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










Publication year range
2.
Semin Hematol ; 48 Suppl 1: S12-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21530768

ABSTRACT

Bendamustine has demonstrated substantial efficacy in the treatment of hematologic malignancies and continues to distinguish itself from other alkylating agents with regard to its activity in tumor cells. The mechanistic and clinical differences associated with bendamustine may be directly related to its unique structural features. Although the precise mechanisms of action are still poorly understood, bendamustine is associated with extensive and durable DNA damage. The increased potency of bendamustine may be due to secondary mechanisms such as inhibition of mitotic checkpoints, inefficient DNA repair, and initiation of p53-dependent DNA-damage stress response, all of which lead to mitotic catastrophe and apoptosis. It has also been hypothesized that the presence of a benzimidazole ring in addition to the nitrogen mustard group may influence the way bendamustine interacts with DNA and/or confer antimetabolite properties. Further elucidation of the mechanisms of action for bendamustine and the signaling pathways involved in the response to bendamustine-induced DNA damage is essential to maximize its therapeutic potential, identify biomarkers for response, and understand the potential for synergy with other agents involved in DNA damage and inhibition of DNA repair. This review will discuss the current understanding and hypotheses regarding bendamustine mechanisms of action and suggest future investigations that would shed light on the many unanswered questions.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Damage/drug effects , Hematologic Neoplasms/drug therapy , Nitrogen Mustard Compounds/pharmacology , Animals , Bendamustine Hydrochloride , DNA/metabolism , Humans
3.
Semin Hematol ; 48 Suppl 1: S37-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21530770

ABSTRACT

Bendamustine is a bifunctional mechlorethamine derivative that shares similarities to other alkylators; however, the presence of a benzimidazole ring may confer "nucleoside-like" properties and may allow the stabilization of the molecule leading to longer lasting DNA damage. Though bendamustine has demonstrated promising response rates in preclinical and clinical studies, particularly in follicular lymphoma, chronic lymphocytic leukemia, diffuse B-cell lymphoma, and mantle cell lymphoma, the unique and exact mechanism of action of this agent remains unclear. Several studies have been initiated to address this question, and it is hoped that emerging data will provide the basis for more effective utilization of this interesting drug. Several recent clinical trials have reported impressive results with bendamustine in lymphoid malignancies, and appropriate clinical use of this agent and the rationale behind its use are of growing importance. This review discussed emerging data and aimed to provide clinical updates and scientific rationales that are relevant to practicing clinicians who provide care to patients with lymphoid malignancies, and/or who are interested in understanding the evolving role of bendamustine in this setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma/drug therapy , Nitrogen Mustard Compounds/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Bendamustine Hydrochloride , DNA Damage/drug effects , Humans , Nitrogen Mustard Compounds/pharmacology
4.
Semin Hematol ; 48 Suppl 1: S4-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21530771

ABSTRACT

Although the alkylating agent bendamustine was developed in Germany in the mid-twentieth century, it has only recently come to the forefront in the rest of the world as an effective chemotherapeutic agent for the treatment of several hematologic malignancies. Based on the activity demonstrated in single-arm and randomized trials, this nitrogen mustard is approved by the US Food and Drug Administration (FDA) for the treatment of chronic lymphocytic leukemia and rituximab-refractory indolent non-Hodgkin lymphoma. The unique structural and mechanistic features of bendamustine differentiate it from other alkylating agents, providing increased stability and potency in DNA cross linking and subsequent cytotoxicity. Due to its unusual development, few studies have closely examined the mechanisms of action for this nitrogen mustard and many unanswered questions remain. Additionally, phase I and pharmacokinetic studies are limited, although increased understanding of the clinical pharmacology of bendamustine led to development of dosing recommendations by international experts based on the available data. The clinical activity of bendamustine as a single agent and in combination with other chemotherapeutic and immunotherapeutic drugs, coupled with its potential lack of cross-resistance with many other chemotherapy agents, make bendamustine an attractive therapy for patients with newly diagnosed and refractory hematologic malignancies. This review will discuss the development of bendamustine, its structural and pharmacologic characteristics, and current data regarding the optimal dosing of this agent in specific clinical settings.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Nitrogen Mustard Compounds/chemistry , Nitrogen Mustard Compounds/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Bendamustine Hydrochloride , Humans , Nitrogen Mustard Compounds/pharmacokinetics , Nitrogen Mustard Compounds/pharmacology
5.
BJU Int ; 108(7): 1210-4, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21314886

ABSTRACT

OBJECTIVE: • To study the pharmacokinetic and toxicity profile of intravesically administered TMX-101, with its active ingredient R-837, a synthetic Toll-like receptor (TLR)-7 agonist, in a pig model. MATERIALS AND METHODS: • TLR-7 expression was determined by immunohistochemistry in human and pig bladder tissue. • Four groups of six pigs received a 1-h intravesical instillation with R-837 of different formulations. • Pharmacokinetic analysis was performed on plasma. Toxicity evaluation included monitoring the well-being of the animals, peripheral blood cell counts, and interleukin-6 and creatinine measurements. Urine was collected for R-837 measurement and dipstick analysis. • In total, three pigs per group were sacrificed 24 h post-treatment, and the remaining animals were sacrificed after 1 week. Histopathological examination of the bladder wall was performed. RESULTS: • TLR-7 was homogeneously expressed in human and pig urothelium. • R-837 and vehicle were well tolerated without deterioration in animal well-being. • Systemic R-837 absorption was low. • Mean maximum plasma concentration of R-837 differed depending on the formulation. Post-treatment, plasma levels were negligible at 24 h. • Histopathological examination of the bladders did not show significant abnormalities, apart from the intended inflammatory reaction in the R-837 treated groups. CONCLUSION: • Intravesically administered R-837 in pigs, which showed a similar TLR-7 distribution in bladder tissue as humans, is well tolerated and causes no bladder wall toxicity, and formulations with poloxamer and hydroxypropyl-ß-cyclodextrin showed less systemic absorption.


Subject(s)
Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/toxicity , Aminoquinolines/pharmacokinetics , Aminoquinolines/toxicity , Adjuvants, Immunologic/administration & dosage , Administration, Intravesical , Aminoquinolines/administration & dosage , Animals , Imiquimod , Swine
6.
Clin Cancer Res ; 14(1): 309-17, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18172283

ABSTRACT

PURPOSE: Bendamustine has shown clinical activity in patients with disease refractory to conventional alkylator chemotherapy. The purpose of this study was to characterize the mechanisms of action of bendamustine and to compare it with structurally related compounds. EXPERIMENTAL DESIGN: Bendamustine was profiled in the National Cancer Institute in vitro antitumor screen. Microarray-based gene expression profiling, real-time PCR, immunoblot, cell cycle, and functional DNA damage repair analyses were used to characterize response to bendamustine and compare it with chlorambucil and phosphoramide mustard. RESULTS: Bendamustine displays a distinct pattern of activity unrelated to other DNA-alkylating agents. Its mechanisms of action include activation of DNA-damage stress response and apoptosis, inhibition of mitotic checkpoints, and induction of mitotic catastrophe. In addition, unlike other alkylators, bendamustine activates a base excision DNA repair pathway rather than an alkyltransferase DNA repair mechanism. CONCLUSION: These results suggest that bendamustine possesses mechanistic features that differentiate it from other alkylating agents and may contribute to its distinct clinical efficacy profile.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cell Cycle/drug effects , DNA Repair/drug effects , Gene Expression/drug effects , Nitrogen Mustard Compounds/pharmacology , Apoptosis/drug effects , Bendamustine Hydrochloride , Blotting, Western , Cell Line, Tumor , Chlorambucil/pharmacology , Cyclophosphamide/pharmacology , Drug Screening Assays, Antitumor , Flow Cytometry , Humans
7.
Mol Cancer Ther ; 4(12): 1860-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16373701

ABSTRACT

The p16(INK4A)/CDKN2A gene on chromosome 9p21 is a site of frequent allelic loss in human cancers, and in a subset of cases, homozygous deletions at this locus encompass the telomeric methylthioadenosine phosphorylase (MTAP) gene. The MTAP gene product is the principal enzyme involved in purine synthesis via the salvage pathway, such that MTAP-negative cancers are solely dependent on de novo purine synthesis mechanisms. Inhibitors of the de novo pathway can then be used to selectively blockade purine synthesis in cancer cells while causing minimal collateral damage to normal cells. In this study, we determine that 10 of 28 (35%) biliary tract cancers show complete lack of Mtap protein expression. In vitro analysis using a selective inhibitor of the de novo purine synthesis pathway, L-alanosine, shows robust growth inhibition in MTAP-negative biliary cancer cell lines CAK-1 and GBD-1 accompanied by striking depletion of intracellular ATP and failure to rescue this depletion via addition of exogenous methylthioadenosine, the principal substrate of the MTAP gene product; in contrast, no significant effects were observed in MTAP-expressing HuCCT1 and SNU308 cell lines. Colony formation studies confirmed that L-alanosine reduced both number and size of CAK-1 colonies in soft agar assays. Knockdown of Mtap protein by RNA interference in L-alanosine-resistant HuCCT1 cells conferred sensitivity to this agent, confirming that intracellular Mtap protein levels determine response to L-alanosine. Inhibitors of de novo purine synthesis can be a potential mechanism-based strategy for treatment of biliary tract cancers, one third of which show complete loss of MTAP function.


Subject(s)
Biliary Tract Neoplasms/genetics , Gene Deletion , Homozygote , Purine-Nucleoside Phosphorylase/genetics , Base Sequence , Biliary Tract Neoplasms/enzymology , Biliary Tract Neoplasms/pathology , Cell Line, Tumor , DNA Primers , Humans , Polymerase Chain Reaction
8.
Am J Surg Pathol ; 29(11): 1497-504, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16224217

ABSTRACT

The gene that encodes methylthioadenosine phosphorylase (MTAP), an enzyme involved in adenine and methionine salvage pathways, is located on chromosome 9p21 telomeric to the p16INK4A/CDKN2A tumor suppressor gene. Inactivation of the p16INK4A/CDKN2A gene occurs by three different mechanisms: hypermethylation of the gene promoter, intragenic mutation coupled with loss of the second allele, and homozygous deletion. Immunohistochemical labeling for the p16INK4A/CDKN2A gene product parallels gene status but does not elucidate the mechanism of gene inactivation. Since the MTAP gene is often co-deleted with p16INK4A/CDKN2A, concurrent immunolabeling for both proteins can identify cases with homozygous p16INK4A/CDKN2A gene deletion. MTAP loss itself has therapeutic implications since it may confer selective sensitivity to inhibitors of de novo purine biosynthesis, such as L-alanosine. Twelve tissue microarrays were constructed from 92 cases of Barrett-associated adenocarcinomas and precursor lesions and 112 cases of gastric adenocarcinoma and precursor lesions comprising 1161 individual cores. Multiple cores were arrayed from any given case, and when available, included the entire histologic spectrum of intestinal metaplasia-dysplasia-carcinoma. Tissue microarrays were labeled with monoclonal antibodies against MTAP protein (clone 6.9, Salmedix, Inc) and p16 (clone 16P07, Neomarkers). Complete loss of labeling was considered negative, while any labeling (p16: nuclear; MTAP: cytoplasmic and nuclear) was considered positive. Loss of MTAP labeling occurred exclusively in conjunction with loss of p16 labeling, confirming that the previous findings from this group that concurrent loss of MTAP and p16 labeling is a surrogate marker of 9p21 homozygous deletions. Complete loss of MTAP and p16 was seen in 4 of 25 (16%) patients with Barrett's esophagus, 4 of 18 (22%) with low-grade dysplasia, 5 of 39 (13%) with high-grade dysplasia, 17 of 78 (22%) with invasive adenocarcinoma, and 8 of 36 (22%) of metastases. There were 7 cases of esophageal adenocarcinoma with loss of both MTAP and p16 for which precursor lesions were available. In 6 on these 7 cases (85%), the precursor lesion(s) had loss of both MTAP and p16. Lack of MTAP and p16 expression was seen in 11 of 106 (10%) cases of gastric adenocarcinoma. All metaplastic (30 biopsies from 20 cases) and dysplastic (15 biopsies from 13 cases) gastric tissues had both intact MTAP and p16INK4A/CDKN2A gene products. No precursor lesions were available from the gastric cancers that had loss of both MTAP and p16. Two benign gastric hyperplastic polyps also had intact p16 and MTAP. Concurrent MTAP and p16 loss detected by immunohistochemistry can serve as a convenient surrogate for p16INK4A/CDKN2A gene homozygous deletion in archival tissues. Inactivation of p16INK4A/CDKN2A by homozygous deletion appears to be an early event in Barrett carcinogenesis, occurring in noninvasive precursor lesions, including nondysplastic Barrett mucosa, in subsets of cases. In the absence of MTAP, cells depend exclusively on the de novo synthesis pathway for production of adenosine. This loss of MTAP during 9p21 homozygous deletion might be exploited therapeutically using de novo purine synthesis antimetabolites to treat a subset of invasive gastroesophageal adenocarcinomas and esophageal precursor lesions.


Subject(s)
Adenocarcinoma/genetics , Esophageal Neoplasms/genetics , Genes, p16/physiology , Purine-Nucleoside Phosphorylase/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Esophageal Neoplasms/pathology , Gene Deletion , Humans , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Stomach Neoplasms/pathology
9.
Blood ; 106(2): 706-12, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15802527

ABSTRACT

In this study we report that R-etodolac (SDX-101), at clinically relevant concentrations, induces potent cytotoxicity in drug-sensitive multiple myeloma (MM) cell lines, as well as in dexamethasone (MM.1R)-, doxorubicin (Dox40/RPMI8226)-, and bortezomib (DHL4)-resistant cell lines. Immunoblot analysis demonstrates that R-etodolac induces apoptosis characterized by caspase-8, -9, and -3 and PARP (poly-ADP [adenosine diphosphate]-ribose polymerase) cleavage and down-regulation of cyclin D1 expression. Subcytotoxic doses of R-etodolac up-regulate myeloid cell leukemia-1 proapoptotic variant (Mcl-1S), while enhancing dexamethasone (Dex)-induced caspase activation and apoptosis. The combination of R-etodolac with Dex results in a highly synergistic cytotoxic effect. R-etodolac also induces apoptosis against primary cells isolated from patients with MM refractory to chemotherapy. Although interleukin 6 (IL-6) and insulin-like growth factor-1 (IGF-1) abrogate Dex-induced MM cell cytotoxicity, neither IL-6 nor IGF-1 protects against R-etodolac-induced cytotoxicity in MM cells. R-etodolac also inhibits viability of MM cells adherent to bone marrow stromal cells (BMSCs), thereby overcoming a mechanism of drug resistance commonly observed with other conventional chemotherapeutic agents. Our data, therefore, indicate that R-etodolac circumvents drug resistance in MM cells at clinically relevant concentrations, targets Mcl-1, and can be synergistically combined with Dex.


Subject(s)
Antineoplastic Agents/pharmacology , Dexamethasone/administration & dosage , Etodolac/pharmacology , Multiple Myeloma/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Caspases/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cyclin D1/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Etodolac/administration & dosage , Etodolac/chemistry , Humans , Insulin-Like Growth Factor I/pharmacology , Interleukin-6/pharmacology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Stereoisomerism
10.
Mod Pathol ; 18(7): 959-63, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15832197

ABSTRACT

The p16INK4A/CDKN2A (p16) gene on chromosome 9p21 is inactivated in >90% of invasive pancreatic cancers. In 40% of pancreatic cancers the p16 gene is inactivated by homozygous deletion, in 40% by an intragenic mutation coupled with loss of the second allele, and in 10-15% by hypermethylation of the p16 gene promoter. Immunohistochemical labeling for the p16 gene product parallels gene status, but does not provide information of the mechanism of p16 gene inactivation. The methylthioadenosine phosphorylase gene (MTAP) gene also resides on chromosome 9p21, approximately 100 kb telomeric to the p16 gene. The MTAP gene is frequently contained within p16 homozygous deletions, producing concordant loss of both p16 and MTAP gene expression. Concordant loss of both p16 and MTAP protein expression can therefore be used as a surrogate marker for p16 homozygous deletion. Here we immunolabeled a series of pancreatic intraepithelial neoplasia (PanIN) lesions of various histologic grades for the p16 and MTAP gene products using a high-throughput PanIN tissue microarray (TMA) format. We demonstrate concordant loss of p16 and MTAP protein expression in 6/73 (8%) PanINs, including five high-grade lesions and one low-grade lesion. Immunolabeling for both p16 and MTAP protein expression provides a tool to evaluate tissues with intact morphology for p16 gene homozygous deletions. The concordant loss of expression of both genes in PanIN lesions demonstrates that homozygous deletions of the p16 tumor suppressor gene can occur in noninvasive precursor lesions.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology , Purine-Nucleoside Phosphorylase/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Deletion , Homozygote , Humans , Immunohistochemistry , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Purine-Nucleoside Phosphorylase/genetics
11.
Proc Natl Acad Sci U S A ; 102(7): 2525-30, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699354

ABSTRACT

Prostate cancer is often slowly progressive, and it can be difficult to treat with conventional cytotoxic drugs. Nonsteroidal antiinflammatory drugs inhibit the development of prostate cancer, but the mechanism of chemoprevention is unknown. Here, we show that the R-enantiomer of the nonsteroidal antiinflammatory drug etodolac inhibited tumor development and metastasis in the transgenic mouse adenocarcinoma of the prostate (TRAMP) model, by selective induction of apoptosis in the tumor cells. This proapoptotic effect was associated with loss of the retinoid X receptor (RXRalpha) protein in the adenocarcinoma cells, but not in normal prostatic epithelium. R-etodolac specifically bound recombinant RXRalpha, inhibited RXRalpha transcriptional activity, and induced its degradation by a ubiquitin and proteasome-dependent pathway. The apoptotic effect of R-etodolac could be controlled by manipulating cellular RXRalpha levels. These results document that pharmacologic antagonism of RXRalpha transactivation is achievable and can have profound inhibitory effects in cancer development.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Etodolac/metabolism , Etodolac/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Retinoid X Receptor alpha/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Proliferation/drug effects , Etodolac/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Conformation/drug effects , RNA, Small Interfering/genetics , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/genetics , Stereoisomerism , Transcriptional Activation/drug effects , Transfection
12.
Blood ; 105(11): 4455-62, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15718423

ABSTRACT

Myeloid cell leukemia-1 (MCL-1) acts as a key survival factor for chronic lymphocytic leukemia (CLL) cells. In addition, dissipation of cellular bioenergy may impose a lethal effect on these quiescent cells. Previously, in multiple myeloma cell lines we demonstrated that halogenated adenosine (8-Cl-Ado) was phosphorylated to triphosphate (8-Cl-adenosine triphosphate [ATP]), which preferentially incorporated into mRNA and inhibited RNA synthesis by premature transcription termination. Furthermore, 8-Cl-ATP accumulation was associated with a decline in cellular bioenergy. Based on these actions, we hypothesized that 8-Cl-Ado would be ideal to target CLL lymphocytes. In the present study we demonstrate that leukemic lymphocytes incubated with 8-Cl-Ado display time- and dose-dependent increase in the accumulation of 8-Cl-ATP, with a parallel depletion of the endogenous ATP pool. Inhibition of global RNA synthesis resulted in a significant decline in the expression of transcripts with a short half-life such as MCL1. Consistent to this, protein expression of MCL-1 but not B-cell lymphoma-2 (BCL-2) was decreased. Furthermore, 8-Cl-ATP induced programmed cell death, as suggested by caspases activation, cleavage of caspase 3, and PARP (poly-adenosine diphosphate [ADP]-ribose polymerase), and increased DNA fragmentation. In conclusion, 8-Cl-Ado induces apoptosis in CLL lymphocytes by targeting cellular bioenergy as well as RNA transcription and translation of key survival genes such as MCL1.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Apoptosis/drug effects , Energy Metabolism/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Transcription, Genetic/drug effects , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacokinetics , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis
14.
Cancer Biol Ther ; 4(1): 83-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15662124

ABSTRACT

Methylthioadenosine phosphorylase (MTAP) plays an important role in the salvage pathway for the synthesis of adenosine. Novel chemotherapeutic strategies exploiting the selective loss of MTAP function in cancers have been proposed. The MTAP gene, on chromosome 9p21, is frequently included within homozygous deletions of the p16INK4A/ CDKN2A gene. Biallelic deletions of the p16INK4A/CDKN2A gene are found in 40% of pancreatic cancers, suggesting that the MTAP gene may be frequently inactivated in pancreatic cancer and that selected patients with pancreatic cancer may benefit from therapies targeting this loss. We immunolabeled six xenografted pancreatic cancers with known MTAP and p16INK4A/CDKN2A gene status and found that immunolabeling mirrored gene status. Loss of expression of both MTAP and p16 was observed only in those pancreatic cancers with homozygous deletions that encompassed both the MTAP and p16INK4A/CDKN2A genes. We then immunolabeled a series of 320 microarrayed infiltrating pancreatic adenocarcinomas, 35 biliary adenocarcinomas, 54 ampullary cancers, and 35 noninvasive intraductal papillary mucinous neoplasms. Immunolabeling for MTAP was lost in 91 of the 300 (30%) evaluable pancreatic cancers, 9 of 54 (17%) ampullary cancers, 4 of 33 (12%) biliary cancers, and in 1 of 35 (3%) IPMNs. All neoplasms with loss of MTAP labeling also demonstrated loss of p16 labeling. These results suggest that MTAP expression is lost in approximately 30% of infiltrating pancreatic cancers and in a lower percentage of other periampullary neoplasms, that this loss is the result of homozygous deletions encompassing both the MTAP and p16INK4A/CDKN2A genes. Thus, pancreatic cancer is a promising cancer type in which to explore novel chemotherapeutic strategies to exploit the selective loss of MTAP function.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Common Bile Duct Neoplasms/genetics , Common Bile Duct Neoplasms/pathology , Gene Deletion , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Purine-Nucleoside Phosphorylase/genetics , Adenocarcinoma/therapy , Adult , Aged , Aged, 80 and over , Ampulla of Vater , Animals , Common Bile Duct Neoplasms/therapy , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/therapy , Transplantation, Heterologous
15.
Cancer Cell ; 5(6): 565-74, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15193259

ABSTRACT

R-etodolac, a nonsteroidal anti-inflammatory drug, inhibits the progression of CWRSA6 androgen-independent and LuCaP-35 androgen-dependent prostate cancer xenograft growth through downregulation of cyclin D1 expression via the PPARgamma pathway. PPARgamma protein degradation, observed post-R-etodolac treatment, resulted from phospho-MAP kinase (p44/42) induction by R-etodolac negatively regulating PPARgamma function. Negative regulation of PPARgamma was overcome by a combination regimen of R-etodolac with the HER-kinase axis inhibitor, rhuMab 2C4, which demonstrated an additive antitumor effect. We further show that the inhibition of HER-kinase activity by rhuMab 2C4 is sufficient to inhibit PPARgamma protein degradation. This study introduces a novel concept of an in vivo crosstalk between the HER-kinase axis and PPARgamma pathways, ultimately leading to negative regulation of PPARgamma activity and tumor growth inhibition.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Etodolac/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Azo Compounds/pharmacology , Blotting, Western , CD36 Antigens/biosynthesis , Cell Line , Cell Line, Tumor , Coloring Agents/pharmacology , Cyclin D1/biosynthesis , Cyclin D1/metabolism , Down-Regulation , Enzyme Activation , Flow Cytometry , Humans , Lipid Metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Nude , Models, Biological , Models, Chemical , Monocytes/metabolism , NIH 3T3 Cells , Neoplasm Transplantation , Prostatic Neoplasms/pathology , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcriptional Activation , Transfection
16.
Proc Natl Acad Sci U S A ; 101(9): 3118-23, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14973184

ABSTRACT

B cell chronic lymphocytic leukemia (CLL) is characterized by an accumulation of mature, functionally incompetent B cells. Wnts are a large family of secreted glycoproteins involved in cell proliferation, differentiation, and oncogenesis. The classical Wnt signaling cascade inhibits the activity of the enzyme glycogen synthase kinase-3beta, augmenting beta-catenin translocation to the nucleus, and the transcription of target genes. Little is known about the potential roles of Wnt signaling in CLL. In this study, we quantified the gene expression profiles of the Wnt family, and their cognate frizzled (Fzd) receptors in primary CLL cells, and determined the role of Wnt signaling in promoting CLL cell survival. Wnt3, Wnt5b, Wnt6, Wnt10a, Wnt14, and Wnt16, as well as the Wnt receptor Fzd3, were highly expressed in CLL, compared with normal B cells. Three lines of evidence suggested that the Wnt signaling pathway was active in CLL. First, the Wnt/beta-catenin-regulated transcription factor lymphoid-enhancing factor-1, and its downstream target cyclin D1, were overexpressed in CLL. Second, a pharmacological inhibitor of glycogen synthase kinase-3 beta, SB-216763, activated beta-catenin-mediated transcription, and enhanced the survival of CLL lymphocytes. Third, Wnt/beta-catenin signaling was diminished by an analog of a nonsteroidal antiinflammatory drug (R-etodolac), at concentrations that increased apoptosis of CLL cells. Taken together, these results indicate that Wnt signaling genes are overexpressed and are active in CLL. Uncontrolled Wnt signaling may contribute to the defect in apoptosis that characterizes this malignancy.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/blood , Proto-Oncogene Proteins/genetics , Zebrafish Proteins , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Cell Survival/drug effects , Cells, Cultured , Cyclin D1/genetics , Cytoskeletal Proteins/physiology , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Indoles/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/physiology , Maleimides/pharmacology , Multigene Family , Protein-Tyrosine Kinases/genetics , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Trans-Activators/physiology , Transcription Factors/genetics , Wnt Proteins , beta Catenin
17.
Acta Haematol ; 111(1-2): 107-23, 2004.
Article in English | MEDLINE | ID: mdl-14646349

ABSTRACT

Molecular targeting therapies for hematological malignant diseases such as monoclonal antibodies and small molecules have been reviewed. Imatinib mesylate (STI571) targets the tyrosine kinase activity of the BCR-ABL fusion protein in CML, and was superior to IFN-alpha plus low-dose cytarabine in newly diagnosed chronic-phase CML in a phase III randomized study. Imatinib induced apoptosis in BCR-ABL-positive cells in vitro, and activates several signaling pathways such as PI3K/Akt, STAT5 and Ras/MAPK. Combination therapies with imatinib and new strategies for downregulation of intracellular BCR-ABL protein levels have also been investigated from the phenomenon of resistance to imatinib. Anti-CD20 (rituximab) became the first monoclonal antibody approved for the treatment of a relapsed/refractory follicular/low-grade NHL and promising results were obtained from a phase III randomized study. Although antibody-dependent cell-mediated cytotoxicity and complement-mediated cytotoxicity are likely to be the major effectors of B-cell depletion in vivo, direct cytotoxicity by CD20 monoclonal antibody on B-cell lines in vitro has been reported. Anti-CD33 (Mylotarg) and FLT3 inhibitors for AML have also been used in clinical trials and signaling pathways induced by these agents are under intensive investigation. Arsenic trioxide, like all-TRANS-retinoic acid (ATRA), downregulates promyelocytic leukemia protein/retinoic acid receptor-alpha (PML/RARalpha) fusion protein and induced apoptosis in APL cells, and promising results were obtained from ATRA-resistant APL patients. Finally we show our promising in vitro and in vivo data of R-etodolac (a non-steroidal anti-inflammatory drug lacking cyclooxygenase inhibitor activity) against chronic lymphocytic leukemia (CLL) cells.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Hematologic Neoplasms/drug therapy , Humans
18.
Best Pract Res Clin Haematol ; 16(1): 83-9, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12670467

ABSTRACT

Hairy-cell leukaemia cells have a low rate of growth but an even lower rate of apoptosis. Accordingly, this malignancy is an excellent model for studying the effects of drugs on the pathways of apoptosis independently of cell proliferation. The remarkable effectiveness of 2-chlorodeoxyadenosine in hairy-cell leukaemia affirms the feasibility of developing drugs that can destroy even non-proliferating malignant cells. The major nucleotide metabolite of 2-chlorodeoxyadenosine accumulates selectively in lymphocytes and co-activates two key apoptosis-regulating enzymes: poly(ADP-ribose) polymerase and Apaf-1/caspase-9. The ability of 2-chlorodeoxyadenosine to induce durable remissions in hairy-cell leukaemia may also be attributable to its effects on lymphocytes and monocytes in the microenvironment, although this latter effect remains to be proven experimentally.


Subject(s)
Antineoplastic Agents/therapeutic use , Cladribine/therapeutic use , Drug Evaluation/methods , Leukemia, Hairy Cell/drug therapy , Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1 , Caspase 9 , Caspases/drug effects , Caspases/metabolism , Humans , Leukemia, Hairy Cell/metabolism , Leukemia, Hairy Cell/pathology , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proteins/drug effects , Proteins/metabolism
19.
Blood ; 100(5): 1795-801, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12176902

ABSTRACT

Chronic lymphocytic leukemia (CLL) B cells have defects in apoptosis pathways and therefore accumulate in vivo. However, when removed from the patient and cultured in vitro, these malignant cells rapidly undergo apoptosis. Recent studies suggest that leukemia cell survival is influenced by interactions with nonleukemia cells in the microenvironment of lymph nodes, marrow, and other tissues. To model such cell-cell interactions in vitro, we cultured freshly isolated CLL B cells with a follicular dendritic cell line, HK. CLL B cells cocultured with HK cells were protected from apoptosis, either spontaneous or induced by treatment with anticancer drugs. Protection against spontaneous apoptosis could also be induced by coculturing the CLL B cells with normal dendritic cells (DCs) or with a CD40-ligand (CD154)-expressing fibroblast cell line. Examination of the expression of several apoptosis-regulatory proteins revealed that coculture with HK cells or DCs induced up-regulation of the antiapoptotic Bcl-2 family protein Mcl-1 in CLL B cells, whereas CD40 ligation increased expression of Bcl-X(L). Cell-cell contact was required for HK-induced protection, and introducing neutralizing antibodies against various adhesion molecules showed that CD44 was involved in HK-mediated survival, whereas CD40, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) were not. Anti-CD44 antibodies also blocked Mcl-1 induction by HK cells. Mcl-1 antisense oligonucleotides reduced leukemia cell expression of Mcl-1, and significantly suppressed HK-induced protection against apoptosis, whereas control oligonucleotides had no effect. Thus, HK cells protect CLL B cells against apoptosis, at least in part through a CD44-dependent mechanism involving up-regulation of Mcl-1, and this mechanism is distinct from that achieved by CD40 ligation. Consequently, the particular antiapoptotic proteins important for CLL survival may vary depending on the microenvironment.


Subject(s)
Cell Communication , Dendritic Cells/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2 , Apoptosis/immunology , CD40 Ligand/immunology , CD40 Ligand/metabolism , Cell Survival , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/immunology , Tumor Cells, Cultured
20.
Gastroenterology ; 122(5): 1428-41, 2002 May.
Article in English | MEDLINE | ID: mdl-11984528

ABSTRACT

BACKGROUND & AIMS: Impaired mucosal barrier, cytokine imbalance, and dysregulated CD4(+) T cells play important roles in the pathogenesis of experimental colitis and human inflammatory bowel disease. Immunostimulatory DNA sequences (ISS-DNA) and their synthetic oligonucleotide analogs (ISS-ODNs) are derived from bacterial DNA, are potent activators of innate immunity at systemic and mucosal sites, and can rescue cells from death inflicted by different agents. We hypothesized that these combined effects of ISS-DNA could inhibit the damage to the colonic mucosa in chemically induced colitis and thereby limit subsequent intestinal inflammation. METHODS: The protective and the anti-inflammatory effect of ISS-ODN administration were assessed in dextran sodium sulfate-induced colitis and in 2 models of hapten-induced colitis in Balb/c mice. Similarly, these effects of ISS-ODN were assessed in spontaneous colitis occurring in IL-10 knockout mice. RESULTS: In all models of experimental and spontaneous colitis examined, ISS-ODN administration ameliorated clinical, biochemical, and histologic scores of colonic inflammation. ISS-ODN administration inhibited the induction of colonic proinflammatory cytokines and chemokines and suppressed the induction of colonic matrix metalloproteinases in both dextran sodium sulfate- and hapten-induced colitis. CONCLUSIONS: As the colon is continuously exposed to bacterial DNA, these findings suggest a physiologic, anti-inflammatory role for immunostimulatory DNA in the GI tract. Immunostimulatory DNA deserves further evaluation for the treatment of human inflammatory bowel disease.


Subject(s)
Adjuvants, Immunologic/pharmacology , Colitis/prevention & control , DNA/pharmacology , Oligodeoxyribonucleotides/pharmacology , Animals , Cyclooxygenase 2 , Female , Interleukin-10/physiology , Isoenzymes/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandins/biosynthesis , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...