Your browser doesn't support javascript.
loading
Growth factor independence underpins a paroxysmal, aggressive Wnt5aHigh/EphA2Low phenotype in glioblastoma stem cells, conducive to experimental combinatorial therapy.
Trivieri, Nadia; Visioli, Alberto; Mencarelli, Gandino; Cariglia, Maria Grazia; Marongiu, Laura; Pracella, Riccardo; Giani, Fabrizio; Soriano, Amata Amy; Barile, Chiara; Cajola, Laura; Copetti, Massimiliano; Palumbo, Orazio; Legnani, Federico; DiMeco, Francesco; Gorgoglione, Leonardo; Vescovi, Angelo L; Binda, Elena.
Afiliação
  • Trivieri N; Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, 71013, San Giovanni Rotondo, FG, Italy.
  • Visioli A; StemGen SpA, Milan, Italy.
  • Mencarelli G; Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, 71013, San Giovanni Rotondo, FG, Italy.
  • Cariglia MG; Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, 71013, San Giovanni Rotondo, FG, Italy.
  • Marongiu L; Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.
  • Pracella R; Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, 71013, San Giovanni Rotondo, FG, Italy.
  • Giani F; StemGen SpA, Milan, Italy.
  • Soriano AA; Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, 71013, San Giovanni Rotondo, FG, Italy.
  • Barile C; Cancer Stem Cells Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapeutics (ISBReMIT), IRCSS Casa Sollievo della Sofferenza, Opera di San Pio da Pietrelcina, 71013, San Giovanni Rotondo, FG, Italy.
  • Cajola L; StemGen SpA, Milan, Italy.
  • Copetti M; Biostatistics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
  • Palumbo O; Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
  • Legnani F; Department of Neurosurgery, National Neurologic Institute IRCCS C. Besta, Milan, Italy.
  • DiMeco F; Department of Neurosurgery, National Neurologic Institute IRCCS C. Besta, Milan, Italy.
  • Gorgoglione L; Department of Neurosurgery, John Hopkins University, Baltimore, MD, USA.
  • Vescovi AL; Neurosurgery Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
  • Binda E; Scientific Directorate, IRCCS Casa Sollievo della Sofferenza, FG, San Giovanni Rotondo, Italy. vescovia@gmail.com.
J Exp Clin Cancer Res ; 41(1): 139, 2022 Apr 12.
Article em En | MEDLINE | ID: mdl-35414102
ABSTRACT

BACKGROUND:

Glioblastoma multiforme (GBM) is an incurable tumor, with a median survival rate of only 14-15 months. Along with heterogeneity and unregulated growth, a central matter in dealing with GBMs is cell invasiveness. Thus, improving prognosis requires finding new agents to inhibit key multiple pathways, even simultaneously. A subset of GBM stem-like cells (GSCs) may account for tumorigenicity, representing, through their pathways, the proper cellular target in the therapeutics of glioblastomas. GSCs cells are routinely enriched and expanded due to continuous exposure to specific growth factors, which might alter some of their intrinsic characteristic and hide therapeutically relevant traits.

METHODS:

By removing exogenous growth factors stimulation, here we isolated and characterized a subset of GSCs with a "mitogen-independent" phenotype (I-GSCs) from patient's tumor specimens. Differential side-by-side comparative functional and molecular analyses were performed either in vitro or in vivo on these cells versus their classical growth factor (GF)-dependent counterpart (D-GSCs) as well as their tissue of origin. This was performed to pinpoint the inherent GSCs' critical regulators, with particular emphasis on those involved in spreading and tumorigenic potential. Transcriptomic fingerprints were pointed out by ANOVA with Benjamini-Hochberg False Discovery Rate (FDR) and association of copy number alterations or somatic mutations was determined by comparing each subgroup with a two-tailed Fisher's exact test. The combined effects of interacting in vitro and in vivo with two emerging GSCs' key regulators, such as Wnt5a and EphA2, were then predicted under in vivo experimental settings that are conducive to clinical applications. In vivo comparisons were carried out in mouse-human xenografts GBM model by a hierarchical linear model for repeated measurements and Dunnett's multiple comparison test with the distribution of survival compared by Kaplan-Meier method.

RESULTS:

Here, we assessed that a subset of GSCs from high-grade gliomas is self-sufficient in the activation of regulatory growth signaling. Furthermore, while constitutively present within the same GBM tissue, these GF-independent GSCs cells were endowed with a distinctive functional and molecular repertoire, defined by highly aggressive Wnt5aHigh/EphA2Low profile, as opposed to Wnt5aLow/EphA2High expression in sibling D-GSCs. Regardless of their GBM subtype of origin, I-GSCs, are endowed with a raised in vivo tumorigenic potential than matched D-GSCs, which were fast-growing ex-vivo but less lethal and invasive in vivo. Also, the malignant I-GSCs' transcriptomic fingerprint faithfully mirrored the original tumor, bringing into evidence key regulators of invasiveness, angiogenesis and immuno-modulators, which became candidates for glioma diagnostic/prognostic markers and therapeutic targets. Particularly, simultaneously counteracting the activity of the tissue invasive mediator Wnt5a and EphA2 tyrosine kinase receptor addictively hindered GSCs' tumorigenic and invasive ability, thus increasing survival.

CONCLUSION:

We show how the preservation of a mitogen-independent phenotype in GSCs plays a central role in determining the exacerbated tumorigenic and high mobility features distinctive of GBM. The exploitation of the I-GSCs' peculiar features shown here offers new ways to identify novel, GSCs-specific effectors, whose modulation can be used in order to identify novel, potential molecular therapeutic targets. Furthermore, we show how the combined use of PepA, the anti-Wnt5a drug, and of ephrinA1-Fc to can hinder GSCs' lethality in a clinically relevant xenogeneic in vivo model thus being conducive to perspective, novel combinatorial clinical application.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Encefálicas / Glioblastoma Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: J Exp Clin Cancer Res Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Itália

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias Encefálicas / Glioblastoma Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: J Exp Clin Cancer Res Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Itália