426 results on '"Tumor initiating cells"'
Search Results
52. OS06.3A LSD1-directed therapy curtails glioblastoma tumorigenicity by limiting the adaptation of tumor initiating cells to stressful environments
- Author
-
B Costanza, Giuliana Pelicci, E Ceccacci, D Corà, Daniela Osti, S Faletti, G Marotta, and Cristina Richichi
- Subjects
Cancer Research ,Oncology ,medicine ,Cancer research ,Oral Presentations ,Neurology (clinical) ,Limiting ,Biology ,Adaptation ,medicine.disease ,nervous system diseases ,Glioblastoma ,Tumor Initiating Cells - Abstract
BACKGROUND Glioblastoma (GBM) is a fatal tumor whose aggressiveness, heterogeneity, therapy resistance and poor blood-brain-barrier penetration hinder the amelioration of the standard-of-care. Included in the GBM mass are the tumor initiating cells (TICs), representing the driver of GBM growth and relapse in virtue of their stem-like traits and therapy-resistance. Being constantly exposed to environmental stress, including nutrients deficiency, hypoxia and therapeutic insults, all GBM cells -and TICs in particular- have to be highly adaptive in order to survive. Thus, their ability to cope with stress could be targeted to curtail TICs maintenance and the whole GBM aggressiveness. The key of TICs adaptation relies, among the others, on their epigenetic plasticity, hence encouraging epigenetic drugs testing. MATERIAL AND METHODS By exploiting patient-derived GBM TICs and orthotopic xenograft models, we tested the antitumorigenic features of a novel, selective, orally bioavailable and brain-penetrant Lysine-specific histone demethylase 1 inhibitor (LSD1i). We confirmed the specificity of its effects by LSD1 genetic targeting. A combination of RNA-seq, Chromatin Immunoprecipitation(ChIP)-seq, Mass Spectrometry and reverse genetic experiments unraveled LSD1 molecular players in GBM TICs. RESULTS We identified LSD1 as a druggable target in human GBM: LSD1i treatment, mirrored by LSD1 genetic targeting, impairs growth, viability, stem-like traits and in vivo tumorigenicity of GBM TICs. Mechanistically, LSD1 is crucial for the expression of the activating transcription factor 4 (ATF4), which coordinates the integrated stress response (ISR) to manage stressful stimuli as nutrient deprivation and endoplasmic reticulum stress. By mimicking these stress cues in vitro, we found that LSD1i triggers a delayed but unabated ATF4 translation which provokes an over-lasting ISR, eventually culminating in GBM TICs apoptosis. Lastly, LSD1 demethylase activity is dispensable for ATF4 induction. Rather, LSD1i exerts its anti-tumorigenic potential by interfering with LSD1 scaffolding function in GBM TICs. CONCLUSION LSD1-directed therapy is likely a promising strategy to hinder GBM. By sensitizing GBM TICs to stress, LSD1i endangers the GBM TICs pool. The effectiveness of LSD1i administration in different patient-derived GBM TICs and xenografts, regardless of their molecular profile, places a strong rationale toward the clinical translation of this approach for GBM management. FUNDING Italian association for Cancer Research (AIRC) and Italian Ministry of Health
- Published
- 2021
53. Targeting tumor-initiating cells and compensatory YAP pathway to overcome sorafenib resistance in hepatocellular carcinoma
- Author
-
MA Wörns, Peter R. Galle, Arndt Weinmann, Jens U. Marquardt, F Keggenhoff, S Pereira, Snorri S. Thorgeirsson, Stefanie Heilmann-Heimbach, Carolin Czauderna, D Castven, J Schmitt, Tobias Keck, Kai Breuhahn, D Becker, J Hajduk, V Shah, and Harald Binder
- Subjects
business.industry ,Hepatocellular carcinoma ,Cancer research ,Medicine ,business ,medicine.disease ,Tumor Initiating Cells ,Sorafenib resistance - Published
- 2021
- Full Text
- View/download PDF
54. Yin and Yang of Pluripotency: Results of Analysis of Genes Overexpressed In Tumor-Initiating Cells of Krebs-2 Ascites Carcinoma
- Author
-
Ekaterina A. Potter, O.V. Efremova, N. A. Kolchanov, A. S. Proskurina, S. S. Bogachev, Yaroslav R. Efremov, and E.V. Dolgova
- Subjects
0301 basic medicine ,Applied Mathematics ,Biomedical Engineering ,Biology ,medicine.disease ,Tumor Initiating Cells ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Ascites ,medicine ,Carcinoma ,Cancer research ,medicine.symptom ,Gene - Abstract
Functional analysis of 167 genes overexpressed in Krebs-2 tumor initiating cells was performed. Genes were assigned to the three functional groups that determine the malignant phenotype of cancer cells. These groups represent the following features of tumor cells: proliferative self-sufficiency, invasive growth and metastasis, and multiple drug resistance. Malignancy of cancer stem cells was found to be provided by the same genes that provide the stemness of normal pluri-/multipotent stem cells. These results suggest that the malignancy is just the ability to maintain the stem cell specific genes expression profile, and, as a consequence, the stemness itself regardless of the controlling effect of stem niches.
- Published
- 2019
- Full Text
- View/download PDF
55. A new spontaneously transformed syngeneic model of high-grade serous ovarian cancer with a tumor-initiating cell population.
- Author
-
Curtis W. McCloskey, Reuben L. Goldberg, Lauren E. Carter, Lisa F. Gamwell, Ensaf M. Al-Hujaily, Olga eCollins, Elizabeth A. Macdonald, Kenneth eGarson, Manijeh eDaneshmand, Euridice eCarmona, and Barbara C. Vanderhyden
- Subjects
stem cell ,Cancer stem cell ,ovarian cancer ,tumor initiating cells ,ovarian surface epithelium ,high grade serous cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Improving screening and treatment options for patients with epithelial ovarian cancer has been a major challenge in cancer research. Development of novel diagnostic and therapeutic approaches, particularly for the most common subtype, high-grade serous ovarian cancer (HGSC), has been hampered by controversies over the origin of the disease and a lack of spontaneous HGSC models to resolve this controversy. Over long-term culture in our laboratory, an ovarian surface epithelial (OSE) cell line spontaneously transformed (STOSE). The objective of this study was to determine if the STOSE cell line is a good model of HGSC. STOSE cells grow faster than early passage parental M0505 cells with a doubling time of 13h and 48h, respectively. STOSE cells form colonies in soft agar, an activity for which M0505 cells have negligible capacity. Microarray analysis identified 1755 downregulated genes and 1203 upregulated genes in STOSE compared to M0505 cells, many associated with aberrant Wnt/β-Catenin and Nf-κB signaling. Upregulation of Ccnd1 and loss of Cdkn2a in STOSE tumors is consistent with changes identified in human ovarian cancers by The Cancer Genome Atlas. Intraperitoneal injection of STOSE cells into SCID and syngeneic FVB/N mice produced pan-CK+, WT1+, inhibin- and PAX8+ tumors, a histotype resembling human HGSC. Based on evidence that a SCA1+ stem cell-like population exists in M0505 cells, we examined a subpopulation of SCA1+ cells that is present in STOSE cells. Compared to SCA1- cells, SCA1+ STOSE cells have increased colony-forming capacity and form palpable tumors 8 days faster after intrabursal injection into FVB/N mice. This study has identified the STOSE cells as the first spontaneous murine model of HGSC and provides evidence for the OSE as a possible origin of HGSC. Furthermore, this model provides a novel opportunity to study how normal stem-like OSE cells may transform into tumor-initiating cells.
- Published
- 2014
- Full Text
- View/download PDF
56. Identification of cis-HOX-HOXC10 axis as a therapeutic target for colorectal tumor-initiating cells without APC mutations
- Author
-
Lulu Zhang, Pingping Zhu, Zusen Fan, Kaili Wang, Zhiwei Wang, Guangtan Zhang, Zhenzhen Chen, Benyu Liu, and Jiayi Wu
- Subjects
Male ,Untranslated region ,Carcinogenesis ,Adenomatous polyposis coli ,Colorectal cancer ,QH301-705.5 ,RNA Stability ,Nonsense mutation ,colorectal cancer ,medicine.disease_cause ,self-renewal ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,metastasis ,Molecular Targeted Therapy ,RNA, Messenger ,Cell Self Renewal ,Biology (General) ,Wnt Signaling Pathway ,Aged ,Pyrans ,Aged, 80 and over ,Homeodomain Proteins ,Mice, Knockout ,Regulation of gene expression ,biology ,tumor initiating cells ,business.industry ,Wnt signaling pathway ,RNA-Binding Proteins ,RNA, Circular ,medicine.disease ,HOXC10 ,Frizzled Receptors ,Gene Expression Regulation, Neoplastic ,Adenomatous Polyposis Coli ,Mutation ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Female ,cis-HOX ,Colorectal Neoplasms ,business - Abstract
Summary: Colorectal cancer (CRC) is one of the most common cancers worldwide, in which adenomatous polyposis coli (APC) mutations are frequently and uniquely observed. Here we find that cis-HOX (circular RNA stabilizing HOXC10) is robustly expressed in colorectal tumor-initiating cells (TICs). cis-HOX knockout decreases colorectal TIC numbers and impairs the self-renewal, tumorigenesis, and metastatic capacities of TICs, whereas cis-HOX overexpression drives colorectal TIC self-renewal and metastasis. Mechanistically, cis-HOX binds to HOXC10 mRNA to attenuate its decay through blocking the K-homology splicing regulatory protein (KSRP)-binding sequence of HOXC10 3′ UTR. HOXC10 is highly expressed in colorectal tumors and TICs and triggers Wnt/β-catenin activation by activating FZD3 expression. HOXC10 inhibitor salinomycin exerts efficient therapeutic effects in APC-wild-type colorectal tumors, but not in tumors with APC nonsense mutations. Therefore, the cis-HOX-HOXC10 pathway drives colorectal tumorigenesis, stemness, and metastasis and serves as a potential therapeutic target for APC-wild-type colorectal tumors.
- Published
- 2021
57. Influence of CD133+ expression on patients' survival and resistance of CD133+ cells to anti-tumor reagents in gastric cancer.
- Author
-
Chen, De-Hu, Lu, Rui-Qi, Ni, Xiao-Chun, Wu, Ju-Gang, Wang, Shou-Lian, Jiang, Bo-Jian, and Yu, Ji-Wei
- Subjects
ANTINEOPLASTIC agents ,STOMACH cancer ,CARCINOGENS ,CELL lines ,CELL proliferation - Abstract
Objective To investigate the influence of CD133 + expression on patients' survival and resistance of CD133 + cells to anti-tumor agents in gastric cancer (GC). Methods Influence of CD133 expression on prognosis was analyzed employing samples from patients with GC. GC cell lines were utilized to separate CD133 + and CD133 − subpopulations by immunomagnetic separation and to analyze the biological features of two subpopulations in vitro and in vivo , especially in resistant to anti-tumor reagents and its apoptotic mechanism. Results The lower CD133 + group showed a significantly better survival compared with the higher CD133 + group. The highest content of CD133 + subpopulations for KATO-III cells had stronger proliferative ability than CD133 − subpopulations. A single CD133 + cell was capable of generating new cell colony and the tumorigenicity rate in nude mice was 100% for CD133 + clonal spheres or for CD133 + cells, but 0% for CD133 − cells. Furthermore, the higher expression levels of Oct-4, Sox-2, Musashi-1 and ABCG2 in CD133 + clonal spheres were identified compared with CD133 + cells or CD133 − cells. Under the treatment of anti-tumor reagents, CD133 + cells had lower suppression rates compared with CD133 − cells while lower level of Bcl-2 and higher level of Bax were found in CD133 + cells compared with CD133 − cells. Conclusions The patients with lower CD133 + expression had a better survival. Enriched CD133 + cells in clonal sphere shared the ability to be self-renewable, proliferative, tumorigenic and resistant to anti-tumor agents as probably regulated by Bcl-2 and Bax. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
58. Nanoparticle-mediated drug delivery for treating melanoma.
- Author
-
Mundra, Vaibhav, Li, Wei, and Mahato, Ram I
- Abstract
Melanoma originated from melanocytes is the most aggressive type of skin cancer with limited treatment options. New targeted therapeutic options with the discovery of BRAF and MEK inhibitors have shown significant survival benefits. Despite the recent progress, development of chemoresistance and systemic toxicity remains a challenge for treating metastatic melanoma. While the response from the first line of treatment against melanoma using dacarbazine remains only 5-10%, the prolonged use of targeted therapy against mutated oncogene BRAF develops chemoresistance. In this review, we will discuss the nanoparticle-based strategies for encapsulation and conjugation of drugs to the polymer for maximizing their tumor distribution through enhanced permeability and retention effect. We will also highlight photodynamic therapy and design of melanoma-targeted nanoparticles. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
59. Targeting osteopontin suppresses glioblastoma stem-like cell character and tumorigenicity in vivo.
- Author
-
Lamour, Virginie, Henry, Aurélie, Kroonen, Jérôme, Nokin, Marie‐Julie, von Marschall, Zofia, Fisher, Larry W., Chau, Tieu‐Lan, Chariot, Alain, Sanson, Marc, Delattre, Jean‐Yves, Turtoi, Andrei, Peulen, Olivier, Rogister, Bernard, Castronovo, Vincent, and Bellahcène, Akeila
- Abstract
Osteopontin (OPN) is a secreted protein involved in most aspects of tumor progression and metastasis development. Elevated OPN expression has been reported in multiple types of cancer including glioblastoma (GBM), the highest grade and most aggressive brain tumor. GBMs contain a subpopulation of glioma-initiating cells (GICs) implicated in progression, therapeutic resistance and recurrence. We have previously demonstrated that OPN silencing inhibited GBM cell growth in vitro and in vivo. Moreover, activation of CD44 signaling upon OPN ligation has been recently implicated in the acquisition of a stem cell phenotype by GBM cells. The present study is aimed to explore OPN autocrine function using shRNA silencing strategy in GICs enriched from GBM cell lines and a human primary GBM grown in EGF and bFGF defined medium. The removal of these growth factors and addition of serum induced a significant loss of OPN expression in GICs. We showed that OPN-silenced GICs were unable to grow as spheres and this capacity was restored by exogenous OPN. Importantly, the expression of Sox2, Oct3/4 and Nanog, key stemness transcription factors, was significantly decreased in GICs upon OPN targeting. We identified Akt/mTOR/p70S6K as the main signaling pathway triggered following OPN-mediated EGFR activation in GICs. Finally, in an orthotopic xenograft mouse model, the tumorigenic potential of U87-MG sphere cells was completely abrogated upon OPN silencing. Our demonstration of endogenous OPN major regulatory effects on GICs stemness phenotype and tumorigenicity implies a greater role than anticipated for OPN in GBM pathogenesis from initiation and progression to probable recurrence. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
60. Cancer stem cell targeting: Are we there yet?
- Author
-
Jung, Yuchae and Kim, Woo-Young
- Abstract
Several decades after the first notion that cancer cells are heterogeneous, not only histologically but also at the level of tumorigenicity, the existence of a tumor initiation population (so-called cancer stem cells) is now widely accepted. Here, we review the historical background leading up to the identification of this special cancer cell population and its role in the resistance of tumors to conventional therapies. In addition, we briefly review the clinical targeting strategy of the specific signaling network for cancer stem cells. Current clinical trials with newly developed drugs or older drugs for other uses are summarized to provide an understanding of the current status of strategies to target cancer stem cells. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
61. Response to Comment on 'Tumor-initiating cells establish an IL-33–TGF-β niche signaling loop to promote cancer progression'
- Author
-
Sachiko Taniguchi, Sushil Kumar, Ajit Elhance, Avery Van Duzer, Naoki Oshimori, and Justin J. Leitenberger
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,Cell ,Clone (cell biology) ,Cancer ,medicine.disease ,Tumor Initiating Cells ,Interleukin 33 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cancer research ,biology.protein ,medicine ,Bone marrow ,Antibody ,030217 neurology & neurosurgery ,Transforming growth factor - Abstract
Kamphuis et al . argue that macrophages accumulated in the proximity of tumor-initiating cells do not express the high-affinity immunoglobulin E receptor FcεRIα. Although we cannot exclude the possibility of nonspecific binding of anti-FcεRIα antibody (clone MAR-1), we provide evidence that macrophages in squamous cell carcinomas express FcεRIα and that IL-33 induces FcεRIα expression in bone marrow cell–derived macrophages.
- Published
- 2021
- Full Text
- View/download PDF
62. Functional States in Tumor-Initiating Cell Differentiation in Human Colorectal Cancer
- Author
-
Ava Oberlack, Mathias Kalxdorf, Katharina Jechow, Mark Kriegsmann, Stephan M Tirier, Jeroen Krijgsveld, Christian Conrad, Roland Eils, Karin Laaber, Torsten Müller, Teresa G Krieger, Martin Schneider, Friederike Herbst, Mario Huerta, Sebastian M. Dieter, Claudia R. Ball, Robert Lorenz Chua, Foo Wei Ten, Martina K. Zowada, Katharina Kriegsmann, Jeongbin Park, and Hanno Glimm
- Subjects
0301 basic medicine ,Metabolic state ,Cancer Research ,Colorectal cancer ,Cell ,colorectal cancer ,Biology ,tumor-initiating cells ,tumor cell differentiation ,lcsh:RC254-282 ,Article ,Tumor Initiating Cells ,03 medical and health sciences ,0302 clinical medicine ,tumor heterogeneity ,medicine ,single-cell RNA-sequencing ,Treatment resistance ,patient-derived cancer models ,Cell subpopulations ,medicine.disease ,Tumor initiating cell ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,transcriptional programs ,Cancer cell ,Cancer research ,tumor metabolism - Abstract
Simple Summary Different types of cells with tumor-initiating cell (TIC) activity contribute to colorectal cancer (CRC) progression and resistance to anti-cancer treatment. In this study, we aimed to understand whether different cell types exist within a patient-derived tumor culture, distinguishable by different patterns of their gene expression. By mRNA sequencing of patient-derived CRC cultures at the single-cell level, we defined expression programs that closely resemble differentiated cell populations of the normal intestine. Here, cell type-associated subpopulations showed differences in functional properties such as cell growth and energy metabolism. Subsequent functional analyses in vitro and in vivo demonstrated that metabolic states are linked to TIC activity in primary CRC cultures. We also show that TIC activity is dependent on oxidative phosphorylation, which may therefore represent a target for novel therapies. Abstract Intra-tumor heterogeneity of tumor-initiating cell (TIC) activity drives colorectal cancer (CRC) progression and therapy resistance. Here, we used single-cell RNA-sequencing of patient-derived CRC models to decipher distinct cell subpopulations based on their transcriptional profiles. Cell type-specific expression modules of stem-like, transit amplifying-like, and differentiated CRC cells resemble differentiation states of normal intestinal epithelial cells. Strikingly, identified subpopulations differ in proliferative activity and metabolic state. In summary, we here show at single-cell resolution that transcriptional heterogeneity identifies functional states during TIC differentiation. Furthermore, identified expression signatures are linked to patient prognosis. Targeting transcriptional states associated to cancer cell differentiation might unravel novel vulnerabilities in human CRC.
- Published
- 2021
63. Single-cell chromatin accessibility profiling of glioblastoma identifies an invasive cancer stem cell population associated with lower survival
- Author
-
Fiona J. Coutinho, Florence M.G. Cavalli, Paul Guilhamon, H. Artee Luchman, Naghmeh Rastegar, Rozina Hassam, Michelle Kushida, Divya Singhal, Heather Whetstone, Xiaoguang Hao, Jennifer A. Chan, Christopher Arlidge, Graham MacLeod, Stephane Angers, Katrina Ellestad, Michael D. Taylor, Ana Nikolic, Molly S. Shoichet, Bettina Nadorp, Mathieu Lupien, Peter B. Dirks, Laura Smith, Seyed Ali Madani Tonekaboni, Benjamin Haibe-Kains, Charles Chesnelong, Samuel Weiss, Nishani Rajakulendran, and Marco Gallo
- Subjects
Male ,0301 basic medicine ,cancer stem cell ,endocrine system ,QH301-705.5 ,Science ,Population ,Cell ,Biology ,General Biochemistry, Genetics and Molecular Biology ,single cell ATAC ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Cell Line, Tumor ,medicine ,Humans ,Biology (General) ,Cell Self Renewal ,education ,Transcription factor ,Cancer Biology ,education.field_of_study ,tumor initiating cells ,General Immunology and Microbiology ,General Neuroscience ,fungi ,Cancer ,General Medicine ,medicine.disease ,Chromatin ,ATAC ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Medicine ,Female ,Single-Cell Analysis ,Stem cell ,Glioblastoma ,Research Article ,Human - Abstract
Chromatin accessibility discriminates stem from mature cell populations, enabling the identification of primitive stem-like cells in primary tumors, such as glioblastoma (GBM) where self-renewing cells driving cancer progression and recurrence are prime targets for therapeutic intervention. We show, using single-cell chromatin accessibility, that primary human GBMs harbor a heterogeneous self-renewing population whose diversity is captured in patient-derived glioblastoma stem cells (GSCs). In-depth characterization of chromatin accessibility in GSCs identifies three GSC states: Reactive, Constructive, and Invasive, each governed by uniquely essential transcription factors and present within GBMs in varying proportions. Orthotopic xenografts reveal that GSC states associate with survival, and identify an invasive GSC signature predictive of low patient survival, in line with the higher invasive properties of Invasive state GSCs compared to Reactive and Constructive GSCs as shown by in vitro and in vivo assays. Our chromatin-driven characterization of GSC states improves prognostic precision and identifies dependencies to guide combination therapies.
- Published
- 2021
- Full Text
- View/download PDF
64. Role of autophagy in the maintenance and function of cancer stem cells.
- Author
-
VITALE, ILIO, MANIC, GWENOLA, DANDREA, VITO, and DE MARIA, RUGGERO
- Subjects
CANCER stem cells ,AUTOPHAGY ,TUMORS ,CANCER ,CYTOLOGY - Abstract
Recent advances in experimental technologies and cancer models have made possible to demonstrate that the tumor is a dynamic system comprising heterogeneous populations of cancer cells organized in a hierarchical fashion with cancer stem cells (CSCs) at the apex. CSCs are immature cells characterized by self-renewal property and long-term repopulation potential. CSCs have been causally linked to cancer initiation, propagation, spreading, recurrence and relapse as well as to resistance to anticancer therapy. A growing body of evidence suggests that the function and physiology of CSCs may be influenced by genetic/epigenetic factors and tumor environment. In this context, macroautophagy is a lysosomal degradative process (herein referred to as autophagy) critical for the adaptive response to stress and the preservation of cellular and tissue homeostasis in all eukaryotes that may have a crucial role of in the origin, maintenance and invasiveness of CSCs. The activation of the autophagic machinery is also considered as an adaptive response of CSCs to perturbation of tumor microenvironment, caused for instance by anticancer therapy. Nevertheless, compelling preclinical and clinical evidence on the cytoprotective role of autophagy for CSCs is still missing. Here, we summarize the results on the contribution of autophagy in CSCs and how it impacts tumorigenesis and tumor progression. We also discuss the therapeutical potential of the modulation of autophagy as a means to eradicate CSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
65. Pediatric and Adult High-Grade Glioma Stem Cell Culture Models Are Permissive to Lytic Infection with Parvovirus H-1
- Author
-
Rafael Josupeit, Sebastian Bender, Sonja Kern, Barbara Leuchs, Thomas Hielscher, Christel Herold-Mende, Jörg R. Schlehofer, Christiane Dinsart, Olaf Witt, Jean Rommelaere, and Jeannine Lacroix
- Subjects
oncolytic virus ,parvovirus H-1 ,H-1PV ,DIPG ,pediatric glioblastoma ,tumor initiating cells ,glioma stem-like cells ,high-grade glioma ,Microbiology ,QR1-502 - Abstract
Combining virus-induced cytotoxic and immunotherapeutic effects, oncolytic virotherapy represents a promising therapeutic approach for high-grade glioma (HGG). A clinical trial has recently provided evidence for the clinical safety of the oncolytic parvovirus H-1 (H-1PV) in adult glioblastoma relapse patients. The present study assesses the efficacy of H-1PV in eliminating HGG initiating cells. H-1PV was able to enter and to transduce all HGG neurosphere culture models (n = 6), including cultures derived from adult glioblastoma, pediatric glioblastoma, and diffuse intrinsic pontine glioma. Cytotoxic effects induced by the virus have been observed in all HGG neurospheres at half maximal inhibitory concentration (IC50) doses of input virus between 1 and 10 plaque forming units per cell. H-1PV infection at this dose range was able to prevent tumorigenicity of NCH421k glioblastoma multiforme (GBM) “stem-like” cells in NOD/SCID mice. Interestingly NCH421R, an isogenic subclone with equal capacity of xenograft formation, but resistant to H-1PV infection could be isolated from the parental NCH421k culture. To reveal changes in gene expression associated with H-1PV resistance we performed a comparative gene expression analysis in these subclones. Several dysregulated genes encoding receptor proteins, endocytosis factors or regulators innate antiviral responses were identified and represent intriguing candidates for to further study molecular mechanisms of H-1PV resistance.
- Published
- 2016
- Full Text
- View/download PDF
66. Molecular Biology of Osteosarcoma
- Author
-
Paweł Sobczuk, Michal Fiedorowicz, Piotr Rutkowski, Ewa Bartnik, Anna M. Czarnecka, Wiktoria Firlej, Kamil Synoradzki, and Paweł Grieb
- Subjects
0301 basic medicine ,musculoskeletal diseases ,Cancer Research ,theranostics ,Cabozantinib ,medicine.medical_treatment ,molecular mechanisms ,Review ,lcsh:RC254-282 ,Targeted therapy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Germline mutation ,osteosarcoma ,medicine ,Bloom syndrome ,neoplasms ,tumor initiating cells ,Retinoblastoma ,business.industry ,Cancer ,medicine.disease ,targeted therapy ,molecular imaging ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,chemistry ,Li–Fraumeni syndrome ,030220 oncology & carcinogenesis ,Cancer research ,Osteosarcoma ,business - Abstract
Osteosarcoma (OS) is the most frequent primary bone cancer in children and adolescents and the third most frequent in adults. Many inherited germline mutations are responsible for syndromes that predispose to osteosarcomas including Li Fraumeni syndrome, retinoblastoma syndrome, Werner syndrome, Bloom syndrome or Diamond–Blackfan anemia. TP53 is the most frequently altered gene in osteosarcoma. Among other genes mutated in more than 10% of OS cases, c-Myc plays a role in OS development and promotes cell invasion by activating MEK–ERK pathways. Several genomic studies showed frequent alterations in the RB gene in pediatric OS patients. Osteosarcoma driver mutations have been reported in NOTCH1, FOS, NF2, WIF1, BRCA2, APC, PTCH1 and PRKAR1A genes. Some miRNAs such as miR-21, -34a, -143, -148a, -195a, -199a-3p and -382 regulate the pathogenic activity of MAPK and PI3K/Akt-signaling pathways in osteosarcoma. CD133+ osteosarcoma cells have been shown to exhibit stem-like gene expression and can be tumor-initiating cells and play a role in metastasis and development of drug resistance. Although currently osteosarcoma treatment is based on adriamycin chemoregimens and surgery, there are several potential targeted therapies in development. First of all, activity and safety of cabozantinib in osteosarcoma were studied, as well as sorafenib and pazopanib. Finally, novel bifunctional molecules, of potential imaging and osteosarcoma targeting applications may be used in the future.
- Published
- 2020
67. The Multifactorial Role of PARP-1 in Tumor Microenvironment
- Author
-
Daniel Delgado-Bellido, Francisco Javier Oliver, Esteban Zamudio-Martínez, Santiago Serrano-Sáenz, Mónica Fernández-Cortés, Juan Manuel Martí, Ángel García-Díaz, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), Instituto de Salud Carlos III, and Fundación Domingo Martínez
- Subjects
0301 basic medicine ,Cancer Research ,autophagy ,Angiogenesis ,Poly ADP ribose polymerase ,Review ,Biology ,lcsh:RC254-282 ,Tumor Initiating Cells ,PARPs ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Autophagy ,Hypoxia ,PARP inhibitors ,Tumor microenvironment ,Tumor biology ,hypoxia ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,PARylation ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Function (biology) - Abstract
© 2020 by the authors., Poly(ADP-ribose) polymerases (PARPs), represent a family of 17 proteins implicated in a variety of cell functions; some of them possess the enzymatic ability to synthesize and attach poly (ADP-ribose) (also known as PAR) to different protein substrates by a post-translational modification; PARPs are key components in the cellular response to stress with consequences for different physiological and pathological events, especially during neoplasia. In recent years, using PARP inhibitors as antitumor agents has raised new challenges in understanding their role in tumor biology. Notably, the function of PARPs and PAR in the dynamic of tumor microenvironment is only starting to be understood. In this review, we summarized the conclusions arising from recent studies on the interaction between PARPs, PAR and key features of tumor microenvironment such as hypoxia, autophagy, tumor initiating cells, angiogenesis and cancer-associated immune response., This work was supported by Ministerio de Economía y Competividad, Spanish Ministry of Economy and Competitiveness SAF2012-40011-C02-01, SAF2015-70520-R, RTICC RD12/0036/0026, RTI2018-098968-B-I00, CIBER Cáncer ISCIII CB16/12/00421 and Fundación Domingo Martínez to FJO. JMMC, is recipient of a FPI fellowship from Spanish Ministerio de Ciencia; MFC and EZM are recipients of FPU fellowships from the Spanish Ministerio de Ciencia.
- Published
- 2020
68. Metformin turns off the metabolic switch of pancreatic cancer
- Author
-
Marie-Camille Rowell, Gerardo Ferbeyre, and Xavier Deschênes-Simard
- Subjects
Aging ,pancreatic cancer ,Cellular senescence ,Mitochondrion ,tumor-initiating cells ,Tumor Initiating Cells ,Pancreatic cancer ,medicine ,Humans ,Hypoglycemic Agents ,cellular senescence ,Cell Proliferation ,business.industry ,Cell Biology ,medicine.disease ,Metformin ,Pancreatic Neoplasms ,mitochondria ,Editorial ,Gene Expression Regulation ,Neoplastic Stem Cells ,Cancer research ,Transcriptome ,business ,medicine.drug - Published
- 2019
- Full Text
- View/download PDF
69. Salivary gland cancer stem cells.
- Author
-
Adams, April, Warner, Kristy, and Nör, Jacques E.
- Subjects
- *
CANCER stem cells , *SALIVARY gland cancer , *ONCOGENIC viruses , *CELL populations , *CANCER invasiveness , *TUMOR growth , *CANCER chemotherapy - Abstract
Summary: Emerging evidence suggests the existence of a tumorigenic population of cancer cells that demonstrate stem cell-like properties such as self-renewal and multipotency. These cells, termed cancer stem cells (CSC), are able to both initiate and maintain tumor formation and progression. Studies have shown that CSC are resistant to traditional chemotherapy treatments preventing complete eradication of the tumor cell population. Following treatment, CSC are able to re-initiate tumor growth leading to patient relapse. Salivary gland cancers are relatively rare but constitute a highly significant public health issue due to the lack of effective treatments. In particular, patients with mucoepidermoid carcinoma or adenoid cystic carcinoma, the two most common salivary malignancies, have low long-term survival rates due to the lack of response to current therapies. Considering the role of CSC in resistance to therapy in other tumor types, it is possible that this unique sub-population of cells is involved in resistance of salivary gland tumors to treatment. Characterization of CSC can lead to better understanding of the pathobiology of salivary gland malignancies as well as to the development of more effective therapies. Here, we make a brief overview of the state-of-the-science in salivary gland cancer, and discuss possible implications of the cancer stem cell hypothesis to the treatment of salivary gland malignancies. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
70. Transient treatment with epigenetic modifiers yields stable neuroblastoma stem cells resembling aggressive large-cell neuroblastomas.
- Author
-
Ikegaki, Naohiko, Shimada, Hiroyuki, Fox, Autumn M., Regan, Paul L., Jacobs, Joshua R., Hicks, Sakeenah L., Rappaport, Eric F., and Xao X. Tang
- Subjects
- *
NEUROBLASTOMA , *CANCER stem cells , *CANCER treatment , *EPIGENETICS , *NEURAL crest , *STEM cell culture , *CHROMATIN , *LABORATORY mice , *THERAPEUTICS - Abstract
Cancer stem cells (CSCs) are plastic in nature, a characteristic that hampers cancer therapeutics. Neuroblastoma (NB) is a pediatric tumor of neural crest origin, and half of the cases are highly aggressive. By treating NB cell lines [SKNAS, SKNBE(2)C, CHP134, and SY5Y] with epigenetic modifiers for a short time, followed by sphere-forming culture conditions, we have established stem cell-like NB cells that are phenotypically stable for more than a year. These cells are characterized by their high expression of sternness factors, stem cell markers, and open chromatin structure. We referred to these cells as induced CSCs (iCSCs). SKNAS iCSC and SKNBE(2)C iCSC clones (as few as 100 cells) injected s.c. into SCID/Beige mice formed tumors, and in one case, SKNBE(2)C iCSCs metastasized to the adrenal gland, suggesting their increased metastatic potential. SKNAS iCSC xenografts showed the histologic appearance of totally undifferentiated large-cell NBs (LCNs), the most aggressive and deadly form of NB in humans. Immunohistochemical analyses showed that SKNAS iCSC xenografts expressed high levels of the stem cell marker CXCR4, whereas the SKNAS monolayer cell xenografts did not. The patterns of CXCR4 and MYC expression in SKNAS iCSC xenografts resembled those in the LCNs. The xenografts established from the NB iCSCs shared two common features: the LCN phenotype and high-level MYC/MYCN expression. These observations suggest both that NB cells with large and vesicular nuclei, representing their open chromatin structure, are indicative of stem cell-like tumor cells and that epigenetic changes may have contributed to the development of these most malignant NB cells. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
71. Quercetin in elimination of tumor initiating stem-like and mesenchymal transformation property in head and neck cancer.
- Author
-
Chang, Wen–Wei, Hu, Fang–Wei, Yu, Cheng–Chia, Wang, Hsiu–Huan, Feng, Hsiang–Pu, Lan, Chih, Tsai, Lo–Lin, and Chang, Yu–Chao
- Subjects
QUERCETIN ,HEAD & neck cancer treatment ,CANCER cell proliferation ,WESTERN immunoblotting ,FIBROBLASTS ,METASTASIS - Abstract
Background Previously, we enriched a subpopulation of head and neck cancer-derived tumor initiating cells (HNC-TICs) presented high tumorigenic, chemo-radioresistant, and coupled with epithelial-mesenchymal transition (EMT) properties. The purpose of this study was to investigate the therapeutic effect and molecular mechanisms of quercetin on HNC-TICs. Method ALDH1 activity of head and neck cancer cells with quercetin treatment was assessed by the Aldefluor assay flow cytometry analysis. Self-renewal, invasiveness, and EMT capability of HNC-TICs with different doses of quercetin was presented. Results We first observed that the treatment of quercetin significantly downregulated the ALDH1 activity of head and neck cancer cells in a dose-dependent manner ( p < .05). Moreover, quercetin reduced self-renewal property and stemness signatures expression in head and neck cancer-derived sphere cells. The migration ability of head and neck cancer-derived sphere cells was lessened under quercetin treatment partially due to the decreased productions of Twist, N-cadherin, and vimentin. Conclusion Quercetin suppressing HNC-TICs characteristics may therefore be valuable therapeutics clinically in combination with standard treatment modalities. © 2012 Wiley Periodicals, Inc. Head Neck, 2013 [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
72. Sorafenib selectively depletes human glioblastoma tumor-initiating cells from primary cultures.
- Author
-
Carra, Elisa, Barbieri, Federica, Marubbi, Daniela, Pattarozzi, Alessandra, Favoni, Roberto E., Florio, Tullio, and Daga, Antonio
- Published
- 2013
- Full Text
- View/download PDF
73. Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization.
- Author
-
Tirino, Virginia, Desiderio, Vincenzo, Paino, Francesca, De Rosa, Alfredo, Papaccio, Federica, La Noce, Marcella, Laino, Luigi, De Francesco, Francesco, and Papaccio, Gianpaolo
- Subjects
- *
CANCER stem cells , *STEM cells , *PROGENITOR cells , *CANCER-related mortality , *METASTASIS - Abstract
Primary tumors are responsible for 10% of cancer deaths. In most cases, the main cause of mortality is the formation of metastases. Accumulating evidence suggests that a subpopulation of tumor cells with distinct stem-like properties is responsible for tumor initiation, invasive growth, and metastasis formation. This population is defined as cancer stem cells (CSCs). Existing therapies have enhanced the length of survival after diagnosis of cancer but have completely failed in terms of recovery. CSCs appear to be resistant to chemotherapy, may remain quiescent for extended periods, and have affinity for hypoxic environments. The CSCs can be identified and isolated by different methodologies, including isolation by CSC-specific cell surface marker expression, detection of side population phenotype by Hoechst 33342 exclusion, assessment of their ability to grow as floating spheres, and aldehyde dehydrogenase (ALDH) activity assay. None of the methods mentioned are exclusively used to isolate the solid tumor CSCs, highlighting the imperative to delineate more specific markers or to use combinatorial markers and methodologies. This review provides an overview of the main characteristics and approaches used to identify, isolate, and characterize CSCs from solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
74. The importance of HER2 signaling in the tumor-initiating cell population in aromatase inhibitor-resistant breast cancer.
- Author
-
Gilani, Rabia, Kazi, Armina, Shah, Preeti, Schech, Amanda, Chumsri, Saranya, Sabnis, Gauri, Jaiswal, Anil, and Brodie, Angela
- Abstract
Aromatase inhibitors (AIs) are an effective therapy in treating estrogen receptor-positive breast cancer. Nonetheless, a significant percentage of patients either do not respond or become resistant to AIs. Decreased dependence on ER-signaling and increased dependence on growth factor receptor signaling pathways, particularly human epidermal growth factor receptor 2 (EGFR2/HER2), have been implicated in AI resistance. However, the role of growth factor signaling remains unclear. This current study investigates the possibility that signaling either through HER2 alone or through interplay between epidermal growth factor receptor 1 (EGFR/HER1) and HER2 mediates AI resistance by increasing the tumor initiating cell (TIC) subpopulation in AI-resistant cells via regulation of stem cell markers, such as breast cancer resistance protein (BCRP). TICs and BCRP are both known to be involved in drug resistance. Results from in vitro analyses of AI-resistant versus AI-sensitive cells and HER2-versus HER2+ cells, as well as from in vivo xenograft tumors, indicate that (1) AI-resistant cells overexpress both HER2 and BCRP and exhibit increased TIC characteristics compared to AI-sensitive cells; (2) inhibition of HER2 and/or BCRP decrease TIC characteristics in letrozole-resistant cells; and (3) HER2 and its dimerization partner EGFR/HER1 are involved in the regulation of BCRP. Overall, these results suggest that reducing or eliminating the TIC subpopulation with agents that target BCRP, HER2, EGFR/HER1, and/or their downstream kinase pathways could be effective in preventing and/or treating acquired AI resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
75. Basal-like breast cancer stem cells are sensitive to anti-DR5 mediated cytotoxicity.
- Author
-
Londoño-Joshi, Angelina, Oliver, Patsy, Li, Yufeng, Lee, Choo, Forero-Torres, Andres, LoBuglio, Albert, and Buchsbaum, Donald
- Abstract
Breast cancer stem cells (BrCSC) are resistant to common therapeutic modalities including chemotherapy, radiation, and hormonal agents. They are thought to contribute to treatment resistance, relapse, and metastases. This study examines the effect of a monoclonal anti-DR5 antibody (TRA-8) and chemotherapy (adriamycin, taxol) on BrCSC populations from basal-like breast cancer cell lines. Doubly enriched BrCSC (CD44, CD24, ALDH) cells were exposed to TRA-8 and control reagents and examined for cytotoxicity, caspase activation, tumorsphere formation and tumorigenicity. Doubly enriched BrCSC populations expressed cell surface DR5 and were sensitive to TRA-8 mediated cytotoxicity with induction of caspase 8 and 3 activation. TRA-8 at sub-nanomolar concentrations inhibited 2LMP and SUM159 BrCSC tumorsphere formation and was more than 50-fold more inhibitory than TRAIL or anti-DR4 at equimolar concentrations. Chemotherapy treatment of 2LMP and SUM159 cell lines resulted in a relative increase of BrCSC, whereas TRA-8 produced a decrease in the percentage of BrCSC. TRA-8 exposure to 2LMP and SUM159 BrCSC preparations produced significant inhibition of tumorigenicity. DR5 maybe a therapeutic target on the surface of basal-like BrCSC which is amenable to agonistic monoclonal anti-DR5 therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
76. Aberrant microRNAs expression in CD133/CD326 human lung adenocarcinoma initiating cells from A549.
- Author
-
Lin, Sheng, Sun, Jian-guo, Wu, Jing-bo, Long, Hai-xia, Zhu, Cong-hui, Xiang, Tong, Ma, Hu, Zhao, Zhong-quan, Yao, Quan, Zhang, An-mei, Zhu, Bo, and Chen, Zheng-tang
- Abstract
Increasing evidence demonstrates that miRNAs are involved in the dysregulation of tumor initiating cells (TICs) in various tumors. Due to a lack of definitive markers, cell sorting is not an ideal separation method for lung adenocarcinoma initiating cells. In this study, we combined paclitaxel with serum-free medium cultivation (inverse-induction) to enrich TICs from A549 cells, marked by CD133/CD326, defined features of stemness. We next investigated aberrant microRNAs in this subpopulation compared to normal cells with miRNA microarray and found that 50 miRNAs exhibited a greater than 2-fold change in expression. As further validation, 10 miRNAs were chosen to perform quantitative RT-PCR on the A549 cell line and primary samples. The results suggest that aberrant expression of miRNAs such as miR-29ab, miR-183, miR-17-5p and miR-127-3P may play an important role in regulating the bio-behavior of TICs. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
77. Defining the origins of Ras/p53-mediated squamous cell carcinoma.
- Author
-
White, Andrew C., Trana, Kathy, Khuu, Joan, Dang, Christine, Yongyan Cui, Binder, Scott W., and Lowry, William E.
- Subjects
- *
SQUAMOUS cell carcinoma , *CANCER , *PAPILLARY carcinoma , *CANCER cells , *GENOTYPE-environment interaction - Abstract
The precise identity of cancer cells of origin and the molecular events of tumor initiation in epidermal squamous cell carcinoma (5CC) are unknown. Here we show that malignancy potential is related to the developmental capacity of the initiating cancer cell in a genetically defined, intact, and inducible in vivo model. Specifically, these data demonstrate that SCCs can originate from inside the hair follicle stem cell (SC) niche or from immediate progenitors, whereas more developmentally restricted progeny, the transit amplifying (TA) cells, are unable to generate even benign tumors in the same genetic context. Using a temporal model of tumorigenesis in situ, we high-light the phenotypes of cancer progression from the hair follicle SC niche, including hyperplasia, epithelial to mesenchymal transition, and 5CC formation. Furthermore, we provide insights into the inability of hair follicle TA cells to respond'to tumorigenic stimuli. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
78. Cancer Stem Cells and Pediatric Solid Tumors.
- Author
-
Friedman, Gregory K. and Gillespie, G. Yancey
- Subjects
- *
STEM cells , *TUMORS , *CHILDHOOD cancer , *DRUG therapy , *RADIATION , *PEDIATRICS - Abstract
Recently, a subpopulation of cells, termed tumor-initiating cells or tumor stem cells (TSC), has been identified in many different types of solid tumors. These TSC, which are typically more resistant to chemotherapy and radiation compared to other tumor cells, have properties similar to normal stem cells including multipotency and the ability to self-renew, proliferate, and maintain the neoplastic clone. Much of the research on TSC has focused on adult cancers. With considerable differences in tumor biology between adult and pediatric cancers, there may be significant differences in the presence, function and behavior of TSC in pediatric malignancies. We discuss what is currently known about pediatric solid TSC with specific focus on TSC markers, tumor microenvironment, signaling pathways, therapeutic resistance and potential future therapies to target pediatric TSC. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
79. Plasticité du phenotype cellulaire ou une certaine fin de I'insouciance du déterminisme.
- Author
-
Chneiweiss, Hervé
- Subjects
ATOMIC bomb blast effect ,HEMATOPOIETIC stem cell transplantation ,ATOMIC bomb victims ,PHENOTYPIC plasticity ,STEM cells ,BIOLOGY ,ONCOLOGY - Abstract
The article presents a study regarding the discovery of hematopoietic stem cells and their phenotypic plasticity, in response to environmental factors. It states that the discovery was due to the tragical consequences of the Hiroshima and Nagasaki atomic bombs in 1945. Moreover, the mechanisms underlying the plasticity, the frontiers of development biology and oncology are also discussed.
- Published
- 2011
- Full Text
- View/download PDF
80. Specific posttranslational modification regulates early events in mammary carcinoma formation.
- Author
-
Hua-Bei Guo, Johnson, Heather, Randolph, Matthew, Nagy, Tamas, Blalock, Ryan, and Pierce, Michael
- Subjects
- *
ENZYMES , *GLYCOPROTEINS , *BREAST cancer , *CARCINOGENESIS , *CELL culture , *EPITHELIAL cells - Abstract
The expression of an enzyme, GnT-V, that catalyzes a specific posttranslational modification of a family of glycoproteins, namely a branched N-glycan, is transcriptionally up-regulated during breast carcinoma oncogenesis. To determine the molecular basis of how early events in breast carcinoma formation are regulated by GnT-V, we studied both the early stages of mammary tumor formation by using 3D cell culture and a her-2 transgenic mouse mammary tumor model. Overexpression of GnT-V in MCF-10A mammary epithelial cells in 3D culture disrupted acinar morphogenesis with impaired hollow lumen formation, an early characteristic of mammary neoplastic transformation. The disrupted acinar morphogenesis of mammary tumor cells in 3D culture caused by her-2 expression was reversed in tumors that lacked GnT-V expression. Moreover, her-2-induced mammary tumor onset was significantly delayed in the GnT-V null tumors, evidence that the lack of the posttranslational modification catalyzed by GnT-V attenuated tumor formation. Inhibited activation of both PKB and ERK signaling pathways was observed in GnT-V null tumor cells. The proportion of tumor-initiating cells (TICs) in the mammary tumors from GnT-V null mice was significantly reduced compared with controls, and GnT-V null TICs displayed a reduced ability to form secondary tumors in NOD/SCID mice. These results demonstrate that GnT-V expression and its branched glycan products effectively modulate her-2-mediated signaling pathways that, in turn, regulate the relative proportion of tumor initiating cells and the latency of her-2-driven tumor onset. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
81. Oncogene-Driven Hemostatic Changes in Cancer.
- Author
-
Rak, Janusz, Yu, Joanne, and Milsom, Chloe
- Subjects
- *
ONCOGENES , *BLOOD coagulation disorders , *CANCER complications , *HEMOSTASIS , *THROMBOSIS , *DISEASE risk factors - Abstract
A common feature in progression of multiple human malignancies is the protracted deregulation of the coagulation system, often referred to as cancer coagulopathy. The genesis of this syndrome can be traced to changes in the tumor vascular interface, formed through vascular invasion, angiogenesis, and metastasis. The resulting contact between cancer cells and the circulating components of the coagulation system compromise the regulatory barriers that normally control physiological hemostasis. In addition, cancer cells and their stroma often exhibit procoagulant properties. While these changes have long been thought to be unspecific in nature, evidence now exists to suggest that cancer coagulopathy and the related Trousseau syndrome are a function of the genetic tumor progression. Indeed, the expression of several effector molecules of the coagulation and fibrinolytic systems, including: tissue factor (TF), plasminogen activator inhibitor 1 (PAI-1), cyclooxygenase 2 (COX-2), or urokinase (uPA) are often direct regulatory targets of oncogenes (K-ras, EGFR, HER-2, c-MET) and tumor suppressors (p53, PTEN). Moreover, oncogenic alterations act on coagulation indirectly by driving formation of leaky vessels, metastasis, or inflammation. These procoagulant influences of oncogenic pathways are modulated by hypoxia, stress responses and cellular differentiation, the latter involving formation of cancer stem cells and their niches to which coagulation factors may contribute. It is possible that targeting cancer-related coagulopathy may require more cancer-specific measures to reduce thrombosis burden and improve overall survival. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
82. Cancer stem cells – old concepts, new insights.
- Author
-
Vermeulen, L., Sprick, M. R., Kemper, K., Stassi, G., and Medema, J. P.
- Subjects
- *
CANCER , *STEM cells , *GENETIC disorders , *ONCOGENES , *TUMORS - Abstract
Cancer has long been viewed as an exclusively genetic disorder. The model of carcinogenesis, postulated by Nowell and Vogelstein, describes the formation of a tumor by the sequential accumulation of mutations in oncogenes and tumor suppressor genes. In this model, tumors are thought to consist of a heterogeneous population of cells that continue to acquire new mutations, resulting in a highly dynamic process, with clones that out compete others due to increased proliferative or survival capacity. However, novel insights in cancer stem cell research suggest another layer of complexity in the process of malignant transformation and preservation. It has been reported that only a small fraction of the cancer cells in a malignancy have the capacity to propagate the tumor upon transplantation into immuno-compromised mice. Those cells are termed ‘cancer stem cells’ (CSC) and can be selected based on the expression of cell surface markers associated with immature cell types. In this review, we will critically discuss these novel insights in CSC-related research. Where possible we integrate these results within the genetic model of cancer and illustrate that the CSC model can be considered an extension of the classic genetic model rather than a contradictory theory. Finally, we discuss some of the most controversial issues in this field.Cell Death and Differentiation (2008) 15, 947–958; doi:10.1038/cdd.2008.20; published online 15 February 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
83. Lung cancer stem cells – the role in pathogenesis and progressive growth of cancer
- Author
-
Joanna Domagała-Kulawik and Agata Raniszewska
- Subjects
Microbiology (medical) ,cancer stem cells ,tumor initiating cells ,business.industry ,lcsh:R ,Cancer ,lcsh:Medicine ,medicine.disease ,Pathogenesis ,lung cancer ,Infectious Diseases ,oncogenesis ,medicine ,Cancer research ,Stem cell ,Lung cancer ,business - Abstract
Lung cancer is the main cause of cancer death worldwide. Rapid accurate diagnosis, recognition of risk factors and improvement of treatment efficacy represent the main challenges. An advanced stage of the disease at the time of diagnosis, observed in the majority of cases, makes the introduction of radical treatment impossible or ineffective. Immunotherapy was a breakthrough in achieving long-term survival in recent years. The reason for developing this type of treatment is to know the interaction between the host cells and tumor cells. Cancer stem cells theory is increasingly gaining greater significance in the world. The biological heterogeneity of lung cancer may be due to the presence of a small percentage of cancer stem cells (CSCs). CSCs are the results of genetic and epigenetic changes in normal stem cells, progenitor cells or differentiated cells. CSCs are functionally defined by their unlimited self-renewal capacity, multiple differentiation and their ability to imitate tumors. It is believed that CSCs determine unrestricted growth of tumors and their different morphology. Their ability to self-renew can be the cause of relapses even after long periods of remission. CSCs reside in niches, where they receive signals for differentiation and proliferation process. The identification of CSCs is based on the presence of specific molecules (CD133, CD44, CD90, ALDH, EpCAM). An important feature of CSCs is their resistance to conventional cancer treatment: radio therapy and chemotherapy. Presumably, CSCs are responsible for the synthesis of immunosuppressive particles, and recruiting other molecules, which have immunosuppressive properties. The paper presents the current state of knowledge on CSCs in lung cancer detailing adenocarcinoma.
- Published
- 2017
84. Iron addiction: a novel therapeutic target in ovarian cancer
- Author
-
Frank McKeon, Miranda L. Lynch, Christopher P. Crum, Poornima Hegde, Suzy V. Torti, Gang Ning, Frank M. Torti, Wa Xian, Molly Brewer, Zhiyong Deng, Yusuke Yamamoto, Bibbin T. Paul, Nathaniel A. Dyment, Lia Tesfay, Xiaohong Wang, Lance D. Miller, and Debargha Basuli
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Ferroportin ,Transferrin receptor ,Biology ,Molecular oncology ,Article ,Mice ,03 medical and health sciences ,iron ,Growth factor receptor ,Internal medicine ,Genetic model ,Genetics ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Interleukin 6 ,Molecular Biology ,Ovarian Neoplasms ,tumor initiating cells ,Cancer ,medicine.disease ,ferroptosis ,ovarian cancer ,030104 developmental biology ,Endocrinology ,biology.protein ,Cancer research ,Female ,Ovarian cancer - Abstract
Ovarian cancer is a lethal malignancy that has not seen a major therapeutic advance in over 30 years. We demonstrate that ovarian cancer exhibits a targetable alteration in iron metabolism. Ferroportin (FPN), the iron efflux pump, is decreased, and transferrin receptor (TFR1), the iron importer, is increased in tumor tissue from patients with high grade but not low grade serous ovarian cancer. A similar profile of decreased FPN and increased TFR1 is observed in a genetic model of ovarian cancer tumor initiating cells (TICs). The net result of these changes is an accumulation of excess intracellular iron and an augmented dependence on iron for proliferation. A forced reduction in intracellular iron reduces the proliferation of ovarian cancer TICs in vitro, and inhibits both tumor growth and intraperitoneal dissemination of tumor cells in vivo. Mechanistic studies demonstrate that iron increases metastatic spread by facilitating invasion through expression of matrix metalloproteases and synthesis of IL6. We show that the iron dependence of ovarian cancer tumor initiating cells renders them exquisitely sensitive in vivo to agents that induce iron-dependent cell death (ferroptosis) as well as iron chelators, and thus creates a metabolic vulnerability that can be exploited therapeutically.
- Published
- 2017
- Full Text
- View/download PDF
85. Jarid2 is essential for the maintenance of tumor initiating cells in bladder cancer
- Author
-
Chun-Zhi Ai, Shan Jiang, Xiang-Zhen Wang, Min Niu, Shanshan Xu, Xinxing Zhu, Shaoqi Tian, Yi-Zhou Jiang, Guangyao Li, Ya-Wei Yan, Xifeng Lu, Genshen Zhong, Shaojun Tang, and Yu Xue
- Subjects
0301 basic medicine ,Oncology ,Gerontology ,medicine.medical_specialty ,Population ,p16 ,urologic and male genital diseases ,tumor-initiating cells ,Jarid2 ,Tumor Initiating Cells ,03 medical and health sciences ,Cell Line, Tumor ,Internal medicine ,Humans ,Medicine ,histone modification ,University medical ,education ,education.field_of_study ,Bladder cancer ,business.industry ,Polycomb Repressive Complex 2 ,medicine.disease ,Urologic malignancy ,bladder tumors ,030104 developmental biology ,Urinary Bladder Neoplasms ,Cancer cell ,Neoplastic Stem Cells ,Christian ministry ,Stem cell ,business ,Signal Transduction ,Research Paper - Abstract
// Xin-Xing Zhu 1, 2, * , Ya-Wei Yan 1, 2, * , Chun-Zhi Ai 1 , Shan Jiang 1 , Shan-Shan Xu 1 , Min Niu 3 , Xiang-Zhen Wang 4 , Gen-Shen Zhong 5 , Xi-Feng Lu 6 , Yu Xue 7 , Shaoqi Tian 8 , Guangyao Li 9 , Shaojun Tang 10 , Yi-Zhou Jiang 1 1 Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong, China 2 Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China 3 Department of Statics, University of Wisconsin-Madison, Madison, WI, USA 4 Maternal and Child Health Hospital of Nanshan District, Shenzhen, Guangdong, China 5 The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China 6 Department of Physiology, Center for Diabetes, Obesity and Metabolism, Shenzhen University, Shenzhen, Guangdong, China 7 Minnan Normal University, Zhangzhou, Fujian, China 8 The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China 9 Department of Health Outcomes and Policy, College of Medicine, University of Florida, Gainesville, FL, USA 10 Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA * These authors contributed equally to this work Correspondence to: Yi-Zhou Jiang, email: jiangyz@szu.edu.cn Keywords: Jarid2, bladder tumors, tumor-initiating cells, p16, histone modification Received: November 24, 2016 Accepted: February 07, 2017 Published: February 20, 2017 ABSTRACT Bladder cancer is the most common urologic malignancy in China, with an increase of the incidence and mortality rates over past decades. Recent studies suggest that bladder tumors are maintained by a rare fraction of cells with stem cell proprieties. Targeting these bladder tumor initiating cell (TICs) population can overcome the drug-resistance of bladder cancer. However, the molecular and genetic mechanisms regulating TICs in bladder cancer remain poorly defined. Jarid2 is implicated in signaling pathways regulating cancer cell epithelial-mesenchymal transition, and stem cell maintenance. The goal of our study was to examine whether Jarid2 plays a role in the regulation of TICs in bladder cancer. We found that knockdown of Jarid2 was able to inhibit the invasive ability and sphere-forming capacity in bladder cancer cells. Moreover, knockdown of Jarid2 reduced the proportion of TICs and impaired the tumorigenicity of bladder cancer TICs in vivo . Conversely, ectopic overexpression of Jarid2 promoted the invasive ability and sphere-forming capacity in bladder cancer cells. Mechanistically, reduced Jarid2 expression led to the upregulation of p16 and H3K27me3 level at p16 promoter region. Collectively, we provided evidence that Jarid2 via modulation of p16 is a putative novel therapeutic target for treating malignant bladder cancer.
- Published
- 2017
- Full Text
- View/download PDF
86. Breast Cancer: IL1R2 Blockade Suppresses Breast Tumorigenesis and Progression by Impairing USP15‐Dependent BMI1 Stability (Adv. Sci. 1/2020)
- Author
-
Lixing Zhang, Adeel ur Rehman, Lei Zhou, Weilong Chen, Xin Hu, Zhi-Ming Shao, Xueyan He, Ying Du, Yi-Zhou Jiang, Jian Zhang, Dandan Sheng, Tao Li, Xi‐chun Hu, Suling Liu, Dong Wang, Jing Feng, Jiankun Qiang, and Xiaoli Yang
- Subjects
General Chemical Engineering ,General Physics and Astronomy ,Medicine (miscellaneous) ,Interleukin 1 receptor, type II ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Tumor Initiating Cells ,Breast cancer ,breast cancer ,medicine ,General Materials Science ,Inside Front Cover ,Neutralizing antibody ,biology ,tumor initiating cells ,business.industry ,General Engineering ,neutralizing antibody ,medicine.disease ,BMI1 ,Blockade ,USP15 ,IL1R2 ,Cancer research ,biology.protein ,Carcinogenesis ,business - Abstract
In article https://doi.org/10.1002/advs.201901728, Suling Liu and co‐workers report that the “decoy receptor” IL‐1 receptor 2 (IL1R2) is highly expressed on the most tumorigenic intermediate‐state breast tumor initiation cells (BTICs), which fall in between the epithelial‐like state and mesenchymal‐like state. IL‐1β in the tumor microenvironment activates the IL1R2 signaling pathway and then increases intermediate‐state BTIC self‐renewal and tumor progression, which can be specifically blocked by IL1R2 neutralizing antibody.
- Published
- 2020
87. Progastrin production transitions from Bmi1+/Prox1+ to Lgr5high cells during early intestinal tumorigenesis
- Author
-
Bénédicte Brulin, Unmesh Jadhav, Philippe Crespy, Zeinab Homayed, Jihane Boubaker-Vitre, Fanny Grillet, J. Hazerbroucq, Frédéric Hollande, Cyril Breuker, Julie Giraud, Christine Pignodel, J-F. Bourgaux, Ramesh A. Shivdasani, Philippe Jay, Julie Pannequin, Momeneh Foroutan, MORNET, Dominique, Institut de Génomique Fonctionnelle (IGF), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), University of Melbourne, Dana-Farber Cancer Institute [Boston], Harvard Medical School [Boston] (HMS), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Hôpital Universitaire Carémeau [Nîmes] (CHU Nîmes), Centre Hospitalier Universitaire de Nîmes (CHU Nîmes), and Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Cancer Research ,FACS, Fluorescence-Activated Cell Sorting ,[SDV]Life Sciences [q-bio] ,Intestinal polyp ,Enteroendocrine cell ,Biology ,Tumor initiating cells ,lcsh:RC254-282 ,Lgr5 (GPR49) ,03 medical and health sciences ,0302 clinical medicine ,Intestinal mucosa ,Lgr5, leucine-rich repeat containing G protein-coupled receptor 5 ,Neoplastic transformation ,APC, Adenomatous Polyposis Coli ,Original Research ,PG, progastrin ,Progastrin ,Intestinal stem cells ,Wnt signaling pathway ,LGR5 ,CBC, crypt base columnar cells ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Adenomas ,3. Good health ,SPF, Specific Pathogen Free ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,EGFP, Enhanced Green Fluorescence Protein ,Oncology ,BMI1 ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,DAPI, 4′,6-diamidino-2-phenylindole - Abstract
Highlights • Secretion of progastrin is a signature event of early malignant transformation in the colon. • In the healthy epithelium, progastrin is produced by a subset of enteroendocrine cells expressing both Bmi1 and Prox1. • LGR5-high intestinal stem cells are a primary source of progastrin production in early mouse and human intestinal adenomas., Progastrin is an unprocessed soluble peptide precursor with a well-described tumor-promoting role in colorectal cancer. It is expressed at small levels in the healthy intestinal mucosa, and its expression is enhanced at early stages of intestinal tumor development, with high levels of this peptide in hyperplastic intestinal polyps being associated with poor neoplasm-free survival in patients. Yet, the precise type of progastrin-producing cells in the healthy intestinal mucosa and in early adenomas remains unclear. Here, we used a combination of immunostaining, RNAscope labelling and retrospective analysis of single cell RNAseq results to demonstrate that progastrin is produced within intestinal crypts by a subset of Bmi1+/Prox1+/LGR5low endocrine cells, previously shown to act as replacement stem cells in case of mucosal injury. In contrast, our findings indicate that intestinal stem cells, specified by expression of the Wnt signaling target LGR5, become the main source of progastrin production in early mouse and human intestinal adenomas. Collectively our results suggest that the previously identified feed-forward mechanisms between progastrin and Wnt signaling is a hallmark of early neoplastic transformation in mouse and human colonic adenomas.
- Published
- 2020
- Full Text
- View/download PDF
88. IL1R2 Blockade Suppresses Breast Tumorigenesis and Progression by Impairing USP15‐Dependent BMI1 Stability
- Author
-
Jian Zhang, Yi-Zhou Jiang, Dandan Sheng, Dong Wang, Xueyan He, Xiaoli Yang, Xin Hu, Jing Feng, Tao Li, Zhi Ming Shao, Jiankun Qiang, Suling Liu, Xi Chun Hu, Lei Zhou, Ying Du, Weilong Chen, Adeel ur Rehman, and Lixing Zhang
- Subjects
General Chemical Engineering ,Cell ,Population ,General Physics and Astronomy ,Medicine (miscellaneous) ,02 engineering and technology ,010402 general chemistry ,medicine.disease_cause ,01 natural sciences ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Immune system ,breast cancer ,Downregulation and upregulation ,medicine ,General Materials Science ,education ,Neutralizing antibody ,lcsh:Science ,education.field_of_study ,biology ,Full Paper ,tumor initiating cells ,Cell growth ,Chemistry ,General Engineering ,neutralizing antibody ,Full Papers ,021001 nanoscience & nanotechnology ,BMI1 ,0104 chemical sciences ,USP15 ,medicine.anatomical_structure ,IL1R2 ,Cancer research ,biology.protein ,lcsh:Q ,0210 nano-technology ,Carcinogenesis - Abstract
Breast tumor initiating cells (BTICs) with ALDH+CD24−CD44+ phenotype are the most tumorigenic and invasive cell population in breast cancer. However, the molecular mechanisms are still unclear. Here, it is found that a negative immune regulator interleukin‐1 receptor type 2 (IL1R2) is upregulated in breast cancer (BC) tissues and especially in BTICs. BC patients with high IL1R2 expression have a poorer overall survival and relapse‐free survival. High IL1R2 promotes BTIC self‐renewal and BC cell proliferation and invasion. Mechanistically, IL1R2 is activated by IL1β, as demonstrated by the fact that IL1β induces the release of IL1R2 intracellular domain (icd‐IL1R2) and icd‐IL1R2 then interacts with the deubiquitinase USP15 at the UBL2 domain and promotes its activity, which finally induces BMI1 deubiquitination at lysine 81 and stabilizes BMI1 protein. In addition, IL1R2 neutralizing antibody can suppress the protein expression of both IL1R2 and BMI1, and significantly abrogates the promoting effect of IL1R2 on BTIC self‐renewal and BC cell growth both in vitro and in vivo. The current results indicate that blocking IL1R2 with neutralizing antibody provides a therapeutic approach to inhibit BC progression by targeting BTICs., Interleukin‐1 receptor type 2 (IL1R2) increases BMI1 deubiquitination and stability via binding and enhancing the activity of ubiquitin‐specific protease 15 (USP15) in cell nuclei, intrinsically promoting the self‐renewal of breast tumor initiating cells (BTICs) as well as breast cancer cell proliferation and invasion. Targeting IL1R2 with its neutralizing antibody inhibits the self‐renewal of BTICs, breast tumorigenesis, and cancer resistance to docetaxel.
- Published
- 2019
89. NOTCH3-targeted antibody drug conjugates regress tumors by inducing apoptosis in receptor cells and through transendocytosis into ligand cells
- Author
-
Magali Guffroy, Steven Pirie-Shepherd, Puja Sapra, Judy Lucas, Jonathon Golas, Kenneth G. Geles, Joel Bard, Nicole Piche-Nicholas, Riyez Karim, Hans-Peter Gerber, Latha Sridharan, Lioudmila Tchistiakova, Andreas Giannakou, Jing Zhang, Manoj Charati, Tania Franks, Wenyan Zhong, Ting-Ting Yamin, Yijie Gao, Jessie Qian, Marc D. Roy, Andreas Maderna, and Christopher J. O’Donnell
- Subjects
cancer stem cells ,Medicine (General) ,Immunoconjugates ,Notch signaling pathway ,Antineoplastic Agents ,Apoptosis ,DLL4 ,Clathrin ,Article ,General Biochemistry, Genetics and Molecular Biology ,R5-920 ,biotherapeutic ,Cancer stem cell ,Cell Line, Tumor ,Caveolin ,Humans ,Receptor ,Cytotoxicity ,Receptor, Notch3 ,Receptors, Notch ,tumor initiating cells ,antibody drug conjugates ,biology ,Chemistry ,Oncogenes ,Ligand (biochemistry) ,Xenograft Model Antitumor Assays ,notch signaling ,NOTCH3 receptor ,body regions ,transendocytosis ,oncology ,biology.protein ,Cancer research ,Receptor clustering ,JAG1 - Abstract
Summary Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition., Graphical abstract, Highlights NOTCH3 receptor is overexpressed in breast, lung, and ovarian tumors Newly generated NOTCH3-targeted antibody drug conjugates are efficacious and safe NOTCH3 antibodies internalize through different routes depending on signaling status NOTCH3 antibody intercellular trafficking occurs by transendocytosis into ligand cells, Inhibition of Notch signaling in cancer is challenging due to dose-limiting, on-target toxicities associated with inhibition of its normal function. Geles et al. demonstrate that a NOTCH3-targeted antibody drug conjugate without signaling inhibition is efficacious by killing receptor and ligand expressing cells, and thus has a lower potential for side effects.
- Published
- 2021
- Full Text
- View/download PDF
90. Trending Speculations of Tumor-Initiating Cells in Squamous Cell Cancers of Head and Neck
- Author
-
Vidya Kadashetti, Wasim Kamate, Sushma Gugwad, Uzma Belgaumi, Rajendra Baad, and Nupura Vibhute
- Subjects
0301 basic medicine ,Review Article ,Malignancy ,medicine.disease_cause ,Tumor Initiating Cells ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Head and neck ,Tumor cell population ,Squamous cell cancer ,business.industry ,Tumor-initiating cells ,Cancer ,Cell Biology ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Squamous cell cancer of head and neck ,Undifferentiated cell ,Stem cell ,Carcinogenesis ,business ,Developmental Biology - Abstract
Tumor-initiating cells are a diminutive subpopulation of stem cells that have ability of long term self-renewal and generation of varied traits of tumor cell population. Understanding the concept of tumor-initiating cells may have a great implicative intimation for our comprehension of cancer pathobiology and for the delineation of new therapies directed towards these stem cells. The present review is an endeavor to conceptualize the role of tumor-initiating cells in the Squamous Cell Cancers (SCC) of head and neck, their role in tumorigenesis and the possible supplementary approach in the latest treatment modalities.
- Published
- 2017
91. Structure-optimized interpolymer polyphosphazene complexes for effective gene delivery to glioblastoma
- Author
-
Universidade de Santiago de Compostela. Centro de Investigación en Medicina Molecular e Enfermidades Crónicas, Hsu, Wei-Hsin, Sánchez-Gómez, Pilar, Gomez-Ibarlucea, Esther, Ivanov, Delyan P., Rahman, Ruman, Grabowska, Anna M., Csaba, Noemi Stefania, Alexander, Cameron, García Fuentes, Marcos, Universidade de Santiago de Compostela. Centro de Investigación en Medicina Molecular e Enfermidades Crónicas, Hsu, Wei-Hsin, Sánchez-Gómez, Pilar, Gomez-Ibarlucea, Esther, Ivanov, Delyan P., Rahman, Ruman, Grabowska, Anna M., Csaba, Noemi Stefania, Alexander, Cameron, and García Fuentes, Marcos
- Abstract
Safe and efficient gene delivery vectors would enhance the prospects for polynucleotide-based therapies. Herein we describe a new approach towards structurally-optimized gene vector design based on the preparation of clickable poly(allylamino-phosphazene)s that can be converted to several cationic and anionic derivatives via thiol-ene addition. Simultaneous co-incubation of alkylamine- and alkylcarboxylate-poly(phosphazenes) with polynucleotides generated binary-polyelectrolyte nanoparticles. Screening of a series of these complexes for transfection in glioblastoma cells showed that the inclusion of 6-mercaptohexanoic acid substituted poly(phosphazene)s in the complexes resulted in 6-fold and 19-fold higher luciferase expression in U87MG cells and primary GBM1 cell- line, respectively. This effect was attributed to the specific ionization properties of this materials that improved polyplex intracellular trafficking. Transfection in 3D-spheroid models and subcutaneous xenograft U87MG tumors confirmed higher transgene expression for the binary cationic/anionic poly(phosphazene) complexes compared to the related polycation-pDNA complexes and to PEI- pDNA complexes. The data also indicated a notable capacity of the mixed complexes to deliver genes to the inner cores of tumor spheroids. Extension of this approach to siRNA delivery showed that the mixed poly(phosphazene) complexes were able to silence DYRK1A, a gene implicated in tumor initiation and progression, reducing U87MG cell renewal in vitro and delaying tumor growth in vivo.
- Published
- 2019
92. The effect of Transforming Growth Factor Beta family members on tumor initiating cells in primary liver cancer
- Author
-
D Castven, PR Galle, S Dooley, Hauke Lang, L Rodrigues, Jens U. Marquardt, S Pereira, Nmm Meindl-Beinker, and Peter P. Grimminger
- Subjects
biology ,business.industry ,Gastroenterology ,Cancer research ,biology.protein ,Medicine ,Transforming growth factor beta ,Primary liver cancer ,business ,Tumor Initiating Cells - Published
- 2016
- Full Text
- View/download PDF
93. Activation of tumor initiating cells during anti-angiogenic therapies promotes tumor progression and relapse formation in hepatocellular carcinoma
- Author
-
D Becker, Carolin Czauderna, Peter P. Grimminger, Hauke Lang, D Castven, MA Wörns, Jens U. Marquardt, Peter R. Galle, and Snorri S. Thorgeirsson
- Subjects
Tumor progression ,business.industry ,Hepatocellular carcinoma ,Anti angiogenic ,Immunology ,Gastroenterology ,Cancer research ,medicine ,medicine.disease ,business ,Tumor Initiating Cells - Published
- 2016
- Full Text
- View/download PDF
94. Microenvironment mediated alterations to metabolic pathways confer increased chemo-resistance in CD133+ tumor initiating cells
- Author
-
Olivia McGinn, Alice Nomura, Vikas Dudeja, Nivedita Arora, Patricia Dauer, Kaustav Majumdar, Ashok K. Saluja, Charles Uhlrich, Joseph J. Dalluge, Vineet Gupta, and Sulagna Banerjee
- Subjects
0301 basic medicine ,Glucose uptake ,Apoptosis ,Cell Separation ,Mice ,Tandem Mass Spectrometry ,Surgical oncology ,Tumor Microenvironment ,AC133 Antigen ,CD133 ,Chromatography, High Pressure Liquid ,Glucose Transporter Type 1 ,education.field_of_study ,tumor initiating cells ,ROS ,Immunohistochemistry ,Cell Hypoxia ,Mitochondria ,3. Good health ,Oncology ,Metabolome ,Neoplastic Stem Cells ,Metabolic Networks and Pathways ,Research Paper ,Population ,Antineoplastic Agents ,Mice, Transgenic ,Electron Transport Complex IV ,03 medical and health sciences ,Cell Line, Tumor ,Pancreatic cancer ,medicine ,Animals ,Humans ,education ,Electron Transport Complex I ,hypoxia ,business.industry ,Cancer ,Neoplasms, Experimental ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Pancreatic Neoplasms ,Glucose ,030104 developmental biology ,HIF1A ,Drug Resistance, Neoplasm ,Tumor progression ,Immunology ,Cancer research ,Neoplasm Recurrence, Local ,business ,metabolism - Abstract
// Alice Nomura 1, 2 , Patricia Dauer 1, 2 , Vineet Gupta 1, 2 , Olivia McGinn 1 , Nivedita Arora 1 , Kaustav Majumdar 1 , Charles Uhlrich III 1 , Joseph Dalluge 3 , Vikas Dudeja 1, 2 , Ashok Saluja 1, 2 , Sulagna Banerjee 1, 2 1 Division of Basic and Translational Research, Department of Surgery, University of Minnesota, Minneapolis, MN, USA 2 Division of Surgical Oncology, Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA 3 Department of Chemistry, Mass Spectrometry Laboratory, University of Minnesota, Minneapolis, MN, USA Correspondence to: Sulagna Banerjee, email: Sulagna.Banerjee@med.miami.edu Keywords: CD133, tumor initiating cells, metabolism, hypoxia, ROS Received: April 25, 2016 Accepted: July 10, 2016 Published: July 26, 2016 ABSTRACT Chemoresistance in pancreatic cancer has been attributed to tumor-initiating cells (TICs), a minor sub-population of tumor cells. However, the mechanism of chemo-resistance in these cells is still unclear. In the current study, immunohistochemical analysis of LSL-Kras G12D ; LSL-Trp53 R172H; PdxCre (KPC) murine tumors indicated that hypoxic regions developed through tumor progression. This hypoxic “niche” correlated with increased CD133 + population that had an increased HIF1A activity. Consistent with this observation, CD133 + cells had increased glucose uptake and activity of glycolytic pathway enzymes compared to CD133 − cells. Mass spectrometric analysis (UPLC-TQD) following metabolic labeling of CD133 + cells with [ 13 C]-U6 glucose confirmed this observation. Furthermore, although both populations had functionally active mitochondria, CD133 + cells had low mitochondrial complex I and complex IV activity and lesser accumulation of ROS in response to standard chemotherapeutic compounds like paclitaxel, 5FU and gemcitabine. CD133 + cells also showed increased resistance to all three chemotherapeutic compounds and treatment with Glut1 inhibitor (STF31) reversed this resistance, promoting apoptotic death in these cells similar to CD133 − cells. Our study indicates that the altered metabolic profile of CD133 + pancreatic TIC protects them against apoptosis, by reducing accumulation of ROS induced by standard chemotherapeutic agents, thereby confering chemoresistance. Since resistance to existing chemotherapy contributes to the poor prognosis in pancreatic cancer, our study paves the way for identifying novel therapeutic targets for managing chemoresistance and tumor recurrence in pancreatic cancer.
- Published
- 2016
- Full Text
- View/download PDF
95. Salmonella VNP20009-mediated RNA interference of ABCB5 moderated chemoresistance of melanoma stem cell and suppressed tumor growth more potently
- Author
-
Cheng Xiawei, Yuqiang Zhou, Xiaoxin Zhang, Zi-Chun Hua, Yueyang Lai, and Wenmin Cao
- Subjects
0301 basic medicine ,Small interfering RNA ,ATP Binding Cassette Transporter, Subfamily B ,Population ,Melanoma, Experimental ,Apoptosis ,Pharmacology ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Tumor Cells, Cultured ,medicine ,Salmonella VNP20009 ,Animals ,RNA, Small Interfering ,education ,Antineoplastic Agents, Alkylating ,Cyclophosphamide ,Cell Proliferation ,antitumor ,bacterial therapy ,education.field_of_study ,drug resistance ,tumor initiating cells ,Melanoma ,ABCB5 ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Bacterial vaccine ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Bacterial Vaccines ,ATP-Binding Cassette Transporters ,Drug Therapy, Combination ,Stem cell ,Research Paper - Abstract
Drug resistance remains an obstacle hindering the success of chemotherapy. Cancer stem cells (CSCs) have been recently found to confer resistance to chemotherapy. Therefore functional markers of CSCs should be discovered and specific therapies targeting these cells should be developed. In our investigation, a small population of B16F10 cells which was positive for ATP-binding cassette sub-family B member 5 (ABCB5) was isolated. This population displayed characteristics similar to those of CSCs and ABCB5 was identified to confer tumor growth and drug resistance in B16F10 cell line. Although targeting ABCB5 by small short interfering RNA delivered by VNP20009 failed to inhibit tumor growth, the combined treatment of VNP-shABCB5 and chemotherapy can act synergistically to delay tumor growth and enhance survival time in a primary B16F10 mice model. Results suggest that the combined treatment of VNP-shABCB5 and chemotherapy can improve the efficacy of chemotherapeutic drugs. Therefore, this combination therapy is of potential significance for melanoma treatment.
- Published
- 2016
- Full Text
- View/download PDF
96. Abstract 3582: Drugs targeting ovarian cancer tumor-initiating cells enhance oxidative stress and prevent disease recurrence
- Author
-
Christina M. Annunziata and Brittney S. Harrington
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Disease ,business ,Ovarian cancer ,medicine.disease ,medicine.disease_cause ,Oxidative stress ,Tumor Initiating Cells - Abstract
Ovarian cancer (OC) is a global health burden with the poorest 5-year survival rate of the gynecological malignancies. Disease recurrence is the major cause of morbidity and mortality of OC, likely driven by the survival of chemoresistant, self-renewing tumor-initiating cells (TICs). The TIC population in OC acquires features to survive cellular stress and avoid apoptosis but these can also be targeted as vulnerabilities specific to the TIC population of OC to prevent recurrence. To reveal these vulnerabilities, a high-throughput drug screen was performed to identify drugs that showed increased efficacy against OC cells grown in TIC-enriching spheroid (TES) conditions compared to adherently grown cells. Four drugs were selected for further investigation. Disulfiram, Bardoxolone Methyl, Elesclomol and Salinomycin were tested in vitro to confirm the effects on viability of OC cells in adherent or TES conditions. The drugs' efficacy against sphere formation and expression of markers of stemness as high ALDH activity and CD133 expression (ALDH+CD133+) as single agents or in combination with carboplatin was also investigated in vitro. Disulfiram showed the greatest efficacy against spheroid viability and decreased the ALDH+CD133+ population of several OC cell lines. This effect was enhanced when combined with carboplatin. Bardoxolone Methyl, Elesclomol and Salinomycin showed similar efficacy against cell viability in OC cells grown adherently or in TES conditions; Elesclomol significantly inhibited the ALDH+CD133+ population of the OC cells which was again enhanced in combination with carboplatin. RNAseq analysis revealed a strong enrichment of genes involved in oxidative phosphorylation and reactive oxygen species (ROS) pathways in TES conditions compared to adherent conditions. The drugs were investigated for their ability to promote oxidative stress and OC cells were measured for accumulation of intracellular ROS and mitochondrial superoxide after exposure to the drugs. OC cells grown in TES conditions had higher basal intracellular ROS levels than adherently grown cells and the level of ROS was significantly enhanced in TES cells treated with Elesclomol or Disulfiram but no differences were seen in adherently grown cells. Salinomycin and Bardoxolone Methyl increased mitochondrial superoxide accumulation in some OC cell lines under TES conditions. In a model of OC relapse in vitro, Disulfiram and Elesclomol following carboplatin treatment significantly increased cell death compared to carboplatin alone. Disulfiram and Salinomycin were incorporated into a mouse model to test their ability to prevent OC relapse after chemotherapy in vivo. These results demonstrate that targeting key pathways mediating increased oxidative stress in TICs can eliminate this population and provide a means to prevent OC recurrence. Citation Format: Brittney S. Harrington, Christina M. Annunziata. Drugs targeting ovarian cancer tumor-initiating cells enhance oxidative stress and prevent disease recurrence [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3582.
- Published
- 2020
- Full Text
- View/download PDF
97. New quantitative approach reveals heterogeneity in mitochondrial structure-function relations in tumor initiating cells
- Author
-
Anita B. Hjelmeland, Avik Mukherjee, Victor M. Darley-Usmar, Kasturi Mitra, Brian Spurlock, McKenzie E. Foxall, Priyanka Gupta, Malay Kumar Basu, and Danitra Parker
- Subjects
Fission ,Mitochondrion ,Biology ,Mitochondrial Dynamics ,Cell Line ,Tumor Initiating Cells ,Mitochondrial Proteins ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,Animals ,Humans ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,Fusion ,Microscopy, Confocal ,Energetics ,Cell Biology ,Tools and Resources ,Mitochondria ,Mitochondrial structure ,First person ,Neoplastic Stem Cells ,Biophysics ,Female ,Energy Metabolism ,Oxidation-Reduction ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Steady-state mitochondrial structure or morphology is primarily maintained by a balance of opposing fission and fusion events between individual mitochondria, which is collectively referred to as mitochondrial dynamics. The details of the bidirectional relationship between the status of mitochondrial dynamics (structure) and energetics (function) require methods to integrate these mitochondrial aspects. To study the quantitative relationship between the status of mitochondrial dynamics (fission, fusion, matrix continuity and diameter) and energetics (ATP and redox), we have developed an analytical approach called mito-SinCe(2). After validating and providing proof of principle, we applied mito-SinCe(2) on ovarian tumor-initiating cells (ovTICs). Mito-SinCe(2) analyses led to the hypothesis that mitochondria-dependent ovTICs interconvert between three states, that have distinct relationships between mitochondrial energetics and dynamics. Interestingly, fusion and ATP increase linearly with each other only once a certain level of fusion is attained. Moreover, mitochondrial dynamics status changes linearly with ATP or with redox, but not simultaneously with both. Furthermore, mito-SinCe(2) analyses can potentially predict new quantitative features of the opposing fission versus fusion relationship and classify cells into functional classes based on their mito-SinCe(2) states. This article has an associated First Person interview with the first author of the paper.
- Published
- 2019
- Full Text
- View/download PDF
98. Structure-optimized interpolymer polyphosphazene complexes for effective gene delivery to glioblastoma
- Author
-
Wei-Hsin Hsu, Ruman Rahman, Esther Gómez-Ibarlucea, Marcos Garcia-Fuentes, Noemi Csaba, Delyan P. Ivanov, Anna M. Grabowska, Cameron Alexander, Pilar Sánchez-Gómez, and Universidade de Santiago de Compostela. Centro de Investigación en Medicina Molecular e Enfermidades Crónicas
- Subjects
siRNA delivery ,Pharmaceutical Science ,Medicine (miscellaneous) ,02 engineering and technology ,Gene delivery ,Tumor initiating cells ,010402 general chemistry ,01 natural sciences ,Tumor Initiating Cells ,medicine ,Pharmacology (medical) ,Polyphosphazene ,Genetics (clinical) ,Pharmacology ,Polymeric gene delivery ,Chemistry ,Click chemistry ,Biochemistry (medical) ,021001 nanoscience & nanotechnology ,medicine.disease ,0104 chemical sciences ,Cancer research ,Poly(phosphazene)s ,0210 nano-technology ,Glioblastoma - Abstract
This is the peer reviewed version of the following article: Hsu, W.‐H., Sánchez‐Gómez, P., Gomez‐Ibarlucea, E., Ivanov, D.P., Rahman, R., Grabowska, A.M., Csaba, N., Alexander, C. and Garcia‐Fuentes, M. (2019), Structure‐Optimized Interpolymer Polyphosphazene Complexes for Effective Gene Delivery against Glioblastoma. Adv. Therap., 2: 1800126. doi:10.1002/adtp.201800126, which has been published in final form at https://doi.org/10.1002/adtp.201800126. This article may be used for non-commercial purposes in accordance with Wiley Terms and Conditions for Use of Self-Archived Versions Safe and efficient gene delivery vectors would enhance the prospects for polynucleotide-based therapies. Herein we describe a new approach towards structurally-optimized gene vector design based on the preparation of clickable poly(allylamino-phosphazene)s that can be converted to several cationic and anionic derivatives via thiol-ene addition. Simultaneous co-incubation of alkylamine- and alkylcarboxylate-poly(phosphazenes) with polynucleotides generated binary-polyelectrolyte nanoparticles. Screening of a series of these complexes for transfection in glioblastoma cells showed that the inclusion of 6-mercaptohexanoic acid substituted poly(phosphazene)s in the complexes resulted in 6-fold and 19-fold higher luciferase expression in U87MG cells and primary GBM1 cell- line, respectively. This effect was attributed to the specific ionization properties of this materials that improved polyplex intracellular trafficking. Transfection in 3D-spheroid models and subcutaneous xenograft U87MG tumors confirmed higher transgene expression for the binary cationic/anionic poly(phosphazene) complexes compared to the related polycation-pDNA complexes and to PEI- pDNA complexes. The data also indicated a notable capacity of the mixed complexes to deliver genes to the inner cores of tumor spheroids. Extension of this approach to siRNA delivery showed that the mixed poly(phosphazene) complexes were able to silence DYRK1A, a gene implicated in tumor initiation and progression, reducing U87MG cell renewal in vitro and delaying tumor growth in vivo. This work has been funded by Ministerio de Economía y Competitividad (MINECO-RETOS, Grant MAT2017-84361-R, Feder Funds) and Xunta de Galicia (Grupos de Referencia Competitiva, Feder Funds) to MGF and Engineering and Physical Sciences Research Council Grants EP/N006615/1, EP/ N03371X/1 to CA. WHH was a recipient of a contract from EU Erasmus Mundus NanoFar joint doctorate program SI
- Published
- 2019
99. Abstract B13: Immune cells, tumor-initiating cells, and drug sensitivity in claudin-low TNBC: A delicate balance
- Author
-
Swarnima Singh, Jeffrey M. Rosen, Se-Jin Kim, and Xiang Zhang
- Subjects
Drug ,Cancer Research ,Immune system ,Chemistry ,media_common.quotation_subject ,Immunology ,Cancer research ,Sensitivity (control systems) ,Claudin-Low ,media_common ,Balance (ability) ,Tumor Initiating Cells - Abstract
Background: Breast cancer is a heterogeneous disease. Based on gene expression profiling, it can be classified into at least 5 subtypes. They are luminal A and B, normal breast-like, Her2-positive, basal-like, and newly described claudin-low. Among all these subtypes, basal-like and claudin-low tumors have relatively poor prognosis. Claudin-low tumors are characterized by the low expression of cell-cell junction proteins like claudin 3 and E-cadherin along with an enrichment of EMT markers such as SNAI1 and SNAI2 and high levels of mesenchymal markers such as vimentin. Claudin-low tumors are generally more aggressive and the rate of response to primary chemotherapy is around 32%. Initial data have shown that p-53 null claudin-low tumors are sensitive to cyclophosphamide treatment. Cyclophosphamide (CTX) is a chemotherapy drug that has been used in lymphoma, leukemia and some solid tumors. At low and continuous doses, it shows immunostimulatory properties. The overall goal of this proposal is to test the therapeutic response of claudin-low tumors to cyclophosphamide treatment and define the functional role of the immune system in a mouse model of claudin-low breast cancer. Preliminary data have shown that tumor cells in a p53 null claudin-low murine model (T12) possess tumor-initiating abilities and have STAT3 activation. Initial data in claudin-low T12 tumors also suggest that CTX induces CD4+ and CD8+ T-cell homing to the primary tumor. Additionally, wild-type Balb/c mice implanted with T12 tumors respond favorably to CTX treatment as compared to T cell-deficient NSG mice. Our studies have shown that treatment-resistant claudin-low tumors have a high tumor-associated macrophage (TAM) infiltrate. Tumors with poor response to CTX show higher levels of vimentin staining as compared to responsive tumors. Depletion of specific T-cell subsets to determine their functional importance in the response to CTX shows that while CD8+ T-cell depletion does not have a significant effect on treatment response, depletion of CD4+ T cells results in impaired tumor response to CTX treatment. Additionally, TAMs increase upon CD4+ T-cell depletion. We hypothesize that CTX treatment can sensitize T12 primary tumors to CD4+ T-cell mediated apoptosis. We further hypothesize that TAMS can suppress T-cell function and this, along with changes in the tumor EMT status, can promote chemoresistance. TAMs can also interact with tumor-initiating cells (TIC) within claudin-low tumors via the STAT3 pathway to promote survival and proliferation of chemotherapy resistant TICs. Furthermore, depleting immunosuppressive cells such as TAMs may provide us with an additional therapeutic modality for chemotherapy-resistant breast cancer in combination with checkpoint immunotherapy. Reference: 1. Prat A et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 2010;12:R68. Citation Format: Swarnima Singh, Se-Jin Kim, Xiang Zhang, Jeffrey Rosen. Immune cells, tumor-initiating cells, and drug sensitivity in claudin-low TNBC: A delicate balance [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B13.
- Published
- 2020
- Full Text
- View/download PDF
100. Tumor-initiating cells in malignant brain tumors
- Author
-
Prakash Rath
- Subjects
business.industry ,Cancer research ,Medicine ,business ,Tumor Initiating Cells - Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.