Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Med ; 29(9): 2286-2294, 2023 09.
Article in English | MEDLINE | ID: mdl-37592106

ABSTRACT

Idecabtagene vicleucel (ide-cel) is a B-cell-maturation antigen (BCMA)-directed chimeric antigen receptor T cell therapy. We performed a post hoc analysis of a single-arm phase 1 multicenter study in relapsed/refractory multiple myeloma (CRB-401) (n = 62; median follow-up, 18.1 months). The primary endpoint was safety outcomes, and secondary endpoints included overall response rate (ORR), complete response (CR) and very good partial response (VGPR). The study met its primary endpoint with low rates of grade 3/grade 4 cytokine release syndrome (6.5%) and neurotoxicity (1.6%). ORR was 75.8%; 64.5% achieved VGPR or better and 38.7% achieved CR or stringent CR. Among exploratory endpoints, median duration of response, progression-free survival (PFS) and overall survival were 10.3, 8.8 and 34.2 months, respectively, and ide-cel expansion in blood and bone marrow correlated with clinical efficacy and postinfusion reduction of soluble BCMA. Patients with PFS ≥ 18 months had more naive and less exhausted T cells in apheresis material and improved functional T cell phenotype in the drug product compared with those with less durable responses. These results confirm ide-cel safety, tolerability and efficacy and describe T cell qualities that correlate with durable response. Clinicaltrials.gov identifier : NCT02658929 .


Subject(s)
Multiple Myeloma , Receptors, Chimeric Antigen , Humans , Multiple Myeloma/therapy , B-Cell Maturation Antigen , Follow-Up Studies , Cytokine Release Syndrome
2.
N Engl J Med ; 384(8): 705-716, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33626253

ABSTRACT

BACKGROUND: Idecabtagene vicleucel (ide-cel, also called bb2121), a B-cell maturation antigen-directed chimeric antigen receptor (CAR) T-cell therapy, has shown clinical activity with expected CAR T-cell toxic effects in patients with relapsed and refractory multiple myeloma. METHODS: In this phase 2 study, we sought to confirm the efficacy and safety of ide-cel in patients with relapsed and refractory myeloma. Patients with disease after at least three previous regimens including a proteasome inhibitor, an immunomodulatory agent, and an anti-CD38 antibody were enrolled. Patients received ide-cel target doses of 150 × 106 to 450 × 106 CAR-positive (CAR+) T cells. The primary end point was an overall response (partial response or better); a key secondary end point was a complete response or better (comprising complete and stringent complete responses). RESULTS: Of 140 patients enrolled, 128 received ide-cel. At a median follow-up of 13.3 months, 94 of 128 patients (73%) had a response, and 42 of 128 (33%) had a complete response or better. Minimal residual disease (MRD)-negative status (<10-5 nucleated cells) was confirmed in 33 patients, representing 26% of all 128 patients who were treated and 79% of the 42 patients who had a complete response or better. The median progression-free survival was 8.8 months (95% confidence interval, 5.6 to 11.6). Common toxic effects among the 128 treated patients included neutropenia in 117 patients (91%), anemia in 89 (70%), and thrombocytopenia in 81 (63%). Cytokine release syndrome was reported in 107 patients (84%), including 7 (5%) who had events of grade 3 or higher. Neurotoxic effects developed in 23 patients (18%) and were of grade 3 in 4 patients (3%); no neurotoxic effects higher than grade 3 occurred. Cellular kinetic analysis confirmed CAR+ T cells in 29 of 49 patients (59%) at 6 months and 4 of 11 patients (36%) at 12 months after infusion. CONCLUSIONS: Ide-cel induced responses in a majority of heavily pretreated patients with refractory and relapsed myeloma; MRD-negative status was achieved in 26% of treated patients. Almost all patients had grade 3 or 4 toxic effects, most commonly hematologic toxic effects and cytokine release syndrome. (Funded by bluebird bio and Celgene, a Bristol-Myers Squibb company; KarMMa ClinicalTrials.gov number, NCT03361748.).


Subject(s)
Immunotherapy, Adoptive , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/therapeutic use , Adult , Aged , Biomarkers/blood , Cytokine Release Syndrome/etiology , Drug Resistance, Neoplasm , Female , Hematologic Diseases/chemically induced , Humans , Immunotherapy, Adoptive/adverse effects , Male , Middle Aged , Multiple Myeloma/immunology , Progression-Free Survival , Recurrence
3.
Nat Commun ; 12(1): 868, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558511

ABSTRACT

BCMA targeting chimeric antigen receptor (CAR) T cell therapy has shown deep and durable responses in multiple myeloma. However, relapse following therapy is frequently observed, and mechanisms of resistance remain ill-defined. Here, we perform single cell genomic characterization of longitudinal samples from a patient who relapsed after initial CAR T cell treatment with lack of response to retreatment. We report selection, following initial CAR T cell infusion, of a clone with biallelic loss of BCMA acquired by deletion of one allele and a mutation that creates an early stop codon on the second allele. This loss leads to lack of CAR T cell proliferation following the second infusion and is reflected by lack of soluble BCMA in patient serum. Our analysis suggests the need for careful detection of BCMA gene alterations in multiple myeloma cells from relapse following CAR T cell therapy.


Subject(s)
Alleles , B-Cell Maturation Antigen/genetics , Drug Resistance, Neoplasm , Immunotherapy, Adoptive , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Bone Marrow/pathology , Humans , Multiple Myeloma/immunology , Tumor Microenvironment
4.
N Engl J Med ; 380(18): 1726-1737, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31042825

ABSTRACT

BACKGROUND: Preclinical studies suggest that bb2121, a chimeric antigen receptor (CAR) T-cell therapy that targets B-cell maturation antigen (BCMA), has potential for the treatment of multiple myeloma. METHODS: In this phase 1 study involving patients with relapsed or refractory multiple myeloma, we administered bb2121 as a single infusion at doses of 50×106, 150×106, 450×106, or 800×106 CAR-positive (CAR+) T cells in the dose-escalation phase and 150×106 to 450×106 CAR+ T cells in the expansion phase. Patients had received at least three previous lines of therapy, including a proteasome inhibitor and an immunomodulatory agent, or were refractory to both drug classes. The primary end point was safety. RESULTS: Results for the first 33 consecutive patients who received a bb2121 infusion are reported. The data-cutoff date was 6.2 months after the last infusion date. Hematologic toxic effects were the most common events of grade 3 or higher, including neutropenia (in 85% of the patients), leukopenia (in 58%), anemia (in 45%), and thrombocytopenia (in 45%). A total of 25 patients (76%) had cytokine release syndrome, which was of grade 1 or 2 in 23 patients (70%) and grade 3 in 2 patients (6%). Neurologic toxic effects occurred in 14 patients (42%) and were of grade 1 or 2 in 13 patients (39%). One patient (3%) had a reversible grade 4 neurologic toxic effect. The objective response rate was 85%, including 15 patients (45%) with complete responses. Six of the 15 patients who had a complete response have had a relapse. The median progression-free survival was 11.8 months (95% confidence interval, 6.2 to 17.8). All 16 patients who had a response (partial response or better) and who could be evaluated for minimal residual disease (MRD) had MRD-negative status (≤10-4 nucleated cells). CAR T-cell expansion was associated with responses, and CAR T cells persisted up to 1 year after the infusion. CONCLUSIONS: We report the initial toxicity profile of a BCMA-directed cellular immunotherapy for patients with relapsed or refractory multiple myeloma. Antitumor activity was documented. (Funded by Bluebird Bio and Celgene; CRB-401 ClinicalTrials.gov number, NCT02658929.).


Subject(s)
B-Cell Maturation Antigen/antagonists & inhibitors , Immunotherapy, Adoptive , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/therapeutic use , Adult , Aged , CD4-CD8 Ratio , Female , Humans , Immunotherapy, Adoptive/adverse effects , Infusions, Intravenous , Male , Middle Aged , Multiple Myeloma/immunology , Progression-Free Survival , T-Lymphocytes/metabolism
5.
Mol Cancer Ther ; 16(12): 2849-2861, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28878028

ABSTRACT

Prognosis of triple-negative breast cancer (TNBC) remains poor. To identify shared and selective vulnerabilities of basal-like TNBC, the most common TNBC subtype, a directed siRNA lethality screen was performed in 7 human breast cancer cell lines, focusing on 154 previously identified dependency genes of 1 TNBC line. Thirty common dependency genes were identified, including multiple proteasome and RNA splicing genes, especially those associated with the U4/U6.U5 tri-snRNP complex (e.g., PRPF8, PRPF38A). PRPF8 or PRPF38A knockdown or the splicing modulator E7107 led to widespread intronic retention and altered splicing of transcripts involved in multiple basal-like TNBC dependencies, including protein homeostasis, mitosis, and apoptosis. E7107 treatment suppressed the growth of basal-A TNBC cell line and patient-derived basal-like TNBC xenografts at a well-tolerated dose. The antitumor response was enhanced by adding the proteasome inhibitor bortezomib. Thus, inhibiting both splicing and the proteasome might be an effective approach for treating basal-like TNBC. Mol Cancer Ther; 16(12); 2849-61. ©2017 AACR.


Subject(s)
RNA Splicing/genetics , Triple Negative Breast Neoplasms/genetics , Cell Proliferation , Female , Humans , Prognosis , Survival Analysis , Triple Negative Breast Neoplasms/mortality
6.
Cancers (Basel) ; 9(7)2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28718815

ABSTRACT

Chimeric Antigen Receptor (CAR) T-cells are T-cells with recombinant receptors targeted to tumor antigens. CAR-T cell therapy has emerged as a mode of immunotherapy and is now being extensively explored in hematologic cancer. In contrast, CAR-T cell use in solid tumors has been hampered by multiple obstacles. Several approaches have been taken to circumvent these obstacles, including the regional delivery of CAR-T cells. Regional CAR-T cell delivery can theoretically compensate for poor T-cell trafficking and tumor antigen specificity while avoiding systemic toxicity associated with intravenous delivery. We reviewed completed clinical trials for the treatment of glioblastoma and metastatic colorectal cancer and examined the data in these studies for safety, efficacy, and potential advantages that regional delivery may confer over systemic delivery. Our appraisal of the available literature revealed that regional delivery of CAR-T cells in both glioblastoma and hepatic colorectal metastases was generally well tolerated and efficacious in select instances. We propose that the regional delivery of CAR-T cells is an area of potential growth in the solid tumor immunotherapy, and look towards future clinical trials in head and neck cancer, mesothelioma, and peritoneal carcinomatosis as the use of this technique expands.

7.
Mol Cancer Ther ; 14(10): 2279-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26264278

ABSTRACT

Effective therapeutic strategies for in vivo siRNA delivery to knockdown genes in cells outside the liver are needed to harness RNA interference for treating cancer. EpCAM is a tumor-associated antigen highly expressed on common epithelial cancers and their tumor-initiating cells (TIC, also known as cancer stem cells). Here, we show that aptamer-siRNA chimeras (AsiC, an EpCAM aptamer linked to an siRNA sense strand and annealed to the siRNA antisense strand) are selectively taken up and knock down gene expression in EpCAM(+) cancer cells in vitro and in human cancer biopsy tissues. PLK1 EpCAM-AsiCs inhibit colony and mammosphere formation (in vitro TIC assays) and tumor initiation by EpCAM(+) luminal and basal-A triple-negative breast cancer (TNBC) cell lines, but not EpCAM(-) mesenchymal basal-B TNBCs, in nude mice. Subcutaneously administered EpCAM-AsiCs concentrate in EpCAM(+) Her2(+) and TNBC tumors and suppress their growth. Thus, EpCAM-AsiCs provide an attractive approach for treating epithelial cancer.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Breast Neoplasms/pathology , Neoplasms, Glandular and Epithelial/pathology , Neoplastic Stem Cells/physiology , RNA, Small Interfering/administration & dosage , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Base Sequence , Breast Neoplasms/metabolism , Carcinogenesis/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Female , Gene Expression , Gene Knockdown Techniques , Humans , Mice, Nude , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Burden , Polo-Like Kinase 1
8.
Cancer Cell ; 24(2): 182-96, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23948298

ABSTRACT

Basal-like triple-negative breast cancers (TNBCs) have poor prognosis. To identify basal-like TNBC dependencies, a genome-wide siRNA lethality screen compared two human breast epithelial cell lines transformed with the same genes: basal-like BPLER and myoepithelial HMLER. Expression of the screen's 154 BPLER dependency genes correlated with poor prognosis in breast, but not lung or colon, cancer. Proteasome genes were overrepresented hits. Basal-like TNBC lines were selectively sensitive to proteasome inhibitor drugs relative to normal epithelial, luminal, and mesenchymal TNBC lines. Proteasome inhibition reduced growth of established basal-like TNBC tumors in mice and blocked tumor-initiating cell function and macrometastasis. Proteasome addiction in basal-like TNBCs was mediated by NOXA and linked to MCL-1 dependence.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Proteasome Endopeptidase Complex/genetics , RNA, Small Interfering/analysis , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Prognosis , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering/genetics
9.
J Clin Oncol ; 29(6): 747-54, 2011 Feb 20.
Article in English | MEDLINE | ID: mdl-21079135

ABSTRACT

Cancer therapeutics still fall far short of our goals for treating patients with locally advanced or metastatic disease. Until recently, almost all cancer drugs were crude cytotoxic agents that discriminate poorly between cancer cells and normally dividing cells. The development of targeted biologics that recognize tumor cell surface antigens and of specific inhibitors of pathways dysregulated in cancer cells or normal cellular pathways on which a cancer cell differentially depends has provided hope for converting our increasing understanding of cellular transformation into intelligently designed anticancer therapeutics. However, new drug development is painfully slow, and the pipeline of new therapeutics is thin. The discovery of RNA interference (RNAi), a ubiquitous cellular pathway of gene regulation that is dysregulated in cancer cells, provides an exciting opportunity for relatively rapid and revolutionary approaches to cancer drug design. Small RNAs that harness the RNAi machinery may become the next new class of drugs for treating a variety of diseases. Although it has only been 9 years since RNAi was shown to work in mammalian cells, about a dozen phase I to III clinical studies have already been initiated, including four for cancer. So far there has been no unexpected toxicity and suggestions of benefit in one phase II study. However, the obstacles for RNAi-based cancer therapeutics are substantial. This article will discuss how the endogenous RNAi machinery might be harnessed for cancer therapeutics, why academic researchers and biotech and pharmaceutical companies are so excited, and what the obstacles are and how they might be overcome.


Subject(s)
Genetic Therapy/trends , Neoplasms/genetics , Neoplasms/therapy , RNA Interference/physiology , RNA, Small Interfering/therapeutic use , Clinical Trials as Topic , Genetic Therapy/methods , Humans , RNA, Small Interfering/genetics
10.
PLoS One ; 4(9): e7181, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19787069

ABSTRACT

BACKGROUND: The development of metastases involves the dissociation of cells from the primary tumor to penetrate the basement membrane, invade and then exit the vasculature to seed, and colonize distant tissues. The last step, establishment of macroscopic tumors at distant sites, is the least well understood. Four isogenic mouse breast cancer cell lines (67NR, 168FARN, 4TO7, and 4T1) that differ in their ability to metastasize when implanted into the mammary fat pad are used to model the steps of metastasis. Only 4T1 forms macroscopic lung and liver metastases. Because some miRNAs are dysregulated in cancer and affect cellular transformation, tumor formation, and metastasis, we examined whether changes in miRNA expression might explain the differences in metastasis of these cells. METHODOLOGY/PRINCIPAL FINDINGS: miRNA expression was analyzed by miRNA microarray and quantitative RT-PCR in isogenic mouse breast cancer cells with distinct metastatic capabilities. 4T1 cells that form macroscopic metastases had elevated expression of miR-200 family miRNAs compared to related cells that invade distant tissues, but are unable to colonize. Moreover, over-expressing miR-200 in 4TO7 cells enabled them to metastasize to lung and liver. These findings are surprising since the miR-200 family was previously shown to promote epithelial characteristics by inhibiting the transcriptional repressor Zeb2 and thereby enhancing E-cadherin expression. We confirmed these findings in these cells. The most metastatic 4T1 cells acquired epithelial properties (high expression of E-cadherin and cytokeratin-18) compared to the less metastatic cells. CONCLUSIONS/SIGNIFICANCE: Expression of miR-200, which promotes a mesenchymal to epithelial cell transition (MET) by inhibiting Zeb2 expression, unexpectedly enhances macroscopic metastases in mouse breast cancer cell lines. These results suggest that for some tumors, tumor colonization at metastatic sites might be enhanced by MET. Therefore the epithelial nature of a tumor does not predict metastatic outcome.


Subject(s)
Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Cadherins/biosynthesis , Cell Line, Tumor , Cell Transformation, Neoplastic , Cloning, Molecular , Epithelium/metabolism , Female , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Microscopy, Fluorescence/methods , Neoplasm Metastasis , Treatment Outcome , Zinc Finger E-box-Binding Homeobox 1
11.
RNA Biol ; 6(3): 335-40, 2009.
Article in English | MEDLINE | ID: mdl-19535911

ABSTRACT

Because microRNAs regulate cancer cell differentiation, proliferation, survival and metastasis, manipulating microRNA function, either by mimicking or inhibiting miRNAs implicated in cancer, could provide a powerful therapeutic strategy to interfere with key pathways for cancer progression. This review will explore some of the opportunities and obstacles to harnessing microRNA biology for cancer therapy.


Subject(s)
MicroRNAs/therapeutic use , Neoplasms/genetics , Neoplasms/therapy , Animals , Drug Delivery Systems , Gene Expression Regulation, Neoplastic , Genetic Therapy , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , Neoplasms/pathology , Neoplasms/prevention & control
12.
Adv Immunol ; 102: 227-44, 2009.
Article in English | MEDLINE | ID: mdl-19477322

ABSTRACT

MicroRNAs (miRNAs) regulate cell fate during development and in response to environmental cues. Here, we review the emerging story of how miRNAs regulate immune cell development and function.


Subject(s)
Lymphocytes/immunology , MicroRNAs/physiology , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Immunity , Virus Replication
15.
Cancer Res ; 68(20): 8191-4, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922889

ABSTRACT

Inactivation of the transforming growth factor beta (TGFbeta) tumor suppressor pathway is a main step in the development of a variety of human tumors. The miR-106b-25 and miR-17-92 clusters are emerging as key modulators of TGFbeta signaling in gastrointestinal and other tumors, interfering with cell cycle arrest and apoptosis when overexpressed in cancer cells. Genetic ablation of these microRNAs (miRNAs) reveals their physiologic role in the control of liver and central nervous system apoptosis, supporting the notion that miRNA-based homeostatic mechanisms can be usurped by cancer cells to resist TGFbeta tumor suppression.


Subject(s)
MicroRNAs/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins/physiology , Bcl-2-Like Protein 11 , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA-Binding Proteins/genetics , E2F1 Transcription Factor/physiology , Genes, myc , Humans , Membrane Proteins/physiology , Minichromosome Maintenance Complex Component 7 , Multigene Family , Neoplasms/etiology , Nuclear Proteins/genetics , Proto-Oncogene Proteins/physiology , RNA Interference
16.
Cancer Res ; 68(15): 6162-70, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676839

ABSTRACT

MicroRNAs are small noncoding RNAs that regulate the expression of protein-coding genes. To evaluate the involvement of microRNAs in prostate cancer, we determined genome-wide expression of microRNAs and mRNAs in 60 primary prostate tumors and 16 nontumor prostate tissues. The mRNA analysis revealed that key components of microRNA processing and several microRNA host genes, e.g., MCM7 and C9orf5, were significantly up-regulated in prostate tumors. Consistent with these findings, tumors expressed the miR-106b-25 cluster, which maps to intron 13 of MCM7, and miR-32, which maps to intron 14 of C9orf5, at significantly higher levels than nontumor prostate. The expression levels of other microRNAs, including a number of miR-106b-25 cluster homologues, were also altered in prostate tumors. Additional differences in microRNA abundance were found between organ-confined tumors and those with extraprostatic disease extension. Lastly, we found evidence that some microRNAs are androgen-regulated and that tumor microRNAs influence transcript abundance of protein-coding target genes in the cancerous prostate. In cell culture, E2F1 and p21/WAF1 were identified as targets of miR-106b, Bim of miR-32, and exportin-6 and protein tyrosine kinase 9 of miR-1. In summary, microRNA expression becomes altered with the development and progression of prostate cancer. Some of these microRNAs regulate the expression of cancer-related genes in prostate cancer cells.


Subject(s)
Gene Expression Profiling , Genomics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Aged , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Prostatic Neoplasms/enzymology , Ribonuclease III/metabolism , Up-Regulation
17.
Cancer Cell ; 13(3): 272-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328430

ABSTRACT

Deregulation of E2F1 activity and resistance to TGFbeta are hallmarks of gastric cancer. MicroRNAs (miRNAs) are small noncoding RNAs frequently misregulated in human malignancies. Here we provide evidence that the miR-106b-25 cluster, upregulated in a subset of human gastric tumors, is activated by E2F1 in parallel with its host gene, Mcm7. In turn, miR-106b and miR-93 regulate E2F1 expression, establishing a miRNA-directed negative feedback loop. Furthermore, upregulation of these miRNAs impairs the TGFbeta tumor suppressor pathway, interfering with the expression of CDKN1A (p21(Waf1/Cip1)) and BCL2L11 (Bim). Together, these results suggest that the miR-106b-25 cluster is involved in E2F1 posttranscriptional regulation and may play a key role in the development of TGFbeta resistance in gastric cancer.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Cell Cycle/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Stomach Neoplasms/genetics , Transforming Growth Factor beta/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/genetics , Feedback, Physiological , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Minichromosome Maintenance Complex Component 7 , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Time Factors , Transfection , Up-Regulation
18.
Endocr Relat Cancer ; 14(3): 791-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17914108

ABSTRACT

We have recently reported that MicroRNAs (miR)-221 and miR-222 were up-regulated in human thyroid papillary carcinomas in comparison with the normal thyroid tissue. Bioinformatic analysis proposed the p27(Kip1) protein, a key regulator of cell cycle, as a candidate target for the miR-221/222 cluster. Here, we report that the enforced expression of miR-221 and miR-222 was able to reduce p27(Kip1) protein levels in thyroid carcinoma and HeLa cells in the absence of significant changes in specific p27(Kip1) mRNA levels. This effect is direct as miR-221 and miR-222 negatively regulate the expression of the 3'-untranslated region-based reporter construct from the p27(Kip1) gene, and is dependent on two target sites in this region. Consistent with these results, an enforced expression of the miR-221 and miR-222 induced the thyroid papillary carcinoma cell line (TPC-1) to progress to the S phase of the cell cycle. It is likely that the negative regulation of p27(Kip1) by miR-221 and miR-222 might also have a role in vivo since we report an inverse correlation between miR-221 and miR-222 up-regulation and down-regulation of the p27(Kip1) protein levels in human thyroid papillary carcinomas. Therefore, the data reported here demonstrate that miR-221 and miR-222 are endogenous regulators of p27(Kip1) protein expression, and thereby, the cell cycle.


Subject(s)
Carcinoma, Papillary/genetics , Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , MicroRNAs/physiology , Thyroid Neoplasms/genetics , Base Sequence , Cyclin-Dependent Kinase Inhibitor p27 , Down-Regulation , HeLa Cells , Humans , Tumor Cells, Cultured
19.
Cancer Res ; 67(18): 8699-707, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875710

ABSTRACT

Epithelial ovarian cancer (EOC) is the sixth most common cancer in women worldwide and, despite advances in detection and therapies, it still represents the most lethal gynecologic malignancy in the industrialized countries. Unfortunately, still relatively little is known about the molecular events that lead to the development of this highly aggressive disease. The relatively recent discovery of microRNAs (miRNA), a class of small noncoding RNAs targeting multiple mRNAs and triggering translation repression and/or RNA degradation, has revealed the existence of a new level of gene expression regulation. Multiple studies involving various types of human cancers proved that miRNAs have a causal role in tumorigenesis. Here we show that, in comparison to normal ovary, miRNAs are aberrantly expressed in human ovarian cancer. The overall miRNA expression could clearly separate normal versus cancer tissues. The most significantly overexpressed miRNAs were miR-200a, miR-141, miR-200c, and miR-200b, whereas miR-199a, miR-140, miR-145, and miR-125b1 were among the most down-modulated miRNAs. We could also identify miRNAs whose expression was correlated with specific ovarian cancer biopathologic features, such as histotype, lymphovascular and organ invasion, and involvement of ovarian surface. Moreover, the levels of miR-21, miR-203, and miR-205, up-modulated in ovarian carcinomas compared with normal tissues, were significantly increased after 5-aza-2'-deoxycytidine demethylating treatment of OVCAR3 cells, suggesting that the DNA hypomethylation could be the mechanism responsible for their overexpression. Our results indicate that miRNAs might play a role in the pathogenesis of human EOC and identify altered miRNA gene methylation as a possible epigenetic mechanism involved in their aberrant expression.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Cell Line, Tumor , DNA Methylation , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Female , Gene Expression Profiling , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
20.
JAMA ; 297(17): 1901-8, 2007 May 02.
Article in English | MEDLINE | ID: mdl-17473300

ABSTRACT

CONTEXT: While global microRNA (miRNA) expression patterns of many embryologic, physiologic, and oncogenic processes have been described, description of the role of miRNAs in ductal adenocarcinoma of the pancreas is lacking. OBJECTIVE: To define the expression pattern of miRNAs in pancreatic cancer and compare it with those of normal pancreas and chronic pancreatitis. DESIGN AND SETTING: Specimens were obtained at a National Cancer Institute-designated comprehensive cancer center from patients with ductal adenocarcinoma of the pancreas (n = 65) or chronic pancreatitis (n = 42) (January 2000-December 2005). All patients underwent curative pancreatectomy; those with pancreatic cancer were chemotherapy-naive. RNA harvested from resected pancreatic cancers and matched benign adjacent pancreatic tissue as well as from chronic pancreatitis specimens was hybridized to miRNA microarrays. MAIN OUTCOME MEASURES: Identification of differentially expressed miRNAs that could differentiate pancreatic cancer from normal pancreas, chronic pancreatitis, or both, as well as a pattern of miRNA expression predictive of long-term (>24 months) survival. Significance of Analysis of Microarrays and Prediction of Analysis of Microarrays were undertaken to identify miRNAs predictive of tissue type and prognosis. P values were calculated by t test, adjusted for multiple testing. Kaplan-Meier survival curves were constructed using mean miRNA expression (high vs low) as threshold and compared by log-rank analysis. RESULTS: Twenty-one miRNAs with increased expression and 4 with decreased expression were identified that correctly differentiated pancreatic cancer from benign pancreatic tissue in 90% of samples by cross validation. Fifteen overexpressed and 8 underexpressed miRNAs differentiated pancreatic cancer from chronic pancreatitis with 93% accuracy. A subgroup of 6 miRNAs was able to distinguish long-term survivors with node-positive disease from those dying within 24 months. Finally, high expression of miR-196a-2 was found to predict poor survival (median, 14.3 months [95% confidence interval, 12.4-16.2] vs 26.5 months [95% confidence interval, 23.4-29.6]; P = .009). CONCLUSIONS: Pancreatic cancer may have a distinct miRNA expression pattern that may differentiate it from normal pancreas and chronic pancreatitis. miRNA expression patterns may be able to distinguish between long- and short-term survivors, but these findings need to be validated in other study populations.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , MicroRNAs/analysis , Pancreas/chemistry , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/genetics , Blotting, Northern , Genetic Markers , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , Pancreas/anatomy & histology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/pathology , Polymerase Chain Reaction , Prognosis , RNA, Neoplasm/analysis , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...