Molecular Mechanisms and Clinical Treatments of Neurofibromatosis Type 1 Related Tumors

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cells of the Nervous System".

Deadline for manuscript submissions: 31 August 2024 | Viewed by 1000

Special Issue Editors


E-Mail Website
Guest Editor
Department of Biomedical Sciences, Università degli Studi di Padova, 35100 Padua, Italy
Interests: mitochondrial pathophysiology; tumor metabolism; molecular chaperones

E-Mail Website
Guest Editor
Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, 35131 Padova, Italy
Interests: neurofibromatosis type 1; signal transduction; molecular oncology

Special Issue Information

Dear Colleagues,

Neurofibromatosis type 1 (NF1) is a tumor-predisposing genetic syndrome where inactivating mutations in the NF1 gene elicit Ras signaling dysregulation and prime the onset of benign tumors in central and peripheral nerves. Ras hyperactivation drives tumor growth by orchestrating changes in biochemical and metabolic circuits of transformed cells with an altered network of interactions in the tumor microenvironment. The dysregulated crosstalk between tumor cells and environmental cues prompts further (epi)genetic lesions and the transition towards malignancy, as exemplified by the evolution from neurofibromas, i.e., benign Schwann cell tumors, to malignant peripheral nerve sheath tumors. Hence, NF1 constitutes an extraordinary model that is used to study progression from early tumor stages to aggressive neoplastic forms. In spite of the huge advances made in the comprehension of this process, translation to effective therapies remains limited, and the clinical settings of NF1 patients warrant major improvements.

This Special Issue aims to draw a comprehensive picture of the biological determinants that support the pro-tumor activity of Ras signaling along the stepwise transformation towards malignancy. The integration of this information into the clinical management of NF1 patients can trailblaze more advanced and effective therapeutic options, and can establish the basis for studying the complex interactions that induce neoplastic progression in a broader set of Ras-related tumor models.

Dr. Andrea Rasola
Dr. Federica Chiara
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • neurofibromatosis type 1
  • rare tumors
  • genetic syndrome
  • plexiform neurofibroma
  • malignant peripheral nerve sheath tumor
  • tumor suppressor
  • Ras signalling
  • tumor microenvironment
  • anti-neoplastic therapy
  • neoplastic transformation

Published Papers (1 paper)

Order results
Result details
Select all
Export citation of selected articles as:

Research

11 pages, 4595 KiB  
Communication
The Use of Hexokinase 2-Displacing Peptides as an Anti-Neoplastic Approach for Malignant Peripheral Nerve Sheath Tumors
by Francesco Ciscato, Ionica Masgras, Alessandro Gori, Marco Fantuz, Greta Bergamaschi, Denis Komarov, Martina La Spina, Shiva Ghasemi-Firouzabadi, Marco Pizzi, Angelo Paolo Dei Tos, Federica Chiara, Alessandro Carrer and Andrea Rasola
Cells 2024, 13(13), 1162; https://doi.org/10.3390/cells13131162 - 8 Jul 2024
Viewed by 573
Abstract
Malignant Peripheral Nerve Sheath Tumors (MPNSTs) are aggressive sarcomas that can arise both sporadically and in patients with the genetic syndrome Neurofibromatosis type 1 (NF1). Prognosis is dismal, as large dimensions, risk of relapse, and anatomical localization make surgery poorly effective, and no [...] Read more.
Malignant Peripheral Nerve Sheath Tumors (MPNSTs) are aggressive sarcomas that can arise both sporadically and in patients with the genetic syndrome Neurofibromatosis type 1 (NF1). Prognosis is dismal, as large dimensions, risk of relapse, and anatomical localization make surgery poorly effective, and no therapy is known. Hence, the identification of MPNST molecular features that could be hit in an efficient and selective way is mandatory to envision treatment options. Here, we find that MPNSTs express high levels of the glycolytic enzyme Hexokinase 2 (HK2), which is known to shield cancer cells from noxious stimuli when it localizes at MAMs (mitochondria-associated membranes), contact sites between mitochondria and endoplasmic reticulum. A HK2-targeting peptide that dislodges HK2 from MAMs rapidly induces a massive death of MPNST cells. After identifying different matrix metalloproteases (MMPs) expressed in the MPNST microenvironment, we have designed HK2-targeting peptide variants that harbor cleavage sites for these MMPs, making such peptides activatable in the proximity of cancer cells. We find that the peptide carrying the MMP2/9 cleavage site is the most effective, both in inhibiting the in vitro tumorigenicity of MPNST cells and in hampering their growth in mice. Our data indicate that detaching HK2 from MAMs could pave the way for a novel anti-MPNST therapeutic strategy, which could be flexibly adapted to the protease expression features of the tumor microenvironment. Full article
Show Figures

Figure 1

Back to TopTop