animal models, and butyrate, phenylbutyrate, and valproate induce gamma globin in human patients. The usefulness of these compounds, however, is limited by requirements for large doses because of their rapid metabolism and their tendency to inhibit cell proliferation, which limits the pool of erythroid progenitors in which gamma globin can be induced. Selected short-chain fatty acid derivatives (SCFADs) were recently found to induce gamma globin and to stimulate the proliferation of hematopoietic cells in vitro. These SCFADs are now evaluated in vivo in nonanemic transgenic mice containing the human beta globin gene locus and in anemic phlebotomized baboons. In mice treated with a SCFAD once daily for 5 days, gamma globin mRNA increased 2-fold, reticulocytes increased 3- to 7-fold, and hematocrit levels increased by 27%. Administration of 3 SCFADs in anemic baboons increased F-reticulocytes 2- to 15-fold over baseline and increased total hemoglobin levels by 1 to 2 g/dL per week despite ongoing significant daily phlebotomy. Pharmacokinetic studies demonstrated 90% oral bioavailability of 2 SCFADs, and targeted plasma levels were maintained for several hours after single oral doses equivalent to 10% to 20% of doses required for butyrate. These findings identify SCFADs that stimulate gamma globin gene expression and erythropoiesis in vivo, activities that are synergistically beneficial for treatment of the beta hemoglobinopathies and useful for the oral treatment of other anemias.


Subject(s)
Erythropoiesis/drug effects , Fatty Acids, Volatile/pharmacology , Gene Expression Regulation/drug effects , Globins/biosynthesis , Anemia/genetics , Anemia/metabolism , Animals , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chromosomes, Artificial, Yeast , Drug Evaluation, Preclinical , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Fatty Acids, Volatile/pharmacokinetics , Genes, Reporter , Globins/genetics , Half-Life , Humans , Luciferases/biosynthesis , Luciferases/genetics , Mice , Mice, Transgenic , Papio , Phenylpropionates/pharmacology , Promoter Regions, Genetic , Recombinant Proteins , Reticulocyte Count , Transfection
17.
Cancer Res ; 62(17): 4985-91, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12208750

ABSTRACT

Molecules differentially expressed or overexpressed by malignant cells can serve in detecting and tracking of tumor. Additionally, they potentially can be applied in histologic-specific antitumor therapy. Few breast cancer-associated candidate molecules have been identified. Here we describe the use of combinatorial immunoglobulin [antigen-binding fragment of immunoglobulin molecule (Fab) fragment] phage libraries generated from patients with breast carcinoma to identify cancer-associated gene expression. The libraries were enriched for tumor-binding Fab by 3 logs and yielded a group of antibodies against DNA-binding protein B (DbpB), a 35-kDa thrombin-inducible nuclear factor and member of the Y-box family of proteins, which are known to act both negatively in selective gene suppression and positively as promoters of gene transcription. Sequencing of the anti-DbpB showed a degree of heterogeneity and bp substitutions suggesting that the Fabs selected from the combinatorial library represented a varied anti-DbpB immune response and did not simply arise from in vitro amplification by PCR of a single or limited numbers of immunoglobulin genes. Sequencing of the DbpB molecule expressed in malignant breast cancer showed no evidence of tumor-specific mutations. Evaluation of levels of DbpB gene product expression however showed the molecule to be constitutively expressed in normal nonmalignant breast tissues but to have consistently differentially higher expression in breast cancer. Immunohistological staining revealed DbpB to be present both intracellularly and on the cell surface, which suggests it may be a means whereby malignant cells repair and replicate DNA in a selectively advantageous manner as compared with nonmalignant cells. DbpB expression in breast cancer may advance the basic understanding of the role of Y-box binding proteins as regulatory agents, and in defining malignant cell phenotypes. In addition, DbpB and the antibodies generated against it may have direct application in tumor detection and in molecule-targeted immunotherapy.


Subject(s)
Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Proteins/biosynthesis , DNA-Binding Proteins , Immunoglobulins/genetics , Transcription Factors , Amino Acid Sequence , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Combinatorial Chemistry Techniques , DNA, Complementary/genetics , Genes, Immunoglobulin , Genomic Library , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulins/metabolism , Molecular Sequence Data , NFI Transcription Factors , Nuclear Proteins , Peptide Library , Y-Box-Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...