Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cytotherapy ; 24(7): 742-749, 2022 07.
Article in English | MEDLINE | ID: mdl-35219582

ABSTRACT

As cancer immunotherapies continue to expand across all areas of oncology, it is imperative to establish a standardized approach for defining and capturing clinically important toxicities, such as cytokine release syndrome (CRS). In this paper, we provide considerations for categorizing the variety of adverse events that may accompany CRS and for recognizing that presentations of CRS may differ among various immunotherapies (e.g., monoclonal antibodies, CAR T cell therapies and T cell engagers, which can include bispecific antibodies and other constructs). The goals of this paper are to ensure accurate and consistent identification of CRS in patients receiving immunotherapies in clinical studies to aid in reporting; enable more precise evaluation of the therapeutic risk-benefit profile and cross-study analyses; support evidence-based monitoring and management of important toxicities related to cancer immunotherapies; and improve patient care and outcomes. These efforts will become more important as the number and variety of molecular targets for immunotherapies broaden and as therapies with novel mechanisms continue to be developed.


Subject(s)
Cytokine Release Syndrome , Immunotherapy , Neoplasms , Antibodies, Bispecific , Clinical Trials as Topic , Cytokine Release Syndrome/etiology , Humans , Immunotherapy/adverse effects , Immunotherapy, Adoptive/adverse effects , Neoplasms/therapy
2.
Open Med (Wars) ; 12: 76-85, 2017.
Article in English | MEDLINE | ID: mdl-28730166

ABSTRACT

Genomic instability, including genetic mutations and chromosomal rearrangements, can lead to cancer development. Aberrant DNA methylation occurs commonly in cancer cells. The aim of this study is to determine the effects of a specific chromosomal lesion the BCR-ABL translocation t(9:22), in establishing DNA methylation profiles in cancer. Materials and methods We compared DNA methylation of 1,505 selected promoter CpGs in chronic myelogenous leukemia (CML), acute lymphoblastic leukemia (ALL) with and without the Philadelphia chromosome t(9:22), CD34+ hematopoietic stem cells transfected with BCR-ABL, and other tumors without BCR-ABL (acute promyelocytic leukemia (APL) and gastrointestinal stromal tumors (GIST). In this study, the DNA methylation profile of CML was more closely related to APL, another myeloid leukemia, than Ph+ ALL. Although DNA methylation profiles were consistent within a specific tumor type, overall DNA methylation profiles were no influenced by BCR-ABL gene translocation in the cancers and tissues studied. We conclude that DNA methylation profiles may reflect the cell of origin in cancers rather than the chromosomal lesions involved in leukemogenesis.

3.
Clin Cancer Res ; 23(16): 4550-4555, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28420721

ABSTRACT

Purpose: A phase I study was conducted to determine the MTD, dose-limiting toxicities (DLT), and pharmacokinetics of fenretinide delivered as an intravenous emulsion in relapsed/refractory hematologic malignancies.Experimental Design: Fenretinide (80-1,810 mg/m2/day) was administered by continuous infusion on days 1 to 5, in 21-day cycles, using an accelerated titration design.Results: Twenty-nine patients, treated with a median of three prior regimens (range, 1-7), were enrolled and received the test drug. Ninety-seven courses were completed. An MTD was reached at 1,280 mg/m2/day for 5 days. Course 1 DLTs included 6 patients with hypertriglyceridemia, 4 of whom were asymptomatic; 2 patients experienced DLT thrombocytopenia (asymptomatic). Of 11 patients with response-evaluable peripheral T-cell lymphomas, two had complete responses [CR, progression-free survival (PFS) 68+ months; unconfirmed CR, PFS 14+ months], two had unconfirmed partial responses (unconfirmed PR, PFS 5 months; unconfirmed PR, PFS 6 months), and five had stable disease (2-12 cycles). One patient with mature B-cell lymphoma had an unconfirmed PR sustained for two cycles. Steady-state plasma levels were approximately 10 mcg/mL (mid-20s µmol/L) at 640 mg/m2/day, approximately 14 mcg/mL (mid-30s µmol/L) at 905 mg/m2/day, and approximately 22 mcg/mL (mid-50s µmol/L) at 1,280 mg/m2/day.Conclusions: Intravenous fenretinide obtained significantly higher plasma levels than a previous capsule formulation, had acceptable toxicities, and evidenced antitumor activity in peripheral T-cell lymphomas. A recommended phase II dosing is 600 mg/m2 on day 1, followed by 1,200 mg/m2 on days 2 to 5, every 21 days. A registration-enabling phase II study in relapsed/refractory PTCL (ClinicalTrials.gov identifier: NCT02495415) is ongoing. Clin Cancer Res; 23(16); 4550-5. ©2017 AACR.


Subject(s)
Fenretinide/therapeutic use , Hematologic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , California , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Fenretinide/administration & dosage , Fenretinide/pharmacokinetics , Hematologic Neoplasms/pathology , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local , Remission Induction , Thrombocytopenia/chemically induced , Young Adult
5.
Semin Hematol ; 52(1): 12-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25578414

ABSTRACT

Thrombopoietin (TPO) is a growth factor that stimulates megakaryocytes to increases platelet counts. Due to concerns around the development of autoantibodies in clinical studies of TPO, a novel peptide that bound the TPO receptor was used to develop a TPO mimetic. This peptide has no shared homology with TPO, and therefore avoids the risk of development of antibodies that cross-react with endogenous TPO. The ability of the artificial peptide to bind and stimulate the TPO receptor was improved by dimerization of the peptide, and the stability of the peptide was improved by linking the peptides to the Fc portion of an antibody. Romiplostim (Nplate, Amgen, Inc, Thousand Oaks, CA) is the first fully engineered fusion of a novel peptide and antibody or "peptibody" that can stimulate platelet production.


Subject(s)
Recombinant Fusion Proteins/pharmacology , Thrombopoiesis/drug effects , Thrombopoietin/pharmacology , Animals , Antibodies , Blood Platelets/cytology , Blood Platelets/drug effects , Humans , Receptors, Fc
6.
Br J Haematol ; 167(3): 337-45, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25039607

ABSTRACT

Low endogenous erythropoietin levels and limited red blood cell transfusion history can predict response to erythropoiesis-stimulating agents in anaemic patients with myelodysplastic syndromes (MDS). The relationship between endogenous thrombopoietin (THPO) levels and platelet response to romiplostim is unknown. Variables including baseline endogenous THPO levels, transfusion needs, and platelet response were analysed in a randomized trial of 250 thrombocytopenic, lower-risk MDS patients (International Prognostic Scoring System low/intermediate-1). A predictive scoring system was developed based on log-likelihood ratios and logistic coefficients. Patients with HI-P (haematological improvement - platelets) responses had lower mean baseline THPO levels (P = 0·0497) and were more likely to have <6 platelet units transfused in the past year (P = 0·0027), as did patients with platelet responses ≥50% of weeks on romiplostim (P = 0·001 and P = 0·0037, respectively). A model for predicting response to romiplostim was developed and validated in a separate MDS cohort (N = 72). Patients in low-, intermediate-, and high-response groups had response rates of 17·4%, 29·6%, and 50·7%, respectively, for HI-P, and 17·4%, 33·8%, and 65·2%, respectively, for ≥50% response. For thrombocytopenic patients with lower-risk MDS, lower baseline THPO levels (<500 pg/ml) and limited platelet transfusion history predicted a greater likelihood of a subsequent platelet response to romiplostim.


Subject(s)
Hematinics/pharmacology , Models, Biological , Myelodysplastic Syndromes/drug therapy , Recombinant Fusion Proteins/pharmacology , Thrombocytopenia/drug therapy , Thrombopoiesis/drug effects , Thrombopoietin/pharmacology , Aged , Blood Transfusion , Combined Modality Therapy , Female , Hematinics/therapeutic use , Humans , Logistic Models , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/therapy , Platelet Count , Randomized Controlled Trials as Topic , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Risk Assessment , Thrombocytopenia/etiology , Thrombopoietin/blood , Thrombopoietin/therapeutic use
7.
Cancer ; 120(12): 1838-46, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24706489

ABSTRACT

BACKGROUND: Thrombocytopenia in patients with myelodysplastic syndrome (MDS) is associated with shortened survival and an increased risk of evolution to acute myeloid leukemia (AML). In this study, the authors evaluated the efficacy of romiplostim in patients who had thrombocytopenia with low-risk/intermediate-1-risk MDS. METHODS: Patients who had thrombocytopenia with low-risk/intermediate-1-risk MDS (N = 250) were randomized 2:1 to receive romiplostim or placebo weekly for 58 weeks. RESULTS: The primary endpoint- the number of clinically significant bleeding events (CSBEs) per patient-had a hazard ratio for romiplostim:placebo of 0.83 (95% confidence interval, 0.66-1.05; P = .13). CSBEs were reduced significantly in the romiplostim group for patients who had baseline platelet counts ≥20 × 10(9) /L (P < .0001). For patients who had baseline platelet counts <20 × 10(9) /L, there was no difference in the number of CSBEs, but the platelet transfusion rates were higher in the placebo group (P < .0001), which may have affected the overall CSBE results in this group with severe thrombocytopenia. The incidence of bleeding events was reduced significantly in the romiplostim group (relative risk, 0.92), as were protocol-defined platelet transfusions (relative risk, 0.77). Platelet response rates according to 2006 International Working Group criteria were higher for the group that received romiplostim (odds ratio, 15.6). On the basis of interim data, an independent data monitoring committee advised halting study drug because of concerns regarding excess blasts and AML rates with romiplostim (interim hazard ratio, 2.51). At 58 weeks, the AML rates were 6% in the romiplostim group and 4.9% in the placebo group (hazard ratio, 1.20; 95% confidence interval, 0.38-3.84), and the overall survival rates were similar. CONCLUSIONS: Romiplostim treatment in patients with low-risk/intermediate-1-risk MDS increased platelet counts and decreased the number of bleeding events and platelet transfusions. Although study drug was discontinued because of an initial concern of AML risk, survival and AML rates were similar with romiplostim and placebo.


Subject(s)
Myelodysplastic Syndromes/drug therapy , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Thrombocytopenia/drug therapy , Thrombopoietin/therapeutic use , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/pathology , Placebos , Survival Analysis , Thrombocytopenia/pathology , Treatment Outcome
8.
Leuk Lymphoma ; 54(2): 321-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22906162

ABSTRACT

Patients with myelodysplastic syndrome (MDS) receiving hypomethylating agents commonly develop thrombocytopenia. This double-blind study evaluated the efficacy and safety of romiplostim, a peptibody protein that increases platelets, in patients with MDS receiving decitabine. Patients received romiplostim 750 µg (n = 15) or placebo (n = 14) and decitabine. Median platelet counts at the beginning of each decitabine cycle trended lower in placebo-treated than in romiplostim-treated patients. Bleeding events occurred in 43% of placebo-treated and 27% of romiplostim-treated patients, and platelet transfusions were administered to 57% of placebo-treated and 47% of romiplostim-treated patients. Overall clinical therapeutic response was achieved by 21% of placebo-treated and 33% of romiplostim-treated patients. Treatment was generally well tolerated. Progression to acute myeloid leukemia (AML) occurred in one patient per group. Adding romiplostim to decitabine treatment is well tolerated and may be beneficial, as indicated by trends toward higher platelet counts at the beginning of each treatment cycle and lower platelet transfusion rates and percentages of patients with bleeding events.


Subject(s)
Myelodysplastic Syndromes/drug therapy , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Thrombopoietin/therapeutic use , Aged , Aged, 80 and over , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Decitabine , Female , Humans , Male , Middle Aged , Platelet Count , Receptors, Fc/administration & dosage , Receptors, Thrombopoietin/agonists , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Thrombopoietin/administration & dosage , Thrombopoietin/adverse effects , Treatment Outcome
9.
J Hematol Oncol ; 5: 71, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23190430

ABSTRACT

BACKGROUND: Lenalidomide treatment in myelodysplastic syndrome (MDS) may lead to thrombocytopenia and dose reductions/delays. This study evaluated the safety and tolerability of the thrombopoietin mimetic romiplostim and its effects on the incidence of clinically significant thrombocytopenic events (CSTEs) in lower risk MDS patients receiving lenalidomide. METHODS: Patients were assigned to weekly placebo (n = 12) or romiplostim 500 µg (n = 14) or 750 µg (n = 13) for four 28-day lenalidomide cycles. RESULTS: The treatment groups were generally similar with respect to baseline disease characteristics. Del(5q) abnormalities were noted in 1 (8%) patient in the placebo group, 3 (21%) in the romiplostim 500 µg group, and two (15%) in the 750 µg group. CSTEs were noted in 8 (67%) patients in the placebo group, 4 (29%) in the romiplostim 500 µg group, and 8 (62%) in the romiplostim 750 µg group. Throughout the study, median platelet counts trended lower in placebo-treated than in romiplostim-treated patients. Thrombocytopenia-related adjustments in lenalidomide occurred in 6 (50%) patients in the placebo group, 5 (36%) in the romiplostim 500 µg group, and 2 (15%) in the 750 µg group. Although the percentages of patients who received platelet transfusions were similar across treatment groups, there was a trend toward lower numbers of transfusions in both romiplostim groups during each treatment cycle. There were two serious treatment-related adverse events during the treatment period (cerebrovascular accident, placebo; worsening thrombocytopenia, romiplostim 500 µg). Two patients (romiplostim 500 and 750 µg, respectively) had an increase in bone marrow blasts to >20% during treatment, but had no post-treatment biopsy to confirm or exclude the diagnosis of progression to AML. CONCLUSIONS: These data suggest that romiplostim administered to MDS patients during lenalidomide treatment may decrease the frequency of dose reductions/delays due to thrombocytopenia. Additional study is needed to confirm the results of this preliminary trial. TRIAL REGISTRATION: ClinicalTrials.gov NCT00418665.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Double-Blind Method , Female , Humans , Lenalidomide , Male , Middle Aged , Placebos , Receptors, Fc/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives , Thrombopoietin/administration & dosage , Thrombopoietin/adverse effects
10.
J Biomed Sci ; 19: 13, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22300442

ABSTRACT

BACKGROUND: Retrotransposons have been extensively studied in plants and animals and have been shown to have an impact on human genome dynamics and evolution. Their ability to move within genomes gives retrotransposons to affect genome instability. METHODS: we examined the polymorphic inserted AluYa5, evolutionary young Alu, in the progesterone receptor gene to determine the effects of Alu insertion on molecular environment. We used mono-allelic inserted cell lines which carry both Alu-present and Alu-absent alleles. To determine the epigenetic change and gene expression, we performed restriction enzyme digestion, Pyrosequencing, and Chromatin Immunoprecipitation. RESULTS: We observed that the polymorphic insertion of evolutionally young Alu causes increasing levels of DNA methylation in the surrounding genomic area and generates inactive histone tail modifications. Consequently the Alu insertion deleteriously inactivates the neighboring gene expression. CONCLUSION: The mono-allelic Alu insertion cell line clearly showed that polymorphic inserted repetitive elements cause the inactivation of neighboring gene expression, bringing aberrant epigenetic changes.


Subject(s)
Alu Elements/genetics , DNA Methylation/genetics , Genome, Human , Transcription, Genetic , Alleles , Cell Line , Epigenesis, Genetic/genetics , Evolution, Molecular , Gene Expression Regulation , Genomic Instability , Humans , Receptors, Progesterone/genetics
11.
Int J Epidemiol ; 41(1): 126-39, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20846947

ABSTRACT

BACKGROUND: Estimates of global DNA methylation from repetitive DNA elements, such as Alu and LINE-1, have been increasingly used in epidemiological investigations because of their relative low-cost, high-throughput and quantitative results. Nevertheless, determinants of these methylation measures in healthy individuals are still largely unknown. The aim of this study was to examine whether age, gender, smoking habits, alcohol drinking and body mass index (BMI) are associated with Alu or LINE-1 methylation levels in blood leucocyte DNA of healthy individuals. METHODS: Individual data from five studies including a total of 1465 healthy subjects were combined. DNA methylation was quantified by PCR-pyrosequencing. RESULTS: Age [ß = -0.011% of 5-methyl-cytosine (%5 mC)/year, 95% confidence interval (CI) -0.020 to -0.001%5 mC/year] and alcohol drinking (ß = -0.214, 95% CI -0.415 to -0.013) were inversely associated with Alu methylation. Compared with females, males had lower Alu methylation (ß = -0.385, 95% CI -0.665 to -0.104) and higher LINE-1 methylation (ß = 0.796, 95% CI 0.261 to 1.330). No associations were found with smoking or BMI. Percent neutrophils and lymphocytes in blood counts exhibited a positive (ß = 0.036, 95% CI 0.010 to 0.061) and negative (ß = -0.038, 95% CI -0.065 to -0.012) association with LINE-1 methylation, respectively. CONCLUSIONS: Global methylation measures in blood DNA vary in relation with certain host and lifestyle characteristics, including age, gender, alcohol drinking and white blood cell counts. These findings need to be considered in designing epidemiological investigations aimed at identifying associations between DNA methylation and health outcomes.


Subject(s)
Alu Elements/genetics , DNA Methylation , DNA/blood , Long Interspersed Nucleotide Elements/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Alcohol Drinking , Body Mass Index , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Risk Factors , Sex Factors , Smoking , Young Adult
12.
Nucleic Acids Res ; 39(1): 104-18, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20841325

ABSTRACT

DNA methyltransferases (DNMTs) play an important role in establishing and maintaining DNA methylation. Aberrant expression of DNMTs and their isoforms has been found in many types of cancer, and their contribution to aberrant DNA methylation has been proposed. Here, we generated HEK 293T cells stably transfected with each of 13 different DNMTs (DNMT1, two DNMT3A isoforms, nine DNMT3B isoforms and DNMT3L) and assessed the DNA methylation changes induced by each DNMT. We obtained DNA methylation profiles of DNA repetitive elements and 1505 CpG sites from 808 cancer-related genes. We found that DNMTs have specific and overlapping target sites and their DNA methylation target profiles are a reflection of the DNMT domains. By examining H3K4me3 and H3K27me3 modifications in the 808 gene promoter regions using promoter ChIP-on-chip analysis, we found that specific de novo DNA methylation target sites of DNMT3A1 are associated with H3K4me3 modification that are transcriptionally active, whereas the specific target sites of DNMT3B1 are associated with H3K27me3 modification that are transcriptionally inactive. Our data suggest that different DNMT domains are responsible for targeting DNA methylation to specific regions of the genome, and this targeting might be associated with histone modifications.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methylation , DNA Methyltransferase 3A , Genes, Neoplasm , HEK293 Cells , Histones/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , Repetitive Sequences, Nucleic Acid , DNA Methyltransferase 3B
13.
Oncol Lett ; 2(1): 181-187, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22870150

ABSTRACT

To define the correlation between BCR promoter DNA methylation and response to imatinib in chronic myeloid leukemia (CML), we investigated BCR promoter DNA methylation in three groups of subjects. The first group included chronic phase patients enrolled in an imatinib dose escalation trial. In the trial, patients who failed to achieve optimal response with 400 mg/day (suboptimal responders) received an escalated imatinib dose. The level of BCR promoter DNA methylation was quantitated at baseline six months after dose escalation. The second group included patients who achieved complete cytogenetic remission after receiving 400 mg/day of imatinib (optimal responders), and the third group were the healthy controls. In the suboptimal responders, an increased BCR promoter DNA methylation at six months compared with the baseline was related to a rapid reduction in the BCR-ABL/ABL transcript level following dose escalation (p=0.001) and a longer time to treatment failure (TTFx) of the dose-escalated imatinib (p=0.008). When multivariate analysis was performed with regard to the baseline BCR-ABL transcript level, baseline BCR promoter DNA methylation, and a change in the BCR promoter DNA methylation following dose escalation, the increase in the BCR promoter DNA methylation following dose escalation was an independent predictive factor for TTFx of dose-escalated imatinib (hazard ratio, 0.294; p=0.015). The baseline BCR promoter DNA methylation level in the suboptimal responders was lower than that in BCR promoter DNA methylation in the optimal responders (p=0.001) and healthy controls (p<0.001). In both the optimal and suboptimal responders, BCR promoter DNA methylation had an inverse correlation with the duration of the 400 mg/day imatinib use. In conclusion, increased BCR promoter DNA methylation strongly correlates with a more favorable imatinib response in CML patients.

14.
Epigenetics ; 5(8): 750-7, 2010.
Article in English | MEDLINE | ID: mdl-20861661

ABSTRACT

This study aims to determine the optimal dosing and administration route for azacitidine to reduce global DNA methylation levels in the peripheral blood of patients with hematologic malignancies. Seventeen patients were enrolled into one of five dose level treatment groups (3 at 25 mg/m², 4 at 50 mg/m², 4 at 75 mg/m², 3 at 100 mg/m², and 3 at 150 mg/m²) and received IV azacitidine at their respective dose on days 1-5 of cycle one. All patients received 75 mg/m²/day IV on days 1-5 of cycle 2. Subcutaneous dosing of 75 mg/m²/day on days 1-5 was used in cycle 3. Peripheral blood was collected on days 1, 3, and 5 of each cycle, and global DNA methylation was measured using bisulfite-PCR pyrosequencing of the DNA repetitive element LINE-1. 14 patients were evaluable for response with 2 CR, 2 PR, 7 SD and 3 PD reported. LINE-1 DNA methylation decreased by 1.37, 2.29, 4.81, 1.94 and 4.05% on day 5 for the 25 mg/m², 50 mg/m², 75 mg/m², 100mg/m² and 150 mg/m² cycle one dose levels respectively. Mean decrease in LINE-1 DNA methylation was 3.7% with 75 mg/m² IV and 3.4% by subcutaneous administration. The data indicates that 75 mg/m² azacitidine given IV or SC effectively leads to global DNA methylation reduction by LINE-1 analysis.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Azacitidine/administration & dosage , DNA Methylation/drug effects , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Clinical Trials, Phase I as Topic , Dose-Response Relationship, Drug , Female , Humans , Injections, Subcutaneous , Long Interspersed Nucleotide Elements , Male , Middle Aged , Time Factors
15.
PLoS Genet ; 6(4): e1000917, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421991

ABSTRACT

It was recently shown that a large portion of the human transcriptome can originate from within repetitive elements, leading to ectopic expression of protein-coding genes. However the mechanism of transcriptional activation of repetitive elements has not been definitively elucidated. For the first time, we directly demonstrate that hypomethylation of retrotransposons can cause altered gene expression in humans. We also reveal that active LINE-1s switch from a tetranucleosome to dinucleosome structure, acquiring H2A.Z- and nucleosome-free regions upstream of TSSs, previously shown only at active single-copy genes. Hypomethylation of a specific LINE-1 promoter was also found to induce an alternate transcript of the MET oncogene in bladder tumors and across the entire urothelium of tumor-bearing bladders. These data show that, in addition to contributing to chromosomal instability, hypomethylation of LINE-1s can alter the functional transcriptome and plays a role not only in human disease but also in disease predisposition.


Subject(s)
Long Interspersed Nucleotide Elements/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-met/genetics , Receptors, Growth Factor/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Cell Line, Tumor , Chromatin Assembly and Disassembly , DNA Methylation/genetics , Humans , Middle Aged , Oncogenes , Transcriptional Activation
16.
Epigenomics ; 2(1): 71-86, 2010 Feb.
Article in English | MEDLINE | ID: mdl-22122748

ABSTRACT

The recent approval of azacitidine (Vidaza®), decitabine (Dacogen®) and vorinostat (Zolinza™) for myelodysplastic syndrome and cutaneous T-cell lymphoma has led to a wave of interest in epigenetic therapy. These DNA methylation inhibitors and the histone deacetylase inhibitor clearly have demonstrated activity in hematologic malignancies, but the future role of epigenetic therapy in solid tumors is still unknown. What is not commonly known is that azacitidine and decitabine were originally developed as cytotoxic nucleoside analogs and clinical trials were previously conducted in a variety of cancer types prior to the knowledge of their ability to inhibit DNA methylation. We review the experience of azacitidine and decitabine in early clinical trials and demonstrate the activity of epigenetic therapy in solid tumors.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Epigenesis, Genetic/drug effects , Immunotherapy/methods , Neoplasms/drug therapy , Azacitidine/chemistry , Azacitidine/therapeutic use , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/physiology , Decitabine , Dose-Response Relationship, Drug , Epigenesis, Genetic/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Molecular Structure , Neoplasms/immunology
17.
J Hematol Oncol ; 2: 46, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19909541

ABSTRACT

Tyrosine kinase inhibitors (TKIs) like dasatinib and nilotinib are indicated as second-line treatment for chronic myeloid leukemia resistant or intolerant to the current first-line TKI imatinib. These are agents are well tolerated, but potent and as such should be monitored for potentially serious side-effects like fluid retention and pleural effusions. Here we present key clinical trial data and safety considerations for all FDA approved TKIs in context for effective management of fluid retention and pleural effusions. Altering the dasatinib regimen from 70 mg twice daily to 100 mg daily reduces the risk of pleural effusion for patients taking dasatinib. Should pleural effusion develop, dasatinib should be interrupted until the condition resolves. Patients with a history of pleural effusion risk factors should be monitored closely while taking dasatinib. Patients receiving imatinib and nilotinib are not without risk of fluid retention. All patients should also be educated to recognize and report key symptoms of fluid retention or pleural effusion. Pleural effusions are generally managed by dose interruption/reduction and other supportive measures in patients with chronic myeloid leukemia receiving dasatinib therapy.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Pleural Effusion/chemically induced , Pleural Effusion/epidemiology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/adverse effects , Thiazoles/adverse effects , Algorithms , Dasatinib , Humans , Incidence , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Water-Electrolyte Imbalance/chemically induced , Water-Electrolyte Imbalance/epidemiology
18.
Hum Mol Genet ; 18(24): 4808-17, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19776032

ABSTRACT

DNA methylation is known to be associated with cell differentiation, aging, disease and cancer. There exists an expanding base of knowledge regarding tissue-specific DNA methylation, but we have little information about person-specific DNA methylation. Here, we analyze the DNA methylation patterns of multiple tissues from multiple individuals using a high-throughput quantitative assay of genome-wide DNA methylation, namely the Illumina GoldenGate BeadArray. DNA methylation patterns were largely conserved across 11 different tissues (r = 0.852) and across six individuals (r = 0.829), and we found that DNA was highly methylated in non-CpG islands and/or CpG sites that are not occupied by either H3K4me3 or H3K27me3 (P < 0.05). Finally, we found that the Illumina GoldenGate assay features a large number of probes (265/1505 probes, 17.6%) that contain single-nucleotide polymorphisms, which may interfere with DNA methylation analyses in genome-wide studies.


Subject(s)
CpG Islands , DNA Methylation , Epigenesis, Genetic , Autopsy , Genome-Wide Association Study/methods , Humans , Polymorphism, Single Nucleotide
19.
J Hematol Oncol ; 2: 30, 2009 Jul 24.
Article in English | MEDLINE | ID: mdl-19630970

ABSTRACT

BACKGROUND: In the last decade the importance of ethnicity, socio-economic and gender differences in relation to disease incidence, diagnosis, and prognosis has been realized. Differences in these areas have become a major health policy focus in the United States. Our study was undertaken to examine the demographic and clinical features of chronic myelogenous leukemia (CML) patients presenting initially at the LAC+USC Medical Center, which serves an ethnically diverse population. RESULTS: Patients were evenly split by gender, overwhelmingly Hispanic (60.9%), and quite young (median age 39, range 17-65) compared with previously reported CML patient populations. Previous CML studies identified significant anemia (Hgb <12 g/dl), significant thrombocytosis (platelets >450 x 109/l), and significant leukocytosis (WBC >50 x 109/l) as significant adverse pretreatment prognostic factors. Using these indicators, in addition to the validated Hasford and Sokal scores, patients were stratified and analyzed via gender and ethnicity. A significantly greater proportion of women presented with significant anemia (p = 0.019, Fisher's exact test) and significant thrombocytosis (p = 0.041, Fisher's exact test) compared to men, although no differences were found in risk stratification or treatment response. MCV values for women were significantly (p = 0.02, 2-sample t-test) lower than those for men, suggesting iron deficiency anemia. Focusing on ethnicity, Hispanics as a whole had significantly lower Hasford risk stratification (p = 0.046, Fisher's exact test), and significantly greater likelihood (p = 0.016, Fisher's exact test) of achieving 3-month complete haematological remission (CHR) compared with non-Hispanics at LAC+USC Medical Center, though differences in treatment outcome were no longer significant with analysis limited to patients treated with first-line imatinib. CONCLUSION: Female CML patients at LAC+USC Medical Center present with more significant adverse pre-treatment prognostic factors compared to men, but achieve comparable outcomes. Hispanic patients present with lower risk profile CML and achieve better treatment responses compared to non-Hispanic patients as a whole; these ethnic differences are no longer significant when statistical analysis is limited to patients given imatinib as first-line therapy. Our patients achieve response rates inferior to those of large-scale national studies. This constellation of findings has not been reported in previous studies, and is likely reflective of a unique patient population.


Subject(s)
Ethnicity , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Sex Characteristics , Adolescent , Adult , Aged , Cohort Studies , Ethnicity/statistics & numerical data , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/epidemiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/ethnology , Los Angeles/epidemiology , Male , Middle Aged , Prognosis , Research Design , Retrospective Studies , Treatment Outcome , Validation Studies as Topic , Young Adult
20.
Mol Cell Biol ; 29(19): 5366-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19620278

ABSTRACT

Proper DNA methylation patterns are essential for mammalian development and differentiation. DNA methyltransferases (DNMTs) primarily establish and maintain global DNA methylation patterns; however, the molecular mechanisms for the generation and inheritance of methylation patterns are still poorly understood. We used sucrose density gradients of nucleosomes prepared by partial and maximum micrococcal nuclease digestion, coupled with Western blot analysis to probe for the interactions between DNMTs and native nucleosomes. This method allows for analysis of the in vivo interactions between the chromatin modification enzymes and their actual nucleosomal substrates in the native state. We show that little free DNA methyltransferase 3A and 3B (DNMT3A/3B) exist in the nucleus and that almost all of the cellular contents of DNMT3A/3B, but not DNMT1, are strongly anchored to a subset of nucleosomes. This binding of DNMT3A/3B does not require the presence of other well-known chromatin-modifying enzymes or proteins, such as proliferating cell nuclear antigen, heterochromatin protein 1, methyl-CpG binding protein 2, Enhancer of Zeste homolog 2, histone deacetylase 1, and UHRF1, but it does require an intact nucleosomal structure. We also show that nucleosomes containing methylated SINE and LINE elements and CpG islands are the main sites of DNMT3A/3B binding. These data suggest that inheritance of DNA methylation requires cues from the chromatin component in addition to hemimethylation.


Subject(s)
CpG Islands , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Nucleosomes/enzymology , Cell Line , Chromatin , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Binding , Repetitive Sequences, Nucleic Acid , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL
...