Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(23): 14522-14529, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33034194

ABSTRACT

Histone demethylase LSDl (KDMlA) belongs to the flavin adenine dinucleotide (FAD) dependent family of monoamine oxidases and is vital in regulation of mammalian biology. Dysregulation and overexpression of LSD1 are hallmarks of a number of human diseases, particularly cancers that are characterized as morphologically poorly differentiated. As such, inhibitors of LSD1 have potential to be beneficial as a cancer therapy. The most clinically advanced inhibitors of LSDl are covalent inhibitors derived from tranylcypromine (TCP). Herein, we report the discovery of a novel series of reversible and selective LSDl inhibitors. Exploration of structure-activity relationships (SARs) and optimization of ADME properties resulted in the identification of clinical candidate CC-90011. CC-90011 exhibits potent on-target induction of cellular differentiation in acute myeloid leukemia (AML) and small cell lung cancer (SCLC) cell lines, and antitumor efficacy in patient-derived xenograft (PDX) SCLC models. CC-90011 is currently in phase 2 trials in patients with first line, extensive stage SCLC (ClinicalTrials.gov identifier: NCT03850067).


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Organic Chemicals/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Humans , Organic Chemicals/chemistry , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 29(1): 103-106, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30409536

ABSTRACT

The histone demethylase LSD1 is a key enzyme in the epigenetic regulation of gene transcription. Here we present our efforts to discover small molecule reversible inhibitors of LSD1 as an attractive approach to treat hematologic malignancies and certain solid tumors. Using structure-based drug design, we designed and synthesized a novel series of heteroaromatic imidazole inhibitors that demonstrate potent inhibition of the demethylase activity and low nanomolar cell-based activity. This novel LSD1 inhibitor series was further optimized by attenuating the hERG inhibition and improving oral bioavailability.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Imidazoles/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
3.
Cell ; 138(3): 592-603, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19665978

ABSTRACT

Human breast tumors contain a breast cancer stem cell (BCSC) population with properties reminiscent of normal stem cells. We found 37 microRNAs that were differentially expressed between human BCSCs and nontumorigenic cancer cells. Three clusters, miR-200c-141, miR-200b-200a-429, and miR-183-96-182 were downregulated in human BCSCs, normal human and murine mammary stem/progenitor cells, and embryonal carcinoma cells. Expression of BMI1, a known regulator of stem cell self-renewal, was modulated by miR-200c. miR-200c inhibited the clonal expansion of breast cancer cells and suppressed the growth of embryonal carcinoma cells in vitro. Most importantly, miR-200c strongly suppressed the ability of normal mammary stem cells to form mammary ducts and tumor formation driven by human BCSCs in vivo. The coordinated downregulation of three microRNA clusters and the similar functional regulation of clonal expansion by miR-200c provide a molecular link that connects BCSCs with normal stem cells.


Subject(s)
Breast Neoplasms/genetics , Breast/cytology , Gene Expression Profiling , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Stem Cells/metabolism , Cell Line , Cell Line, Tumor , Down-Regulation , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Semin Radiat Oncol ; 19(2): 78-86, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19249645

ABSTRACT

A growing body of evidence indicates that subpopulations of cancer stem cells (CSCs) drive and maintain many types of human malignancies. These findings have important implications for the development and evaluation of oncologic therapies and present opportunities for potential gains in patient outcome. The existence of CSCs mandates careful analysis and comparison of normal tissue stem cells and CSCs to identify differences between the two cell types. The development of CSC-targeted treatments will face a number of potential hurdles, including normal stem cell toxicity and the acquisition of treatment resistance, which must be considered in order to maximize the chance that such therapies will be successful.


Subject(s)
Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Humans
5.
Nature ; 458(7239): 780-3, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19194462

ABSTRACT

The metabolism of oxygen, although central to life, produces reactive oxygen species (ROS) that have been implicated in processes as diverse as cancer, cardiovascular disease and ageing. It has recently been shown that central nervous system stem cells and haematopoietic stem cells and early progenitors contain lower levels of ROS than their more mature progeny, and that these differences are critical for maintaining stem cell function. We proposed that epithelial tissue stem cells and their cancer stem cell (CSC) counterparts may also share this property. Here we show that normal mammary epithelial stem cells contain lower concentrations of ROS than their more mature progeny cells. Notably, subsets of CSCs in some human and murine breast tumours contain lower ROS levels than corresponding non-tumorigenic cells (NTCs). Consistent with ROS being critical mediators of ionizing-radiation-induced cell killing, CSCs in these tumours develop less DNA damage and are preferentially spared after irradiation compared to NTCs. Lower ROS levels in CSCs are associated with increased expression of free radical scavenging systems. Pharmacological depletion of ROS scavengers in CSCs markedly decreases their clonogenicity and results in radiosensitization. These results indicate that, similar to normal tissue stem cells, subsets of CSCs in some tumours contain lower ROS levels and enhanced ROS defences compared to their non-tumorigenic progeny, which may contribute to tumour radioresistance.


Subject(s)
Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Radiation Tolerance/physiology , Reactive Oxygen Species/metabolism , Animals , Breast Neoplasms/physiopathology , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , Female , Gene Expression , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Mice, Inbred C57BL
6.
Curr Opin Genet Dev ; 18(1): 48-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18356041

ABSTRACT

The theory of cancer stem cells states that a subset of cancer cells within a tumor has the ability to self-renew and differentiate. Only those cells within a tumor that have these two properties are called cancer stem cells. This concept was first demonstrated in the study of leukemia where only cells with specific surface antigen profiles were able to cause leukemia when engrafted into immunodeficient mice. In recent years solid tumors were studied utilizing similar techniques in mice. Human tumors where evidence of cancer stem cells has been published include tumors of the breast, brain, pancreas, head and neck, and colon. If this difference in tumorigenicity of cancer cells also occurs in patients, then the ability to enrich for cancer stem cells lays an important groundwork for future studies where mechanisms involved in cancer stem cells can now be investigated.


Subject(s)
Neoplasms/etiology , Neoplastic Stem Cells/physiology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Female , Hematopoietic Stem Cells/metabolism , Humans , Leukemia/metabolism , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Animal/etiology , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism
7.
Stem Cells ; 26(2): 364-71, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17975224

ABSTRACT

In human breast cancers, a phenotypically distinct minority population of tumorigenic (TG) cancer cells (sometimes referred to as cancer stem cells) drives tumor growth when transplanted into immunodeficient mice. Our objective was to identify a mouse model of breast cancer stem cells that could have relevance to the study of human breast cancer. To do so, we used breast tumors of the mouse mammary tumor virus (MMTV)-Wnt-1 mice. MMTV-Wnt-1 breast tumors were harvested, dissociated into single-cell suspensions, and sorted by flow cytometry on Thy1, CD24, and CD45. Sorted cells were then injected into recipient background FVB/NJ female syngeneic mice. In six of seven tumors examined, Thy1+CD24+ cancer cells, which constituted approximately 1%-4% of tumor cells, were highly enriched for cells capable of regenerating new tumors compared with cells of the tumor that did not fit this profile ("not-Thy1+CD24+"). Resultant tumors had a phenotypic diversity similar to that of the original tumor and behaved in a similar manner when passaged. Microarray analysis comparing Thy1+CD24+ tumor cells to not-Thy1+CD24+ cells identified a list of differentially expressed genes. Orthologs of these differentially expressed genes predicted survival of human breast cancer patients from two different study groups. These studies suggest that there is a cancer stem cell compartment in the MMTV-Wnt-1 murine breast tumor and that there is a clinical utility of this model for the study of cancer stem cells.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Cell Separation/methods , Female , Flow Cytometry , Humans , Mammary Tumor Virus, Mouse/pathogenicity , Mice , Mice, Transgenic , Neoplasm Transplantation , Neoplastic Stem Cells/classification , Oligonucleotide Array Sequence Analysis , Retroviridae Infections/genetics , Retroviridae Infections/pathology , Transplantation, Isogeneic , Tumor Virus Infections/genetics , Tumor Virus Infections/pathology , Wnt1 Protein/genetics
8.
Proc Natl Acad Sci U S A ; 104(24): 10158-63, 2007 Jun 12.
Article in English | MEDLINE | ID: mdl-17548814

ABSTRACT

Recent observations indicate that, in several types of human cancer, only a phenotypic subset of cancer cells within each tumor is capable of initiating tumor growth. This functional subset of cancer cells is operationally defined as the "cancer stem cell" (CSC) subset. Here we developed a CSC model for the study of human colorectal cancer (CRC). Solid CRC tissues, either primary tissues collected from surgical specimens or xenografts established in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, were disaggregated into single-cell suspensions and analyzed by flow cytometry. Surface markers that displayed intratumor heterogeneous expression among epithelial cancer cells were selected for cell sorting and tumorigenicity experiments. Individual phenotypic cancer cell subsets were purified, and their tumor-initiating properties were investigated by injection in NOD/SCID mice. Our observations indicate that, in six of six human CRC tested, the ability to engraft in vivo in immunodeficient mice was restricted to a minority subpopulation of epithelial cell adhesion molecule (EpCAM)(high)/CD44+ epithelial cells. Tumors originated from EpCAM(high)/CD44+ cells maintained a differentiated phenotype and reproduced the full morphologic and phenotypic heterogeneity of their parental lesions. Analysis of the surface molecule repertoire of EpCAM(high)/CD44+ cells led to the identification of CD166 as an additional differentially expressed marker, useful for CSC isolation in three of three CRC tested. These results validate the stem cell working model in human CRC and provide a highly robust surface marker profile for CRC stem cell isolation.


Subject(s)
Antigens, Neoplasm , Biomarkers, Tumor/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Stem Cells/immunology , Activated-Leukocyte Cell Adhesion Molecule/immunology , Animals , Antigens, Neoplasm/immunology , Cell Separation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/surgery , Epithelial Cells/immunology , Flow Cytometry , Humans , Hyaluronan Receptors/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phenotype , Transplantation, Heterologous , Tumor Stem Cell Assay
9.
Annu Rev Med ; 58: 267-84, 2007.
Article in English | MEDLINE | ID: mdl-17002552

ABSTRACT

Although monoclonal in origin, most tumors appear to contain a heterogeneous population of cancer cells. This observation is traditionally explained by postulating variations in tumor microenvironment and coexistence of multiple genetic subclones, created by progressive and divergent accumulation of independent somatic mutations. An additional explanation, however, envisages human tumors not as mere monoclonal expansions of transformed cells, but rather as complex tridimensional tissues where cancer cells become functionally heterogeneous as a result of differentiation. According to this second scenario, tumors act as caricatures of their corresponding normal tissues and are sustained in their growth by a pathological counterpart of normal adult stem cells, cancer stem cells. This model, first developed in human myeloid leukemias, is today being extended to solid tumors, such as breast and brain cancer. We review the biological basis and the therapeutic implications of the stem cell model of cancer.


Subject(s)
Models, Biological , Neoplasms/etiology , Neoplasms/therapy , Neoplastic Stem Cells/physiology , Humans , Neoplasms/pathology
10.
J Neuroophthalmol ; 24(2): 119-24, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15179064

ABSTRACT

A 2-year-old boy with blindness as an isolated symptom was found to have no light perception binocularly because of compression of both optic nerves by a neuroblastoma infiltrating the walls of the optic canals and medial sphenoid bone. Imaging disclosed a primary tumor near the kidney and multiple osseous metastases. Although neuroblastoma commonly causes blindness by metastasis to the orbit, it rarely causes bilateral blindness from intracranial compression of the optic nerves. This is the first report of bilateral blindness as the presenting feature.


Subject(s)
Blindness/etiology , Nerve Compression Syndromes/etiology , Neuroblastoma/complications , Neuroblastoma/secondary , Optic Nerve Diseases/etiology , Skull Neoplasms/complications , Skull Neoplasms/secondary , Sphenoid Bone , Blindness/physiopathology , Child, Preschool , Combined Modality Therapy , Humans , Kidney Neoplasms/pathology , Magnetic Resonance Imaging , Male , Nerve Compression Syndromes/complications , Neuroblastoma/diagnosis , Neuroblastoma/surgery , Neuroblastoma/therapy , Optic Nerve Diseases/complications , Remission Induction , Skull Neoplasms/diagnosis , Skull Neoplasms/therapy , Vision, Binocular , Visual Acuity
SELECTION OF CITATIONS
SEARCH DETAIL
...