Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Org Biomol Chem ; 11(20): 3341-8, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23563530

ABSTRACT

Natural products of the caged Garcinia xanthones (CGX) family are characterized by a unique chemical structure, potent bioactivities and promising pharmacological profiles. We have developed a Claisen/Diels-Alder reaction cascade that, in combination with a Pd(0)-catalyzed reverse prenylation, provides rapid and efficient access to the CGX pharmacophore, represented by the structure of cluvenone. To further explore this pharmacophore, we have synthesized various A-ring oxygenated analogues of cluvenone and have evaluated their bioactivities in terms of growth inhibition, mitochondrial fragmentation, induction of mitochondrial-dependent cell death and Hsp90 client inhibition. We found that installation of an oxygen functionality at various positions of the A-ring influences significantly both the site-selectivity of the Claisen/Diels-Alder reaction and the bioactivity of these compounds, due to remote electronic effects.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Garcinia/chemistry , Oxygen/chemistry , Xanthones/chemical synthesis , Xanthones/pharmacology , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Mitochondria/drug effects , Molecular Structure , Structure-Activity Relationship , Xanthones/chemistry
2.
Chembiochem ; 13(8): 1191-8, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22532297

ABSTRACT

The natural product gambogic acid (GA) has shown significant potential as an anticancer agent as it is able to induce apoptosis in multiple tumor cell lines, including multidrug-resistant cell lines, as well as displaying antitumor activity in animal models. Despite the fact that GA has entered phase I clinical trials, the primary cellular target and mode of action of this compound remain unclear, although many proteins have been shown to be affected by it. By thorough analysis of several cellular organelles, at both the morphological and functional levels, we demonstrate that the primary effect of GA is at the mitochondria. We found that GA induces mitochondrial damage within minutes of incubation at low-micromolar concentrations. Moreover, a fluorescent derivative of GA was able to localize specifically to the mitochondria and was displaced from these organelles after competition with unlabeled GA. These findings indicate that GA directly targets the mitochondria to induce the intrinsic pathway of apoptosis, and thus represents a new member of the mitocans.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Xanthones/pharmacology , Xanthones/pharmacokinetics , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , HeLa Cells , Humans , Subcellular Fractions/metabolism , Xanthones/chemistry
3.
Invest New Drugs ; 30(5): 1841-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21898184

ABSTRACT

The synthetic caged Garcinia xanthone, cluvenone, has potent and selective cytotoxicity against numerous cancer cell lines including those that are multi-drug resistant. The direct target of this structurally and functionally unique agent is unknown and that of the parent natural product, gambogic acid (GA), presently in clinical trials, is not yet entirely clear. For the first time, using fluorescently labeled GA (GA-Bodipy), we determined that GA-Bodipy localized in mitochondria and was effectively displaced by cluvenone in competition experiments indicating that the direct target of cluvenone resided in mitochondria and was shared by GA. In agreement with these findings, treatment of HeLa cells with cluvenone or GA resulted in disruption of mitochondrial morphology within 4 h. Furthermore, experiments using the potential sensitive JC-1 dye demonstrated that cells treated with 1 µM cluvenone for 1 h had significant loss of MMP compared to control cells. Examination of Cyt c levels in leukemia cells treated with 1 µM cluvenone resulted in a 4-fold increase in levels of both cytosolic and mitochondrial Cyt c. In agreement with Cyt c release, caspase 9 activity was increased 2.6-fold after treatment of cells for 5 h with 1 µM cluvenone. Remarkably, the caspase-9 inhibitor, Z-LEHD-FMK, blocked cluvenone-induced apoptosis in a dose-dependent manner with apoptosis being completely blocked by 10 µM of the inhibitor. In conclusion, cluvenone, an agent with potent cytotoxicity against multi-drug resistant tumor cells, has direct targets in mitochondria thus setting precedence for drug discovery efforts against these targets in the treatment of refractory cancers.


Subject(s)
Apoptosis/drug effects , Mitochondria/drug effects , Xanthones/pharmacology , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Drug Resistance, Neoplasm , HeLa Cells , Humans , Leukemia/drug therapy , Leukemia/metabolism
4.
Clin Cancer Res ; 27(8): 2179-2189, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33504555

ABSTRACT

PURPOSE: Previously our randomized phase III trial demonstrated that immunotherapy including dinutuximab, a chimeric anti-GD2 mAb, GM-CSF, and IL2 improved survival for children with high-risk neuroblastoma that had responded to induction and consolidation therapy. These results served as the basis for FDA approval of dinutuximab. We now present long-term follow-up results and evaluation of predictive biomarkers. PATIENTS AND METHODS: Patients recieved six cycles of isotretinoin with or without five cycles of immunotherapy which consists of dinutuximab with GM-CSF alternating with IL2. Accrual was discontinued early due to meeting the protocol-defined stopping rule for efficacy, as assessed by 2-year event-free survival (EFS). Plasma levels of dinutuximab, soluble IL2 receptor (sIL2R), and human anti-chimeric antibody (HACA) were assessed by ELISA. Fcγ receptor 2A and 3A genotypes were determined by PCR and direct sequencing. RESULTS: For 226 eligible randomized patients, 5-year EFS was 56.6 ± 4.7% for patients randomized to immunotherapy (n = 114) versus 46.1 ± 5.1% for those randomized to isotretinoin only (n = 112; P = 0.042). Five-year overall survival (OS) was 73.2 ± 4.2% versus 56.6 ± 5.1% for immunotherapy and isotretinoin only patients, respectively (P = 0.045). Thirteen of 122 patients receiving dinutuximab developed HACA. Plasma levels of dinutuximab, HACA, and sIL2R did not correlate with EFS/OS, or clinically significant toxicity. Fcγ receptor 2A and 3A genotypes did not correlate with EFS/OS. CONCLUSIONS: Immunotherapy with dinutuximab improved outcome for patients with high-risk neuroblastoma. Early stoppage for efficacy resulted in a smaller sample size than originally planned, yet clinically significant long-term differences in survival were observed.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Interleukin-2/administration & dosage , Neuroblastoma/drug therapy , Adolescent , Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Follow-Up Studies , Humans , Infant , Interleukin-2/adverse effects , Isotretinoin/administration & dosage , Isotretinoin/adverse effects , Male , Neuroblastoma/mortality , Progression-Free Survival , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects
5.
Chemistry ; 16(33): 9944-62, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20648491

ABSTRACT

Natural products have been a great source of many small molecule drugs for various diseases. In spite of recent advances in biochemical engineering and fermentation technologies that allow us to explore microorganisms and the marine environment as alternative sources of drugs, more than 70 % of the current small molecule therapeutics derive their structures from plants used in traditional medicine. Natural-product-based drug discovery relies heavily on advances made in the sciences of biology and chemistry. Whereas biology aims to investigate the mode of action of a natural product, chemistry aims to overcome challenges related to its supply, bioactivity, and target selectivity. This review summarizes the explorations of the caged Garcinia xanthones, a family of plant metabolites that possess a unique chemical structure, potent bioactivities, and a promising pharmacology for drug design and development.


Subject(s)
Garcinia/chemistry , Xanthones/chemistry , Xanthones/pharmacology , Animals , Humans , Plant Extracts/chemistry , Plant Extracts/pharmacology , Structure-Activity Relationship , Trees/chemistry
6.
Org Biomol Chem ; 7(23): 4886-94, 2009 Dec 07.
Article in English | MEDLINE | ID: mdl-19907779

ABSTRACT

The combination of unique structure and potent bioactivity exhibited by several family members of the caged Garcinia xanthones, led us to evaluate their pharmacophore. We have developed a Pd(0)-catalyzed method for the reverse prenylation of catechols that, together with a Claisen/Diels-Alder reaction cascade, provides rapid and efficient access to various caged analogues. Evaluation of the growth inhibitory activity of these compounds leads to the conclusion that the intact ABC ring system containing the C-ring caged structure is essential to the bioactivity. Studies with cluvenone (7) also showed that these compounds induce apoptosis and exhibit significant cytotoxicity in multidrug-resistant leukemia cells. As such, the caged Garcinia xanthone motif represents a new and potent pharmacophore.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Drug Evaluation , Garcinia/chemistry , Xanthones/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Xanthones/chemical synthesis , Xanthones/chemistry
7.
PLoS One ; 12(3): e0172632, 2017.
Article in English | MEDLINE | ID: mdl-28296891

ABSTRACT

Renal cell carcinoma (RCC) is among the top ten most common forms of cancer and is the most common malignancy of the kidney. Clear cell renal carcinoma (cc-RCC), the most common type of RCC, is one of the most refractory cancers with an incidence that is on the rise. Screening of plant extracts in search of new anti-cancer agents resulted in the discovery of englerin A, a guaiane sesquiterpene with potent cytotoxicity against renal cancer cells and a small subset of other cancer cells. Though a few cellular targets have been identified for englerin A, it is still not clear what mechanisms account for the cytotoxicity of englerin A in RCC, which occurs at concentrations well below those used to engage the targets previously identified. Unlike any prior study, the current study used a systems biology approach to explore the mechanism(s) of action of englerin A. Metabolomics analyses indicated that englerin A profoundly altered lipid metabolism by 24 h in cc-RCC cell lines and generated significant levels of ceramides that were highly toxic to these cells. Microarray analyses determined that englerin A induced ER stress signaling and an acute inflammatory response, which was confirmed by quantitative PCR and Western Blot analyses. Additionally, fluorescence confocal microscopy revealed that englerin A at 25 nM disrupted the morphology of the ER confirming the deleterious effect of englerin A on the ER. Collectively, our findings suggest that cc-RCC is highly sensitive to disruptions in lipid metabolism and ER stress and that these vulnerabilities can be targeted for the treatment of cc-RCC and possibly other lipid storing cancers. Furthermore, our results suggest that ceramides may be a mediator of some of the actions of englerin A. Lastly, the acute inflammatory response induced by englerin A may mediate anti-tumor immunity.


Subject(s)
Carcinoma, Renal Cell/metabolism , Endoplasmic Reticulum Stress/drug effects , Inflammation/chemically induced , Kidney Neoplasms/metabolism , Lipid Metabolism/drug effects , Sesquiterpenes, Guaiane/pharmacology , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/pathology
8.
PLoS One ; 9(2): e88219, 2014.
Article in English | MEDLINE | ID: mdl-24505435

ABSTRACT

The INK4A locus codes for two independent tumor suppressors, p14ARF and p16/CDKN2A, and is frequently mutated in many cancers. Here we report a novel deletion/substitution from CC to T in the shared exon 2 of p14ARF/p16 in a melanoma cell line. This mutation aligns the reading frames of p14ARF and p16 mid-transcript, producing one protein which is half p14ARF and half p16, chimera ARF (chARF), and another which is half p16 and half non-p14ARF/non-p16 amino acids, p16-Alternate Carboxyl Terminal (p16-ACT). In an effort to understand the cellular impact of this novel mutation and others like it, we expressed the two protein products in a tumor cell line and analyzed common p14ARF and p16 pathways, including the p53/p21 and CDK4/cyclin D1 pathways, as well as the influence of the two proteins on growth and the cell cycle. We report that chARF mimicked wild-type p14ARF by inducing the p53/p21 pathway, inhibiting cell growth through G2/M arrest and maintaining a certain percentage of cells in G1 during nocodazole-induced G2 arrest. chARF also demonstrated p16 activity by binding CDK4. However, rather than preventing cyclin D1 from binding CDK4, chARF stabilized this interaction through p21 which bound CDK4. p16-ACT had no p16-related function as it was unable to inhibit cyclin D1/CDK4 complex formation and was unable to arrest the cell cycle, though it did inhibit colony formation. We conclude that these novel chimeric proteins, which are very similar to predicted p16/p14ARF chimeric proteins found in other primary cancers, result in maintained p14ARF-p53-p21 signaling while p16-dependent CDK4 inhibition is lost.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Melanoma/genetics , Mutant Chimeric Proteins/genetics , Tumor Suppressor Protein p14ARF/genetics , Base Sequence , Cell Cycle , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Exons , Humans , Melanoma/metabolism , Melanoma/pathology , Mutant Chimeric Proteins/metabolism , Mutation , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism
9.
Cancer Res ; 74(14): 3753-63, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25028366

ABSTRACT

Neuroblastoma is a pediatric cancer with significant genomic and biologic heterogeneity. p16 and ARF, two important tumor-suppressor genes on chromosome 9p21, are inactivated commonly in most cancers, but paradoxically overexpressed in neuroblastoma. Here, we report that exon γ in p16 is also part of an undescribed long noncoding RNA (lncRNA) that we have termed CAI2 (CDKN2A/ARF Intron 2 lncRNA). CAI2 is a single-exon gene with a poly A signal located in but independent of the p16/ARF exon 3. CAI2 is expressed at very low levels in normal tissue, but is highly expressed in most tumor cell lines with an intact 9p21 locus. Concordant expression of CAI2 with p16 and ARF in normal tissue along with the ability of CAI2 to induce p16 expression suggested that CAI2 may regulate p16 and/or ARF. In neuroblastoma cells transformed by serial passage in vitro, leading to more rapid proliferation, CAI2, p16, and ARF expression all increased dramatically. A similar relationship was also observed in primary neuroblastomas where CAI2 expression was significantly higher in advanced-stage neuroblastoma, independently of MYCN amplification. Consistent with its association with high-risk disease, CAI2 expression was also significantly associated with poor clinical outcomes, although this effect was reduced when adjusted for MYCN amplification. Taken together, our findings suggested that CAI2 contributes to the paradoxical overexpression of p16 in neuroblastoma, where CAI2 may offer a useful biomarker of high-risk disease.


Subject(s)
Chromosomes, Human, Pair 9 , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Neuroblastoma/pathology , RNA, Long Noncoding/genetics , ADP-Ribosylation Factors/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p16/genetics , Follow-Up Studies , Gene Expression , Gene Order , Humans , Infant , Infant, Newborn , Neoplasm Staging , Neuroblastoma/mortality , Prognosis , RNA, Messenger/genetics
10.
J Exp Clin Cancer Res ; 32: 57, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23958461

ABSTRACT

Renal cell carcinoma (RCC), the most common malignancy of the kidney, is refractory to standard therapy and has an incidence that continues to rise. Screening of plant extracts in search of new agents to treat RCC resulted in the discovery of englerin A (EA), a natural product exhibiting potent selective cytotoxicity against renal cancer cells. Despite the establishment of synthetic routes to the synthesis of EA, very little is known about its mechanism of action. The results of the current study demonstrate for the first time that EA induces apoptosis in A498 renal cancer cells in addition to necrosis. The induction of apoptosis by EA required at least 24 h and was caspase independent. In addition, EA induced increased levels of autophagic vesicles in A498 cells which could be inhibited by nonessential amino acids (NEAA), known inhibitors of autophagy. Interestingly, inhibition of autophagy by NEAA did not diminish cell death suggesting that autophagy is not a cell death mechanism and likely represents a cell survival mechanism which ultimately fails. Apart from cell death, our results demonstrated that cells treated with EA accumulated in the G2 phase of the cell cycle indicating a block in G2/M transition. Moreover, our results determined that EA inhibited the activation of both AKT and ERK, kinases which are activated in cancer and implicated in unrestricted cell proliferation and induction of autophagy. The phosphorylation status of the cellular energy sensor, AMPK, appeared unaffected by EA. The high renal cancer selectivity of EA combined with its ability to induce multiple mechanisms of cell death while inhibiting pathways driving cell proliferation, suggest that EA is a highly unique agent with great potential as a therapeutic lead for the treatment of RCC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Sesquiterpenes, Guaiane/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Phosphorylation
11.
Chem Asian J ; 7(5): 1052-60, 2012 May.
Article in English | MEDLINE | ID: mdl-22415793

ABSTRACT

An efficient formal synthesis of (-)-englerin A (1) is reported. The target molecule is a recently isolated guaiane sesquiterpene that possesses highly potent and selective activity against renal cancer cell-lines. Our enantioselective strategy involved the construction of the BC ring system of compound 1 through a Rh(II)-catalyzed [4+3] cycloaddition reaction followed by subsequent attachment of the A ring through an intramolecular aldol condensation reaction. As such, this strategy allows the synthesis of truncated englerins. Evaluation of these analogues with the A498 renal cancer cell-line suggested that the A ring of englerin is crucial to its antiproliferative activity. Moreover, evaluation of these analogues led to the identification of potent growth-inhibitors of CEM cells with GI(50) values in the range 1-3 µM.


Subject(s)
Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Phyllanthus/chemistry , Sesquiterpenes, Guaiane/chemical synthesis , Sesquiterpenes, Guaiane/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Catalysis , Cell Line, Tumor , Cyclization , Humans , Inhibitory Concentration 50 , Kidney Neoplasms/drug therapy , Models, Molecular , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Rhodium/chemistry , Sesquiterpenes, Guaiane/chemistry , Stereoisomerism
12.
Mol Cancer Ther ; 9(11): 2869-78, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20881270

ABSTRACT

Several caged Garcinia xanthone natural products have potent bioactivity and a documented value in traditional Eastern medicine. Previous synthesis and structure activity relationship studies of these natural products resulted in the identification of the pharmacophore represented by the structure of cluvenone. In the current study, we examined the anticancer activity of cluvenone and conducted gene expression profiling and pathway analyses. Cluvenone was found to induce apoptosis in T-cell acute lymphoblastic leukemia cells (EC50 = 0.25 µmol/L) and had potent growth-inhibitory activity against the NCI60 cell panel, including those that are multidrug-resistant, with a GI50 range of 0.1 to 2.7 µmol/L. Importantly, cluvenone was approximately 5-fold more potent against a primary B-cell acute lymphoblastic leukemia compared with peripheral blood mononuclear cells from normal donors, suggesting that it has significant tumor selectivity. Comparison of cluvenone's growth-inhibitory profile to those in the National Cancer Institute database revealed that compounds with a similar profile to cluvenone were mechanistically unlike known agents, but were associated with cell stress and survival signaling. Gene expression profiling studies determined that cluvenone induced the activation of mitogen-activated protein kinase and NrF2 stress response pathways. Furthermore, cluvenone was found to induce intracellular reactive oxygen species formation. Lastly, the modulation in the expression of several genes associated with T cell and natural killer cell activation and function by cluvenone suggests a role as an immune-modulator. The current work highlights the potential of cluvenone as a chemotherapeutic agent and provides support for further investigation of these intriguing molecules with regard to mechanism and targets.


Subject(s)
Apoptosis/drug effects , Cells/drug effects , Immunomodulation/drug effects , Oxidative Stress/drug effects , Xanthones/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cells/immunology , Cells/metabolism , Drug Evaluation, Preclinical , Garcinia/chemistry , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Immunologic Factors/chemical synthesis , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Microarray Analysis , Models, Biological , Tumor Cells, Cultured , Xanthones/chemical synthesis , Xanthones/chemistry
14.
Org Biomol Chem ; 5(3): 494-500, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-17252132

ABSTRACT

Inspired by the combination of unique structure and potent bioactivities exhibited by several family members of the caged Garcinia xanthones, we developed a synthesis of simplified analogues that maintain the overall caged motif. The caged structure of these compounds was constructed via a site-selective Claisen/Diels-Alder reaction cascade. We found that the fully substituted caged structure, in which are included the C18 and C23 geminal methyl groups, is necessary to maintain bioactivity. Analogue had comparable activity to the natural products of this family, such as gambogic acid. These compounds exhibit cytotoxicity in a variety of tumor cell lines at low micromolar concentrations and were found to induce apoptosis in HUVE cells. In addition, studies with HL-60 and HL-60/ADR cells indicate that these compounds are not affected by the mechanisms of multidrug resistance, conferred by P glycoprotein expression, typical of relapsed cancers and thus represent a new and potent pharmacophore.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Garcinia/chemistry , Xanthones/chemical synthesis , Xanthones/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Alkanes/chemistry , Biological Products/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Multiple , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Structure-Activity Relationship
15.
Blood ; 107(3): 898-903, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16234352

ABSTRACT

The deficiency of methylthioadenosine phosphorylase (MTAP) in T-cell acute lymphoblastic leukemia (T-ALL) and other cancers, while constitutively expressed in normal cells, allows for selective therapy using L-alanosine, an inhibitor of de novo AMP synthesis. We demonstrate that MTAP- T-ALL cells obtained at relapse are as sensitive to L-alanosine toxicity as diagnosis samples. The therapeutic index of L-alanosine can be increased by the use of a MTAP substrate, which protects MTAP+ normal cells. Since MTAP substrates MTA and 5'deoxyadenosine are prone to toxicities associated with adenosine, we synthesized and evaluated a potentially nontoxic MTAP substrate, 9-beta-D-erythrofuranosyladenine (EFA). The cytotoxicity of EFA to hematopoietic progenitors erythroid burst-forming units (BFU-Es) and granulocyte-macrophage colony-forming units (CFU-GMs) was at least 26- to 41-fold less than that of MTA. In addition, EFA selectively rescued MTAP+ MOLT-4 cells from L-alanosine toxicity at 25 microM with negligible toxicity even at 100 microM. As for MTA, significant, albeit incomplete, rescue was achieved at 12.5 microM, but higher concentrations were toxic. EFA at 20 microM or less rescued primary MTAP+ T-ALL cells and normal lymphocytes from L-alanosine toxicity. Collectively, these data indicate that EFA is an effective agent for salvaging MTAP+ cells from L-alanosine toxicity and is superior to MTA due to lower cytotoxicity.


Subject(s)
Adenine/analogs & derivatives , Antibiotics, Antineoplastic/pharmacology , Enzyme Inhibitors/pharmacology , Furans/pharmacology , Leukemia-Lymphoma, Adult T-Cell/enzymology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Adenine/pharmacology , Adenine/therapeutic use , Alanine/adverse effects , Alanine/analogs & derivatives , Alanine/pharmacology , Alanine/therapeutic use , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Deoxyadenosines/pharmacology , Deoxyadenosines/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Erythroid Precursor Cells/enzymology , Erythroid Precursor Cells/pathology , Furans/therapeutic use , Granulocyte Precursor Cells/enzymology , Granulocyte Precursor Cells/pathology , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Purine-Nucleoside Phosphorylase/deficiency , Salvage Therapy , Thionucleosides/pharmacology , Thionucleosides/therapeutic use
16.
Genes Chromosomes Cancer ; 41(4): 297-308, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15390183

ABSTRACT

Despite considerable research into the etiology of neuroblastoma, the molecular basis of this disease has remained elusive. In contrast to the absence of expression of the known tumor suppressor CDKN2A (also known as p16 and INK4A) in a wide variety of tumor types we have found in previous studies that CDKN2A protein is paradoxically highly expressed in many advanced stage neuroblastomas and unrelated to RB1 status. In the present study, we sought to identify the mechanistic relationships that might influence CDKN2A expression and negate its influence on tumor cell proliferation. In this regard, we examined the role of the tumor-suppressor gene CDKN1B (also known as p27 and Kip1) and the oncogene ID2 in relationship to CDKN2A expression, MYCN amplification, and neuroblastoma pathogenesis in 17 neuroblastoma cell lines and 129 samples of primary tumors of all stages. All neuroblastoma cell lines expressed the ID2 transcript and protein. However, although the majority of primary neuroblastomas also expressed the ID2 transcript, expression of the ID2 protein was undetectable or only barely detectable, regardless of transcript expression. In both cell lines and primary tumors, ID2 expression was independent of both CDKN2A and MYCN expression. In primary neuroblastomas, CDKN1B protein was expressed in significantly fewer advanced-stage neuroblastomas than early-stage neuroblastomas, but its expression had no relationship with CDKN2A expression or MYCN amplification. We concluded that the paradoxical expression of CDKN2A in neuroblastoma cannot be explained by inactivation of the tumor-suppressor gene CDKN1B or overexpression of the oncogene ID2. We further concluded that ID2 is not a target of MYCN regulation nor is it a prognostic factor for neuroblastoma. Finally, the loss of CDKN1B in advanced-stage neuroblastoma suggests this protein may play a role in the neuroblastoma disease process.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , DNA-Binding Proteins/biosynthesis , Neuroblastoma/metabolism , Nuclear Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/genetics , Genes, p16 , Humans , Inhibitor of Differentiation Protein 2 , N-Myc Proto-Oncogene Protein , Neuroblastoma/diagnosis , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prognosis , RNA, Messenger/biosynthesis , Repressor Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
17.
Blood ; 100(9): 3319-24, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12384433

ABSTRACT

The novel prodrug of butyric acid, pivaloyloxymethyl butyrate (AN-9), a histone deacetylase inhibitor, shows great promise as an effective and relatively nontoxic anticancer agent for solid malignancies. However, little is known about its effects on hematopoietic malignancies. In this study, we show that 21 primary samples of acute leukemia were sensitive to the antiproliferative effects of AN-9, with a 50% inhibitory concentration (IC(50)) of 45.8 +/- 4.1 microM. In colony-forming assays, primary T-cell acute lymphoblastic leukemia (T-ALL) cells were 3-fold more sensitive to AN-9 than the normal hematopoietic progenitors, erythroid burst-forming units and granulocyte/monocyte colony-forming units. AN-9 induced apoptosis in the T-ALL cell line CEM. A common problem with cancer is chemoresistance, which is often typical of relapsed cancers. Remarkably, a T-ALL sample at diagnosis and an acute myeloid leukemia sample at relapse that were resistant to doxorubicin in vitro were sensitive to AN-9, with an IC(50) of 50 microM for both samples. More strikingly, samples from 2 infants with t(4;11) ALL obtained at diagnosis and relapse each were the most sensitive to AN-9, with IC(50) values of 25 microM and 17 microM, respectively. Furthermore, a doxorubicin-resistant clone of HL60, HL60/ADR, obtained by the transfection of the MDR-1 gene, was equally sensitive to AN-9 cytotoxicity as the parental cells. AN-9 induced the expression of p21 in an infant leukemia sample with 11q23 rearrangement, but not in T- or B-precursor ALL. Collectively, our results suggest that AN-9 is a selective agent for hematopoietic malignancies that can circumvent the mechanisms of chemoresistance limiting most conventional chemotherapy.


Subject(s)
Butyrates/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Leukemia/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Acetylation/drug effects , Acute Disease , Apoptosis/drug effects , Cell Division/drug effects , Child , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Doxorubicin/pharmacology , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, MDR , HL-60 Cells/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Histones/metabolism , Humans , Infant , Inhibitory Concentration 50 , Leukemia/enzymology , Leukemia/genetics , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Leukemia-Lymphoma, Adult T-Cell/enzymology , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms/enzymology , Neoplasms/genetics , Neoplastic Stem Cells/enzymology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Processing, Post-Translational/drug effects , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL