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










Publication year range
1.
Clin Cancer Res ; 29(2): 488-500, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36239995

ABSTRACT

PURPOSE: Therapy resistance and fatal disease progression in glioblastoma are thought to result from the dynamics of intra-tumor heterogeneity. This study aimed at identifying and molecularly targeting tumor cells that can survive, adapt, and subclonally expand under primary therapy. EXPERIMENTAL DESIGN: To identify candidate markers and to experimentally access dynamics of subclonal progression in glioblastoma, we established a discovery cohort of paired vital cell samples obtained before and after primary therapy. We further used two independent validation cohorts of paired clinical tissues to test our findings. Follow-up preclinical treatment strategies were evaluated in patient-derived xenografts. RESULTS: We describe, in clinical samples, an archetype of rare ALDH1A1+ tumor cells that enrich and acquire AKT-mediated drug resistance in response to standard-of-care temozolomide (TMZ). Importantly, we observe that drug resistance of ALDH1A1+ cells is not intrinsic, but rather an adaptive mechanism emerging exclusively after TMZ treatment. In patient cells and xenograft models of disease, we recapitulate the enrichment of ALDH1A1+ cells under the influence of TMZ. We demonstrate that their subclonal progression is AKT-driven and can be interfered with by well-timed sequential rather than simultaneous antitumor combination strategy. CONCLUSIONS: Drug-resistant ALDH1A1+/pAKT+ subclones accumulate in patient tissues upon adaptation to TMZ therapy. These subclones may therefore represent a dynamic target in glioblastoma. Our study proposes the combination of TMZ and AKT inhibitors in a sequential treatment schedule as a rationale for future clinical investigation.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Proto-Oncogene Proteins c-akt , Drug Resistance, Neoplasm/genetics , Temozolomide , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Xenograft Model Antitumor Assays , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use
2.
Clin Cancer Res ; 23(2): 562-574, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27521447

ABSTRACT

PURPOSE: Investigation of clonal heterogeneity may be key to understanding mechanisms of therapeutic failure in human cancer. However, little is known on the consequences of therapeutic intervention on the clonal composition of solid tumors. EXPERIMENTAL DESIGN: Here, we used 33 single cell-derived subclones generated from five clinical glioblastoma specimens for exploring intra- and interindividual spectra of drug resistance profiles in vitro In a personalized setting, we explored whether differences in pharmacologic sensitivity among subclones could be employed to predict drug-dependent changes to the clonal composition of tumors. RESULTS: Subclones from individual tumors exhibited a remarkable heterogeneity of drug resistance to a library of potential antiglioblastoma compounds. A more comprehensive intratumoral analysis revealed that stable genetic and phenotypic characteristics of coexisting subclones could be correlated with distinct drug sensitivity profiles. The data obtained from differential drug response analysis could be employed to predict clonal population shifts within the naïve parental tumor in vitro and in orthotopic xenografts. Furthermore, the value of pharmacologic profiles could be shown for establishing rational strategies for individualized secondary lines of treatment. CONCLUSIONS: Our data provide a previously unrecognized strategy for revealing functional consequences of intratumor heterogeneity by enabling predictive modeling of treatment-related subclone dynamics in human glioblastoma. Clin Cancer Res; 23(2); 562-74. ©2016 AACR.


Subject(s)
Drug Combinations , Drug Resistance, Neoplasm/genetics , Genetic Heterogeneity , Glioblastoma/drug therapy , Animals , Clonal Evolution/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Xenograft Model Antitumor Assays
3.
Nat Immunol ; 16(7): 766-774, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25985233

ABSTRACT

Childhood acute lymphoblastic leukemia (ALL) can often be traced to a pre-leukemic clone carrying a prenatal genetic lesion. Postnatally acquired mutations then drive clonal evolution toward overt leukemia. The enzymes RAG1-RAG2 and AID, which diversify immunoglobulin-encoding genes, are strictly segregated in developing cells during B lymphopoiesis and peripheral mature B cells, respectively. Here we identified small pre-BII cells as a natural subset with increased genetic vulnerability owing to concurrent activation of these enzymes. Consistent with epidemiological findings on childhood ALL etiology, susceptibility to genetic lesions during B lymphopoiesis at the transition from the large pre-BII cell stage to the small pre-BII cell stage was exacerbated by abnormal cytokine signaling and repetitive inflammatory stimuli. We demonstrated that AID and RAG1-RAG2 drove leukemic clonal evolution with repeated exposure to inflammatory stimuli, paralleling chronic infections in childhood.


Subject(s)
B-Lymphocytes/immunology , Clonal Evolution/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cells, B-Lymphoid/immunology , Adolescent , Animals , Antibody Diversity/genetics , Antibody Diversity/immunology , B-Lymphocytes/metabolism , Child , Child, Preschool , Clonal Evolution/genetics , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Female , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Immunoblotting , Infant , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Microscopy, Fluorescence , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
4.
Clin Cancer Res ; 19(15): 4124-36, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23908450

ABSTRACT

PURPOSE: Glioblastoma is a highly malignant, invariably fatal brain tumor for which effective pharmacotherapy remains an unmet medical need. EXPERIMENTAL DESIGN: Screening of a compound library of 160 synthetic and natural toxic substances identified the antihelmintic niclosamide as a previously unrecognized candidate for clinical development. Considering the cellular and interindividual heterogeneity of glioblastoma, a portfolio of short-term expanded primary human glioblastoma cells (pGBM; n = 21), common glioma lines (n = 5), and noncancer human control cells (n = 3) was applied as a discovery platform and for preclinical validation. Pharmacodynamic analysis, study of cell-cycle progression, apoptosis, cell migration, proliferation, and on the frequency of multipotent/self-renewing pGBM cells were conducted in vitro, and orthotopic xenotransplantation was used to confirm anticancer effects in vivo. RESULTS: Niclosamide led to cytostatic, cytotoxic, and antimigratory effects, strongly reduced the frequencies of multipotent/self-renewing cells in vitro, and after exposure significantly diminished the pGBMs' malignant potential in vivo. Mechanism of action analysis revealed that niclosamide simultaneously inhibited intracellular WNT/CTNNB1-, NOTCH-, mTOR-, and NF-κB signaling cascades. Furthermore, combinatorial drug testing established that a heterozygous deletion of the NFKBIA locus in glioblastoma samples could serve as a genomic biomarker for predicting a synergistic activity of niclosamide with temozolomide, the current standard in glioblastoma therapy. CONCLUSIONS: Together, our data advocate the use of pGBMs for exploration of compound libraries to reveal unexpected leads, for example, niclosamide that might be suited for further development toward personalized clinical application.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioblastoma/genetics , Niclosamide/administration & dosage , Animals , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/pathology , Humans , Mice , NF-kappa B/metabolism , Receptors, Notch/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
5.
Stem Cells ; 31(6): 1064-74, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23390110

ABSTRACT

Cellular heterogeneity, for example, the intratumoral coexistence of cancer cells with and without stem cell characteristics, represents a potential root of therapeutic resistance and a significant challenge for modern drug development in glioblastoma (GBM). We propose here that activation of the innate immune system by stimulation of innate immune receptors involved in antiviral and antitumor responses can similarly target different malignant populations of glioma cells. We used short-term expanded patient-specific primary human GBM cells to study the stimulation of the cytosolic nucleic acid receptors melanoma differentiation-associated gene 5 (MDA5) and retinoic acid-inducible gene I (RIG-I). Specifically, we analyzed cells from the tumor core versus "residual GBM cells" derived from the tumor resection margin as well as stem cell-enriched primary cultures versus specimens without stem cell properties. A portfolio of human, nontumor neural cells was used as a control for these studies. The expression of RIG-I and MDA5 could be induced in all of these cells. Receptor stimulation with their respective ligands, p(I:C) and 3pRNA, led to in vitro evidence for an effective activation of the innate immune system. Most intriguingly, all investigated cancer cell populations additionally responded with a pronounced induction of apoptotic signaling cascades revealing a second, direct mechanism of antitumor activity. By contrast, p(I:C) and 3pRNA induced only little toxicity in human nonmalignant neural cells. Granted that the challenge of effective central nervous system (CNS) delivery can be overcome, targeting of RIG-I and MDA5 could thus become a quintessential strategy to encounter heterogeneous cancers in the sophisticated environments of the brain.


Subject(s)
Antineoplastic Agents/pharmacology , Cytosol/immunology , DEAD-box RNA Helicases/immunology , Glioblastoma/drug therapy , Glioblastoma/immunology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon-Induced Helicase, IFIH1 , Ligands , Receptors, Immunologic , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/immunology , Stem Cells/metabolism
6.
Mol Cancer Res ; 10(1): 143-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22258704

ABSTRACT

Musashi1 (Msi1) is an evolutionarily conserved RNA-binding protein (RBP) that has profound implications in cellular processes such as stem cell maintenance, nervous system development, and tumorigenesis. Msi1 is highly expressed in many cancers, including glioblastoma, whereas in normal tissues, its expression is restricted to stem cells. Unfortunately, the factors that modulate Msi1 expression and trigger high levels in tumors are largely unknown. The Msi1 mRNA has a long 3' untranslated region (UTR) containing several AU- and U-rich sequences. This type of sequence motif is often targeted by HuR, another important RBP known to be highly expressed in tumor tissue such as glioblastoma and to regulate a variety of cancer-related genes. In this report, we show an interaction between HuR and the Msi1 3'-UTR, resulting in a positive regulation of Msi1 expression. We show that HuR increased MSI1 mRNA stability and promoted its translation. We also present evidence that expression of HuR and Msi1 correlate positively in clinical glioblastoma samples. Finally, we show that inhibition of cell proliferation, increased apoptosis, and changes in cell-cycle profile as a result of silencing HuR are partially rescued when Msi1 is ectopically expressed. In summary, our results suggest that HuR is an important regulator of Msi1 in glioblastoma and that this regulation has important biological consequences during gliomagenesis.


Subject(s)
Brain Neoplasms/genetics , ELAV Proteins/physiology , Glioblastoma/genetics , Nerve Tissue Proteins/genetics , Protein Biosynthesis/genetics , RNA Stability/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , ELAV Proteins/antagonists & inhibitors , ELAV Proteins/genetics , ELAV Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , HeLa Cells , Humans , Nerve Tissue Proteins/metabolism , Oncogenes/genetics , Protein Biosynthesis/drug effects , RNA Stability/drug effects , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 108(24): 9951-6, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21628563

ABSTRACT

The tyrosine kinase c-Met promotes the formation and malignant progression of multiple cancers. It is well known that c-Met hyperactivation increases tumorigenicity and tumor cell resistance to DNA damaging agents, properties associated with tumor-initiating stem cells. However, a link between c-Met signaling and the formation and/or maintenance of neoplastic stem cells has not been previously identified. Here, we show that c-Met is activated and functional in glioblastoma (GBM) neurospheres enriched for glioblastoma tumor-initiating stem cells and that c-Met expression/function correlates with stem cell marker expression and the neoplastic stem cell phenotype in glioblastoma neurospheres and clinical glioblastoma specimens. c-Met activation was found to induce the expression of reprogramming transcription factors (RFs) known to support embryonic stem cells and induce differentiated cells to form pluripotent stem (iPS) cells, and c-Met activation counteracted the effects of forced differentiation in glioblastoma neurospheres. Expression of the reprogramming transcription factor Nanog by glioblastoma cells is shown to mediate the ability of c-Met to induce the stem cell characteristics of neurosphere formation and neurosphere cell self-renewal. These findings show that c-Met enhances the population of glioblastoma stem cells (GBM SCs) via a mechanism requiring Nanog and potentially other c-Met-responsive reprogramming transcription factors.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , AC133 Antigen , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cellular Reprogramming , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Glycoproteins/genetics , Glycoproteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoblotting , Indoles/pharmacology , Mice , Mice, SCID , Nanog Homeobox Protein , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Peptides/genetics , Peptides/metabolism , Phenotype , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sulfonamides/pharmacology , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Ann Neurol ; 68(2): 264-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20695020

ABSTRACT

Residual tumor cells remain beyond the margins of every glioblastoma (GBM) resection. Their resistance to postsurgical therapy is considered a major driving force of mortality, but their biology remains largely uncharacterized. In this study, residual tumor cells were derived via experimental biopsy of the resection margin after standard neurosurgery for direct comparison with samples from the routinely resected tumor tissue. In vitro analysis of proliferation, invasion, stem cell qualities, GBM-typical antigens, genotypes, and in vitro drug and irradiation challenge studies revealed these cells as unique entities. Our findings suggest a need for characterization of residual tumor cells to optimize diagnosis and treatment of GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Adult , Aged , Antigens, Neoplasm/metabolism , Brain Neoplasms/drug therapy , Cell Culture Techniques , Cell Proliferation , Cell Separation , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured
9.
J Exp Med ; 206(8): 1739-53, 2009 Aug 03.
Article in English | MEDLINE | ID: mdl-19620627

ABSTRACT

B cell lineage acute lymphoblastic leukemia (ALL) arises in virtually all cases from B cell precursors that are arrested at pre-B cell receptor-dependent stages. The Philadelphia chromosome-positive (Ph(+)) subtype of ALL accounts for 25-30% of cases of adult ALL, has the most unfavorable clinical outcome among all ALL subtypes and is defined by the oncogenic BCR-ABL1 kinase and deletions of the IKAROS gene in >80% of cases. Here, we demonstrate that the pre-B cell receptor functions as a tumor suppressor upstream of IKAROS through induction of cell cycle arrest in Ph(+) ALL cells. Pre-B cell receptor-mediated cell cycle arrest in Ph(+) ALL cells critically depends on IKAROS function, and is reversed by coexpression of the dominant-negative IKAROS splice variant IK6. IKAROS also promotes tumor suppression through cooperation with downstream molecules of the pre-B cell receptor signaling pathway, even if expression of the pre-B cell receptor itself is compromised. In this case, IKAROS redirects oncogenic BCR-ABL1 tyrosine kinase signaling from SRC kinase-activation to SLP65, which functions as a critical tumor suppressor downstream of the pre-B cell receptor. These findings provide a rationale for the surprisingly high frequency of IKAROS deletions in Ph(+) ALL and identify IKAROS-mediated cell cycle exit as the endpoint of an emerging pathway of pre-B cell receptor-mediated tumor suppression.


Subject(s)
Ikaros Transcription Factor/physiology , Leukemia, Prolymphocytic, B-Cell/genetics , Philadelphia Chromosome , Pre-B Cell Receptors/physiology , Adaptor Proteins, Signal Transducing/physiology , Adult , Animals , Cell Cycle , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Down-Regulation , Gene Deletion , Genes, abl , Humans , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Leukemia, Prolymphocytic, B-Cell/pathology , Leukemia, Prolymphocytic, B-Cell/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Pre-B Cell Receptors/deficiency , Pre-B Cell Receptors/genetics , Signal Transduction
10.
Mol Cancer ; 6: 67, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17958915

ABSTRACT

BACKGROUND: Ph-positive leukemias are caused by the aberrant fusion of the BCR and ABL genes. Nilotinib is a selective Bcr/Abl tyrosine kinase inhibitor related to imatinib, which is widely used to treat chronic myelogenous leukemia. Because Ph-positive acute lymphoblastic leukemia only responds transiently to imatinib therapy, we have used mouse models to test the efficacy of nilotinib against lymphoblastic leukemia caused by the P190 form of Bcr/Abl. RESULTS: After transplant of 10,000 highly malignant leukemic cells into compatible recipients, untreated mice succumbed to leukemia within 21 days, whereas mice treated with 75 mg/kg nilotinib survived significantly longer. We examined cells from mice that developed leukemia while under treatment for Bcr/Abl kinase domain point mutations but these were not detected. In addition, culture of such cells ex vivo showed that they were as sensitive as the parental cell line to nilotinib but that the presence of stromal support allowed resistant cells to grow out. Nilotinib also exhibited impressive anti-leukemia activity in P190 Bcr/Abl transgenic mice that had developed overt leukemia/lymphoma masses and that otherwise would have been expected to die within 7 days. Visible lymphoma masses disappeared within six days of treatment and leukemic cell numbers in peripheral blood were significantly reduced. Treated mice survived more than 30 days. CONCLUSION: These results show that nilotinib has very impressive anti-leukemia activity but that lymphoblastic leukemia cells can become unresponsive to it both in vitro and in vivo through mechanisms that appear to be Bcr/Abl independent.


Subject(s)
Antineoplastic Agents/therapeutic use , Fusion Proteins, bcr-abl , Leukemia, Experimental/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Pyrimidines/therapeutic use , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Janus Kinase 2/drug effects , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stromal Cells/drug effects , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...