Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Ann Oncol ; 28(5): 1145-1151, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28453708

ABSTRACT

BACKGROUND: While next generation sequencing has enhanced our understanding of the biological basis of malignancy, current knowledge on global practices for sequencing cancer samples is limited. To address this deficiency, we developed a survey to provide a snapshot of current sequencing activities globally, identify barriers to data sharing and use this information to develop sustainable solutions for the cancer research community. METHODS: A multi-item survey was conducted assessing demographics, clinical data collection, genomic platforms, privacy/ethics concerns, funding sources and data sharing barriers for sequencing initiatives globally. Additionally, respondents were asked as to provide the primary intent of their initiative (clinical diagnostic, research or combination). RESULTS: Of 107 initiatives invited to participate, 59 responded (response rate = 55%). Whole exome sequencing (P = 0.03) and whole genome sequencing (P = 0.01) were utilized less frequently in clinical diagnostic than in research initiatives. Procedures to identify cancer-specific variants were heterogeneous, with bioinformatics pipelines employing different mutation calling/variant annotation algorithms. Measurement of treatment efficacy varied amongst initiatives, with time on treatment (57%) and RECIST (53%) being the most common; however, other parameters were also employed. Whilst 72% of initiatives indicated data sharing, its scope varied, with a number of restrictions in place (e.g. transfer of raw data). The largest perceived barriers to data harmonization were the lack of financial support (P < 0.01) and bioinformatics concerns (e.g. lack of interoperability) (P = 0.02). Capturing clinical data was more likely to be perceived as a barrier to data sharing by larger initiatives than by smaller initiatives (P = 0.01). CONCLUSIONS: These results identify the main barriers, as perceived by the cancer sequencing community, to effective sharing of cancer genomic and clinical data. They highlight the need for greater harmonization of technical, ethical and data capture processes in cancer sample sequencing worldwide, in order to support effective and responsible data sharing for the benefit of patients.


Subject(s)
Genetic Association Studies , Neoplasms/genetics , DNA Mutational Analysis , Databases, Genetic , Genetic Predisposition to Disease , Genome, Human , Humans , Molecular Sequence Annotation , Surveys and Questionnaires , Exome Sequencing
2.
Leukemia ; 21(7): 1481-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17476280

ABSTRACT

Monitoring of BCR-ABL transcripts has become established practice in the management of chronic myeloid leukemia. However, nucleic acid amplification techniques are prone to variations which limit the reliability of real-time quantitative PCR (RQ-PCR) for clinical decision making, highlighting the need for standardization of assays and reporting of minimal residual disease (MRD) data. We evaluated a lyophilized preparation of a leukemic cell line (K562) as a potential quality control reagent. This was found to be relatively stable, yielding comparable respective levels of ABL, GUS and BCR-ABL transcripts as determined by RQ-PCR before and after accelerated degradation experiments as well as following 5 years storage at -20 degrees C. Vials of freeze-dried cells were sent at ambient temperature to 22 laboratories on four continents, with RQ-PCR analyses detecting BCR-ABL transcripts at levels comparable to those observed in primary patient samples. Our results suggest that freeze-dried cells can be used as quality control reagents with a range of analytical instrumentations and could enable the development of urgently needed international standards simulating clinically relevant levels of MRD.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Profiling/standards , Polymerase Chain Reaction/methods , Protein-Tyrosine Kinases/genetics , RNA, Messenger/analysis , Freeze Drying , Fusion Proteins, bcr-abl , Humans , Indicators and Reagents , K562 Cells , Polymerase Chain Reaction/standards , Protein-Tyrosine Kinases/analysis , Quality Control , Reference Standards
3.
Proc Natl Acad Sci U S A ; 104(13): 5563-8, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17372210

ABSTRACT

PTEN is an important tumor-suppressor gene associated with many cancers. Through expression profiling of glioblastoma tissue samples and prostate cancer xenografts, we identified a molecular signature for loss of the PTEN tumor suppressor in glioblastoma and prostate tumors. The PTEN signature consists of a minimum of nine genes, several of which are involved in various pathways already implicated in tumor formation. Among these signature genes, the most significant was an increase in insulin growth factor-binding protein 2 (IGFBP-2) mRNA. Up-regulation of IGFBP-2 was confirmed at the protein level by Western blot analysis and validated in samples not included in the microarray analysis. The link between IGFBP-2 and PTEN was of particular interest because elevated serum IGFBP-2 levels have been reported in patients with prostate and brain tumors. To further investigate this link, we determined that IGFBP-2 expression is negatively regulated by PTEN and positively regulated by phosphatidylinositol 3-kinase (PI3K) and Akt activation. In addition, Akt-driven transformation is impaired in IGFBP2(-/-) mouse embryo fibroblasts, implicating a functional role for IGFBP-2 in PTEN signaling. Collectively, these studies establish that PTEN and IGFBP-2 expression are inversely correlated in human brain and prostate cancers and implicate serum IGFBP-2 levels as a potential serum biomarker of PTEN status and PI3K Akt pathway activation in cancer patients.


Subject(s)
Biomarkers/chemistry , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Insulin-Like Growth Factor Binding Protein 2/physiology , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Humans , Insulin-Like Growth Factor Binding Protein 2/metabolism , Male , Mice , Neoplasm Transplantation
4.
Article in English | MEDLINE | ID: mdl-16869786

ABSTRACT

The success of kinase inhibitor therapy in chronic myeloid leukemia (CML) has validated the long-held thesis in the cancer research community that a precise molecular understanding of cancer can directly affect cancer therapy. Now that several years have passed since the approval of imatinib/Gleevec for CML treatment, we have a greater appreciation for the challenges involved in effectively deploying these agents in the clinic. In this paper, I review recent events in the treatment of CML and highlight early applications of kinase inhibitor therapy to other diseases such as glioblastoma. I conclude with a vision that it may be possible, through analysis of tumor proteins secreted into serum, to track distinct molecular features of various cancers in order to select appropriate molecularly targeted therapy and measure treatment response. This new science of cancer biomarkers could radically transform the conduct of clinical trials and speed the evaluation of new molecularly targeted agents.


Subject(s)
Neoplasms/drug therapy , Biomarkers , Drug Resistance, Neoplasm , Genes, Tumor Suppressor , Humans , Mutation , Neoplasms/enzymology , Neoplasms/genetics , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/genetics
5.
Leukemia ; 16(8): 1484-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145689

ABSTRACT

The ability of interferon-alpha (IFN-alpha) to induce dendritic cell (DC) differentiation in chronic myeloid leukemia (CML) was evaluated. Peripheral blood mononuclear cells from CML patients cultured with IFN-alpha and granulocyte-macrophage colony-stimulating factor (GM-CSF) developed a dendritic morphology. Fluorescence in situ hybridization demonstrated that the DCs harbored the bcr/abl translocation. The DCs prepared with IFN-alpha/GM-CSF expressed significantly higher levels of class I and II HLA than those grown in interleukin-4 (IL-4) and GM-CSF. The DCs prepared from newly diagnosed CML patients using IFN-alpha/GM-CSF expressed immunoregulatory proteins at levels comparable to normal DCs. In contrast, DCs cultured from CML patients who did not achieve a cytogenetic response to IFN-alpha expressed significantly lower levels of class I HLA, CD40, CD54, CD80 and CD86 than normal DCs. The expression of CD86 by CML DCs was enhanced when they were cultured with IFN-alpha/IL-4/GM-CSF, or when IFN-alpha/GM-CSF-treated cells were induced to mature by CD40 ligand. The DCs from IFN-alpha failures were less stimulatory than normal DCs in the allogeneic mixed leukocyte reaction. CML patients who had a cytogenetic response to IFN-alpha initially had low numbers of bone marrow DCs that increased significantly with treatment, while nonresponders had more prevalent DCs at baseline that showed no consistent change with treatment. Therefore, IFN-alpha can induce DC differentiation from CML progenitor cells both in vitro and in vivo. The therapeutic activity of IFN-alpha in CML may be due to its ability to stimulate the generation of DCs that can present CML-specific antigens. Resistance to IFN-alpha may result when DC differentiation becomes impaired.


Subject(s)
Dendritic Cells/drug effects , Interferon-alpha/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/drug effects , Antigen Presentation , Antigens, CD/analysis , Antigens, CD/biosynthesis , Antigens, CD/genetics , Biomarkers, Tumor/genetics , Blood Cells/pathology , Bone Marrow Cells/pathology , CD40 Ligand/pharmacology , Cell Differentiation/drug effects , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA Antigens/analysis , HLA Antigens/biosynthesis , HLA Antigens/genetics , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Interferon alpha-2 , Lymphocyte Culture Test, Mixed , Neoplastic Stem Cells/pathology , Recombinant Proteins , Tumor Cells, Cultured/drug effects
7.
Article in English | MEDLINE | ID: mdl-11722980

ABSTRACT

The treatment recommendations for chronic myelogenous leukemia (CML) are evolving rapidly. In the past year, pegylated interferon and STI571 (Gleevec, imatinib mesylate), a Bcr-Abl tyrosine kinase inhibitor, have become commercially available and non-myeloablative stem cell transplants continue to be refined. Clinicians and patients face a bewildering array of treatment options for CML. In this article Dr. Sawyer reviews the clinical results with STI571 and ongoing investigations into mechanisms of resistance to STI571. Given the newness of STI571, a practical overview on the administration of STI571 is presented by Drs. Druker and Ford, focusing on aspects such as optimal dose, management of common side effects, and potential drug interactions. The most recent data on interferon-based regimens are reviewed by Dr. Baccarani in the third section. In the last section Dr. Goldman presents recent results of allogeneic stem cell transplants, including the reduced intensity conditioning regimens. Lastly, the proposed place of each of these treatments in the management of CML patients is addressed to assist in deciding amongst treatment options for CML patients.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Benzamides , Clinical Trials as Topic , Hematopoietic Stem Cell Transplantation , Humans , Imatinib Mesylate , Interferon-alpha/therapeutic use , Piperazines/administration & dosage , Piperazines/therapeutic use , Piperazines/toxicity , Practice Guidelines as Topic , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Pyrimidines/toxicity , Treatment Outcome
10.
Cancer Res ; 61(16): 6276-80, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507082

ABSTRACT

Androgen deprivation therapies for metastatic prostate cancer are useful initially, but progression to androgen independence usually results in relapse within 2 years. The molecular mechanisms underlying the clinically important transition from androgen dependence to androgen independence are poorly described. Several lines of investigation have suggested that insulin-like growth factors (IGFs) are involved in the biology of prostate cancer, but little is known about their relevance to progression to androgen independence. We used three in vivo models of androgen-dependent (AD) human prostate cancer to study this issue. Progression to androgen-independent (AI) growth was associated with a 60-fold increase in expression of IGF-I mRNA in LAPC-9 xenografts and a 28-fold increase in IGF-I expression in LNCAP xenografts, relative to the initial AD neoplasms. IGF type I receptor (IGF-IR) mRNA levels were approximately 2.5-fold and approximately 5-fold higher, respectively, in AI LAPC-9 and LNCaP tumors compared with the original AD neoplasms. AI growth of these xenografts was also associated with significant reductions in IGF binding protein-3 expression. LAPC-4 xenografts, which previously have been shown to exhibit molecular pathology related to HER-2/neu expression with progression to AI, showed relatively minor changes in expression of the genes investigated, but we nevertheless found evidence of increased IGF-IR phosphorylation with progression to androgen independence in this model. Taken together with prior observations, our results suggest that deregulation of expression of genes related to any one of several critical receptor tyrosine kinase regulatory systems, including IGF signaling, may confer androgen independence.


Subject(s)
Insulin-Like Growth Factor I/genetics , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor, IGF Type 1/genetics , Androgens/physiology , Animals , Disease Progression , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 5/biosynthesis , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor I/biosynthesis , Male , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, IGF Type 1/biosynthesis , Transplantation, Heterologous
12.
Semin Hematol ; 38(3 Suppl 8): 15-21, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11526597

ABSTRACT

The tyrosine kinase inhibitor imatinib mesylate (Gleevec, Novartis Pharmaceuticals Corp, East Hanover, NJ) (formerly STI571) blocks the constitutively activated Bcr-Abl tyrosine kinase that is characteristic of chronic myeloid leukemia (CML). Molecular analysis for the presence of residual Bcr-Abl-positive cells in patients with a cytogenetic response following treatment with imatinib mesylate reveals that some patients have undetectable disease using quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) assays capable of detecting 1 in 10(5) Philadelphia chromosome-positive (Ph(+)) cells. To examine whether the leukemia is still Bcr-Abl-dependent in patients who have responded to imatinib mesylate but have relapsed, a quantitative assay that directly measures enzymatic activity of Bcr-Abl toward one of its major signaling substrates has been developed. This assay allows monitoring both of the imatinib mesylate sensitivity of patient cells in vitro, and of the endogenous inhibition of Bcr-Abl kinase activity during imatinib mesylate treatment and relapse. Studies show that imatinib mesylate resistance is associated with restored activation of the Bcr-Abl signal transduction pathway in the majority of cases, indicating that Bcr-Abl remains a valid target for therapeutic intervention. Understanding resistance mechanisms of Ph(+) leukemia to imatinib mesylate will allow design of therapies to overcome such barriers to efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Benzamides , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/therapeutic use , Humans , Imatinib Mesylate , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Recurrence
13.
Proc Natl Acad Sci U S A ; 98(18): 10320-5, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504907

ABSTRACT

PTEN phosphatase acts as a tumor suppressor by negatively regulating the phosphoinositide 3-kinase (PI3K) signaling pathway. It is unclear which downstream components of this pathway are necessary for oncogenic transformation. In this report we show that transformed cells of PTEN(+/-) mice have elevated levels of phosphorylated Akt and activated p70/S6 kinase associated with an increase in proliferation. Pharmacological inactivation of mTOR/RAFT/FRAP reduced neoplastic proliferation, tumor size, and p70/S6 kinase activity, but did not affect the status of Akt. These data suggest that p70/S6K and possibly other targets of mTOR contribute significantly to tumor development and that inhibition of these proteins may be therapeutic for cancer patients with deranged PI3K signaling.


Subject(s)
Phosphoric Monoester Hydrolases/deficiency , Protein Kinase Inhibitors , Protein Kinases , Ribosomal Protein S6 Kinases/metabolism , Tumor Suppressor Proteins , Alleles , Animals , Base Sequence , Cell Transformation, Neoplastic/drug effects , DNA Primers/genetics , Female , Humans , Mice , Mice, Knockout , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
14.
Proc Natl Acad Sci U S A ; 98(18): 10314-9, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504908

ABSTRACT

Recent evidence places the FRAP/mTOR kinase downstream of the phosphatidyl inositol 3-kinase/Akt-signaling pathway, which is up-regulated in multiple cancers because of loss of the PTEN tumor suppressor gene. We performed biological and biochemical studies to determine whether PTEN-deficient cancer cells are sensitive to pharmacologic inhibition of FRAP/mTOR by using the rapamycin derivative CCI-779. In vitro and in vivo studies of isogenic PTEN(+/+) and PTEN(-/-) mouse cells as well as human cancer cells with defined PTEN status showed that the growth of PTEN null cells was blocked preferentially by pharmacologic FRAP/mTOR inhibition. Enhanced tumor growth caused by constitutive activation of Akt in PTEN(+/+) cells also was reversed by CCI-779 treatment, indicating that FRAP/mTOR functions downstream of Akt in tumorigenesis. Loss of PTEN correlated with increased S6 kinase activity and phosphorylation of ribosomal S6 protein, providing evidence for activation of the FRAP/mTOR pathway in these cells. Differential sensitivity to CCI-779 was not explained by differences in biochemical blockade of the FRAP/mTOR pathway, because S6 phosphorylation was inhibited in sensitive and resistant cell lines. These results provide rationale for testing FRAP/mTOR inhibitors in PTEN null human cancers.


Subject(s)
Immunophilins/antagonists & inhibitors , Neoplasms, Experimental/enzymology , Phosphoric Monoester Hydrolases/deficiency , Phosphotransferases (Alcohol Group Acceptor) , Protein Kinase Inhibitors , Protein Kinases , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cells, Cultured , Eukaryotic Initiation Factors , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , PTEN Phosphohydrolase , Phosphoproteins/metabolism , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Ribosomal Protein S6 Kinases/metabolism , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tumor Cells, Cultured
15.
Genes Chromosomes Cancer ; 31(4): 333-44, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11433524

ABSTRACT

Previous molecular genetic analyses identified a region of deletion at 13q21 in a variety of human cancers, suggesting the existence of a tumor suppressor gene(s) at this locus. In our earlier study on prostate cancer, the region of deletion was confined to a 3.1 cM interval between D13S152 and D13S162. At present, however, no known gene located in this interval has been firmly implicated in cancer, and the region remains too large for gene identification. To fine-map the area of interest, we established a contig of bacterial artificial chromosome (BAC) clones, narrowed the region of deletion by loss of heterozygosity (LOH) and homozygosity-mapping-of-deletion (HOMOD) analyses in different types of cancers, and tested a candidate gene from the region for mutation and alteration of expression in prostate cancers. The contig consisted of 75 overlapping BAC clones. In addition to the generation of 47 new sequence-tagged-site (STS) markers from the ends of BAC inserts, 76 known STS and expressed sequence tag markers were mapped to the contig (25 kb per marker on average). The minimal region of deletion was further defined to be about 700 kb between markers D13S791 and D13S166 by LOH analysis of 42 cases of prostate cancer, and by HOMOD analysis of eight prostate cancer cell lines/xenografts and 49 cell lines from cancers of the breast, ovary, endometrium, and cervix, using 18 microsatellite markers encompassing the deletion region. A gene that is homologous to the WT1 tumor suppressor gene, AP-2rep (KLF12), was mapped in this region and was analyzed for its expression and genetic mutation. In addition to low levels of expression in both normal and neoplastic cells of the prostate, this gene did not have any mutations in a group of aggressive prostate cancers and cell lines/xenografts, as assessed by the methods of polymerase chain reaction-single strand conformational polymorphism analysis and direct sequencing. These studies suggest that a 700 kb interval at 13q21 harbors a tumor suppressor gene(s) that seems to be involved in multiple types of cancer, and that the AP-2rep gene is unlikely to be an important tumor suppressor gene in prostate cancer. The BAC contig and high-resolution physical map of the defined region of deletion should facilitate the cloning of a tumor suppressor gene(s) at 13q21.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 13 , Neoplasms/genetics , Physical Chromosome Mapping , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Yeast , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor/genetics , Homozygote , Humans , Kruppel-Like Transcription Factors , Loss of Heterozygosity/genetics , Molecular Sequence Data , Repressor Proteins/genetics , Sequence Tagged Sites , Transcription Factors/genetics , Tumor Cells, Cultured
16.
Science ; 293(5531): 876-80, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11423618

ABSTRACT

Clinical studies with the Abl tyrosine kinase inhibitor STI-571 in chronic myeloid leukemia demonstrate that many patients with advanced stage disease respond initially but then relapse. Through biochemical and molecular analysis of clinical material, we find that drug resistance is associated with the reactivation of BCR-ABL signal transduction in all cases examined. In six of nine patients, resistance was associated with a single amino acid substitution in a threonine residue of the Abl kinase domain known to form a critical hydrogen bond with the drug. This substitution of threonine with isoleucine was sufficient to confer STI-571 resistance in a reconstitution experiment. In three patients, resistance was associated with progressive BCR-ABL gene amplification. These studies provide evidence that genetically complex cancers retain dependence on an initial oncogenic event and suggest a strategy for identifying inhibitors of STI-571 resistance.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Genes, abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Base Sequence , Benzamides , Blast Crisis/genetics , Cell Line , Drug Resistance, Neoplasm/genetics , Gene Amplification , Humans , Hydrogen Bonding , Imatinib Mesylate , Molecular Sequence Data , Philadelphia Chromosome , Phosphorylation , Piperazines/metabolism , Piperazines/therapeutic use , Point Mutation , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-crk , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Recurrence , Signal Transduction
17.
Oncogene ; 20(25): 3301-5, 2001 May 31.
Article in English | MEDLINE | ID: mdl-11423979

ABSTRACT

Aneuploidy is a characteristic of the majority of human cancers, and recent studies suggest that defects of mitotic checkpoints play a role in carcinogenesis. MAD1L1 is a checkpoint gene, and its dysfunction is associated with chromosomal instability. Rare mutations of this gene have been reported in colon and lung cancers. We examined a total of 44 cell lines (hematopoietic, prostate, osteosarcoma, breast, glioblastoma and lung) and 133 fresh cancer cells (hematopoietic, prostate, breast and glioblastoma) for alterations of MAD1L1 by RT-PCR-SSCP and nucleotide sequencing. Eight mutations consisting of missense, nonsense and frameshift mutations were found, together with a number of nucleotide polymorphisms. All the alterations in cell lines were heterozygous. Frequency of mutations was relatively high in prostate cancer (2/7 cell lines and 2/33 tumor specimens). We placed a mutant truncated MAD1L1, found in a lymphoma sample, into HOS, Ht161 and SJSA cell lines and found that it was less inhibitory than wild type MAD1L1 at decreasing cell proliferation. Co-expression experiments showed that the mutant form had a dominant-negative effect. Furthermore, this mutant impaired the mitotic checkpoint as shown by decreased mitotic indices in HOS cells expressing mutant MAD1L1 after culture with the microtubule-disrupting agent, nocodazole. Our results suggest a pathogenic role of MAD1L1 mutations in various types of human cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mitosis/genetics , Mutation , Neoplasm Proteins/genetics , Breast Neoplasms/genetics , Codon, Nonsense , Female , Frameshift Mutation , Hematologic Neoplasms/genetics , Humans , Lung Neoplasms/genetics , Male , Mutation, Missense , Osteosarcoma/genetics , Prostatic Neoplasms/genetics
18.
Anticancer Res ; 21(2A): 971-8, 2001.
Article in English | MEDLINE | ID: mdl-11396190

ABSTRACT

BACKGROUND: Prostate cancer metastases to bone are associated with significant morbidity and mortality. Presently, there is little known about the biological interaction between prostate cancer cells and bone. Development of an animal model using adult human bone will enhance our ability to study the biology of prostate cancer metastasis to bone. METHODS: Bone was harvested from patients undergoing total joint arthroplasty and implanted in the hindlimbs of pre-treated SCID mice. Two months after bone implantation 4 x 104 prostate cancer cells (PC-3 or LAPC-4) were injected near the bone implantation site. The animals were sacrificed approximately 8 to 12 weeks after the injections of the cells. Complete histological analysis including immunostaining was performed. RESULTS: Both the PC-3 and LAPC-4 prostate cancer cells homed to the human bone implant, specifically the reconstituted bone marrow cavity. Analysis of the bone-tumor interaction after injection of PC-3 cells revealed strong labeling for PTHrP, TNF alpha and IL-6, consistent with osteoclast recruitment and osteoclast activity. These cells also were positively stained for CK18. After cellular injection of LAPC-4 cells, there was strong labeling for TNF alpha, IL-6, and IL-1 (osteoclast recruitment and osteolytic activity). PTHrP staining was also noted. The bone cells were strongly stained for osteocalcin, and the tumor cells for PSA. CONCLUSIONS: These data suggest that the tumor cells may induce an osteolytic response to enhance their ability to metastasize to bone. This animal model allows us to study the biologic interaction between prostate cancer cells and human bone and may enhance our understanding of the events associated with prostate cancer metastasis to bone.


Subject(s)
Adenocarcinoma/pathology , Bone Neoplasms/secondary , Disease Models, Animal , Prostatic Neoplasms/pathology , Adenocarcinoma/metabolism , Animals , Bone Development , Bone Neoplasms/metabolism , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms/metabolism
19.
Leukemia ; 15(5): 757-63, 2001 May.
Article in English | MEDLINE | ID: mdl-11368436

ABSTRACT

In order to improve leukemia-free survival we evaluated the feasibility and efficacy of autologous transplantation of interleukin-2 (IL-2)-mobilized peripheral blood progenitor cells for adult patients with acute myelogenous leukemia in first remission. Forty-nine consecutive patients (median age 49, range 21-70) with acute myelogenous leukemia in first remission were enrolled on a study of high-dose cytarabine/mitoxantrone consolidation chemotherapy with post-recovery IL-2 used as a method of in vivo purging for the purpose of autologous peripheral blood progenitor cell transplantation. A median of 2.08 x 10(6) CD34+ peripheral blood progenitor cells/kg were infused 1 day after preparative conditioning with 11.25 Gy total body irradiation and cyclophosphamide (120 mg/kg). Forty-one patients received myeloablative chemoradiotherapy followed by the infusion of IL-2-mobilized autologous peripheral blood progenitor cells. The median times to both neutrophil and platelet recovery were 16 days (range, 2-43) and 23 days (8-318+ days), respectively. Twenty-seven patients remain alive with 24 in continued first complete remission. Median remission duration for all eligible patients is 8 months, and actuarial leukemia-free survival is 49+/-15%. The actuarial risk of relapse is 43+/-16%. Toxicity of autologous peripheral blood progenitor cell transplant included treatment-related death in three patients and serious organ toxicity in 12. Advanced age was a negative prognostic factor for leukemia-free survival. Results were compared to an age-matched historical control treated with autologous transplantation of chemotherapy-mobilized progenitor cells; no significant difference in favor of IL-2 mobilization could be demonstrated. Our results demonstrate that autologous transplantation of IL-2-mobilized peripheral blood progenitor cells is feasible in an unselected population of adult patients with acute myelogenous leukemia in first remission with minimal toxicity but no clear evidence of benefit in leukemia-free survival.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Interleukin-2/pharmacology , Leukemia, Myeloid, Acute/therapy , Adult , Aged , Female , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Survival Rate , Transplantation, Autologous
20.
N Engl J Med ; 344(14): 1031-7, 2001 Apr 05.
Article in English | MEDLINE | ID: mdl-11287972

ABSTRACT

BACKGROUND: BCR-ABL is a constitutively activated tyrosine kinase that causes chronic myeloid leukemia (CML). Since tyrosine kinase activity is essential to the transforming function of BCR-ABL, an inhibitor of the kinase could be an effective treatment for CML. METHODS: We conducted a phase 1, dose-escalating trial of STI571 (formerly known as CGP 57148B), a specific inhibitor of the BCR-ABL tyrosine kinase. STI571 was administered orally to 83 patients with CML in the chronic phase in whom treatment with interferon alfa had failed. Patients were successively assigned to 1 of 14 doses ranging from 25 to 1000 mg per day. RESULTS: Adverse effects of STI571 were minimal; the most common were nausea, myalgias, edema, and diarrhea. A maximal tolerated dose was not identified. Complete hematologic responses were observed in 53 of 54 patients treated with daily doses of 300 mg or more and typically occurred in the first four weeks of therapy. Of the 54 patients treated with doses of 300 mg or more, cytogenetic responses occurred in 29, including 17 (31 percent of the 54 patients who received this dose) with major responses (0 to 35 percent of cells in metaphase positive for the Philadelphia chromosome); 7 of these patients had complete cytogenetic remissions. CONCLUSIONS: STI571 is well tolerated and has significant antileukemic activity in patients with CML in whom treatment with interferon alfa had failed. Our results provide evidence of the essential role of BCR-ABL tyrosine kinase activity in CML and demonstrate the potential for the development of anticancer drugs based on the specific molecular abnormality present in a human cancer.


Subject(s)
Enzyme Inhibitors/administration & dosage , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/administration & dosage , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/administration & dosage , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Benzamides , Blood Cell Count , Dose-Response Relationship, Drug , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Interferon-alpha/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Phosphorylation , Piperazines/adverse effects , Piperazines/pharmacokinetics , Protein-Tyrosine Kinases/metabolism , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Recurrence , Remission Induction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...