ijms-logo

Journal Browser

Journal Browser

Lung Cancer: From Molecular Mechanisms to Novel Therapeutics

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Oncology".

Deadline for manuscript submissions: 20 June 2024 | Viewed by 11024

Special Issue Editors


E-Mail Website
Guest Editor
1. Medical Oncology Unit, AOU Policlinico "G.Martino", 98125 Messina, Italy
2. Department of Human Pathology “G. Barresi”, University of Messina, 98122 Messina, Italy
Interests: lung cancer; targeted therapy; immunotherapy; personalized medicine; biomarkers
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
1. Medical Oncology 2, Istituto Oncologico Veneto IRCCS, 35128 Padova, Italy
2. Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
Interests: lung cancer; mesothelioma; targeted therapy; immunotherapy; biomarkers

Special Issue Information

Dear Colleagues,

Lung cancer remains the most significant cause of cancer-related deaths worldwide. Both molecular-targeted therapy and immunotherapy have revolutionized the diagnostic and therapeutic management of this heterogenous disease, which includes different histologic and molecular subtypes. The number of actionable oncogenic drivers continues to expand, in parallel with the development of novel targeted agents. On the other hand, several immune checkpoint inhibitors, as single agents or in combinatorial regimens, have been approved in metastatic non-small-cell lung cancer without targetable molecular alterations because of the demonstration of significant clinical efficacy and manageable toxicity profiles. Their development has been shifted in the early stages, with promising results. However, despite significant therapeutic advances, several challenges still remain, including the need for additional reliable biomarkers to select patients for immunotherapy, and a better understanding of the complex biologic mechanisms leading to therapeutic resistance. This Special Issue will focus on: a) identification of new druggable driver gene alterations in lung cancer; b) development of new drugs and combination therapies targeting specific molecular pathways; c) current knowledge on primary and secondary mechanisms of resistance to targeted therapies or immunotherapy; d) current and emerging techniques to identify predictive biomarkers in tissue and liquid biopsy.

Dr. Mariacarmela Santarpia
Dr. Giulia Pasello
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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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

  • lung cancer
  • oncogene
  • targeted therapy
  • immunotherapy
  • predictive biomarkers
  • liquid biopsy

Published Papers (7 papers)

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

Research

Jump to: Review, Other

18 pages, 9873 KiB  
Article
The Roles and Regulatory Mechanisms of Tight Junction Protein Cingulin and Transcription Factor Forkhead Box Protein O1 in Human Lung Adenocarcinoma A549 Cells and Normal Lung Epithelial Cells
by Daichi Ishii, Yuma Shindo, Wataru Arai, Takumi Konno, Takayuki Kohno, Kazuya Honda, Masahiro Miyajima, Atsushi Watanabe and Takashi Kojima
Int. J. Mol. Sci. 2024, 25(3), 1411; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25031411 - 24 Jan 2024
Viewed by 853
Abstract
Tight junction (TJ) protein cingulin (CGN) and transcription factor forkhead box protein O1 (FOXO1) contribute to the development of various cancers. Histone deacetylase (HDAC) inhibitors have a potential therapeutic role for some cancers. HDAC inhibitors affect the expression of both CGN and FOXO1. [...] Read more.
Tight junction (TJ) protein cingulin (CGN) and transcription factor forkhead box protein O1 (FOXO1) contribute to the development of various cancers. Histone deacetylase (HDAC) inhibitors have a potential therapeutic role for some cancers. HDAC inhibitors affect the expression of both CGN and FOXO1. However, the roles and regulatory mechanisms of CGN and FOXO1 are unknown in non-small cell lung cancer (NSCLC) and normal human lung epithelial (HLE) cells. In the present study, to investigate the effects of CGN and FOXO1 on the malignancy of NSCLC, we used A549 cells as human lung adenocarcinoma and primary human lung epithelial (HLE) cells as normal lung tissues and performed the knockdown of CGN and FOXO1 by siRNAs. Furthermore, to investigate the detailed mechanisms in the antitumor effects of HDAC inhibitors for NSCLC via CGN and FOXO1, A549 cells and HLE cells were treated with the HDAC inhibitors trichostatin A (TSA) and Quisinostat (JNJ-2648158). In A549 cells, the knockdown of CGN increased bicellular TJ protein claudin-2 (CLDN-2) via mitogen-activated protein kinase/adenosine monophosphate-activated protein kinase (MAPK/AMPK) pathways and induced cell migration, while the knockdown of FOXO1 increased claudin-4 (CLDN-4), decreased CGN, and induced cell proliferation. The knockdown of CGN and FOXO1 induced cell metabolism in A549 cells. TSA and Quisinostat increased CGN and tricellular TJ protein angulin-1/lipolysis-stimulated lipoprotein receptor (LSR) in A549. In normal HLE cells, the knockdown of CGN and FOXO1 increased CLDN-4, while HDAC inhibitors increased CGN and CLDN-4. In conclusion, the knockdown of CGN via FOXO1 contributes to the malignancy of NSCLC. Both HDAC inhibitors, TSA and Quisinostat, may have potential for use in therapy for lung adenocarcinoma via changes in the expression of CGN and FOXO1. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
Show Figures

Figure 1

25 pages, 5296 KiB  
Article
Real-World Data on Combined EGFR-TKI and Crizotinib Treatment for Acquired and De Novo MET Amplification in Patients with Metastatic EGFR-Mutated NSCLC
by Edyta M. Urbanska, Morten Grauslund, Peter R. Koffeldt, Sarah L. B. Truelsen, Johan O. Löfgren, Junia C. Costa, Linea C. Melchior, Jens B. Sørensen and Eric Santoni-Rugiu
Int. J. Mol. Sci. 2023, 24(17), 13077; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241713077 - 23 Aug 2023
Viewed by 1499
Abstract
Amplification of the mesenchymal epithelial transition (MET) gene is a mechanism of acquired resistance to epidermal growth factor receptor (EGFR)-tyrosine-kinase-inhibitors (TKIs) in over 20% of patients with advanced EGFR-mutated (EGFRm+) non-small lung cancer (NSCLC). However, it may also [...] Read more.
Amplification of the mesenchymal epithelial transition (MET) gene is a mechanism of acquired resistance to epidermal growth factor receptor (EGFR)-tyrosine-kinase-inhibitors (TKIs) in over 20% of patients with advanced EGFR-mutated (EGFRm+) non-small lung cancer (NSCLC). However, it may also occur de novo in 2–8% of EGFRm+ NSCLC cases as a potential mechanism of intrinsic resistance. These patients represent a group with unmet needs, since there is no standard therapy currently approved. Several new MET inhibitors are being investigated in clinical trials, but the results are awaited. Meanwhile, as an alternative strategy, combinations of EGFR-TKIs with the MET/ALK/ROS1-TKI Crizotinib may be used in this setting, despite this use is principally off-label. Thus, we studied five of these MET amplified cases receiving EGFR-TKI and Crizotinib doublet after progression on EGFR-TKI treatment to assess the benefits and challenges related to this combination and the possible occurrence of genomic and phenotypic co-alterations. Furthermore, we compared our cases with other real-world reports on Crizotinib/EGFR-TKI combinations, which appeared effective, especially in patients with high-level MET amplification. Yet, we observed that the co-occurrence of other genomic and phenotypical alterations may affect the response to combined EGFR-TKI and Crizotinib. Finally, given the heterogeneity of MET amplification, the diagnostic methods for assessing it may be discrepant. In this respect, we observed that for optimal detection, immunohistochemistry, fluorescence in situ hybridization, and next-generation sequencing should be used together, as these methods possess different sensitivities and complement each other in characterizing MET amplification. Additionally, we addressed the issue of managing EGFR-mutated NSCLC patients with de novo MET amplification causing primary EGFR-TKI resistance. We conclude that, while data from clinical trials with new MET inhibitors are still pending, adding Crizotinib to EGFR-TKI in NSCLC patients acquiring MET amplification at progression on EGFR-TKI monotherapy is a reasonable approach, with a progression-free survival of 3–19 months. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
Show Figures

Figure 1

Review

Jump to: Research, Other

15 pages, 618 KiB  
Review
Treatment of Thoracic SMARCA4-Deficient Undifferentiated Tumors: Where We Are and Where We Will Go
by Vito Longo, Annamaria Catino, Michele Montrone, Elisabetta Sara Montagna, Francesco Pesola, Ilaria Marech, Pamela Pizzutilo, Annalisa Nardone, Antonella Perrone, Monica Gesualdo and Domenico Galetta
Int. J. Mol. Sci. 2024, 25(6), 3237; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25063237 - 13 Mar 2024
Viewed by 987
Abstract
Recently, the fifth edition of the WHO classification recognized the thoracic SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) as a separate entity from conventional non-small cell lung cancer with SMARCA4 deficiency because of the different clinicopathological characteristics of these two diseases. SMARCA4-UT mainly occurs in [...] Read more.
Recently, the fifth edition of the WHO classification recognized the thoracic SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) as a separate entity from conventional non-small cell lung cancer with SMARCA4 deficiency because of the different clinicopathological characteristics of these two diseases. SMARCA4-UT mainly occurs in young to middle-aged adults and involves a large mass compressing the tissues surrounding the mediastinum and lung parenchyma. Unfortunately, SMARCA4-UT shows a high probability of recurrence after upfront surgery as well as radiotherapy resistance; moreover, chemotherapy has low efficacy. Moreover, given the recent classification of SMARCA4-UT, no data concerning specific clinical trials are currently available. However, several case reports show immunotherapy efficacy in patients with this disease not only in a metastatic setting but also in a neoadjuvant manner, supporting the development of clinical trials. In addition, preclinical data and initial clinical experiences suggest that inhibiting pathways such as CDK4/6, AURKA, ATR, and EZH2 may be a promising therapeutic approach to SMARCA4-UT. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
Show Figures

Figure 1

26 pages, 1031 KiB  
Review
Targeting MET in Non-Small Cell Lung Cancer (NSCLC): A New Old Story?
by Calogera Claudia Spagnolo, Giuliana Ciappina, Elisa Giovannetti, Andrea Squeri, Barbara Granata, Chiara Lazzari, Giulia Pretelli, Giulia Pasello and Mariacarmela Santarpia
Int. J. Mol. Sci. 2023, 24(12), 10119; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241210119 - 14 Jun 2023
Cited by 3 | Viewed by 2321
Abstract
In recent years, we have seen the development and approval for clinical use of an increasing number of therapeutic agents against actionable oncogenic drivers in metastatic non-small cell lung cancer (NSCLC). Among them, selective inhibitors, including tyrosine kinase inhibitors (TKIs) and monoclonal antibodies [...] Read more.
In recent years, we have seen the development and approval for clinical use of an increasing number of therapeutic agents against actionable oncogenic drivers in metastatic non-small cell lung cancer (NSCLC). Among them, selective inhibitors, including tyrosine kinase inhibitors (TKIs) and monoclonal antibodies targeting the mesenchymal–epithelial transition (MET) receptor, have been studied in patients with advanced NSCLC with MET deregulation, primarily due to exon 14 skipping mutations or MET amplification. Some MET TKIs, including capmatinib and tepotinib, have proven to be highly effective in this molecularly defined subgroup of patients and are already approved for clinical use. Other similar agents are being tested in early-stage clinical trials with promising antitumor activity. The purpose of this review is to provide an overview of MET signaling pathways, MET oncogenic alterations primarily focusing on exon 14 skipping mutations, and the laboratory techniques used to detect MET alterations. Furthermore, we will summarize the currently available clinical data and ongoing studies on MET inhibitors, as well as the mechanisms of resistance to MET TKIs and new potential strategies, including combinatorial approaches, to improve the clinical outcomes of MET exon 14-altered NSCLC patients. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
Show Figures

Figure 1

19 pages, 303 KiB  
Review
Targeted Toxicities: Protocols for Monitoring the Adverse Events of Targeted Therapies Used in the Treatment of Non-Small Cell Lung Cancer
by Jacobi B. Hines, Benjamin Bowar, Emma Levine, Alessandra Esposito, Marina C. Garassino and Christine M. Bestvina
Int. J. Mol. Sci. 2023, 24(11), 9429; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24119429 - 29 May 2023
Cited by 2 | Viewed by 1553
Abstract
Targeted therapies have revolutionized the treatment for many patients with non-small cell lung cancer (NSCLC). Multiple new oral targeted therapies have been approved in the last decade; however, their overall efficacy may be reduced by poor adherence, treatment interruptions, or dose reductions due [...] Read more.
Targeted therapies have revolutionized the treatment for many patients with non-small cell lung cancer (NSCLC). Multiple new oral targeted therapies have been approved in the last decade; however, their overall efficacy may be reduced by poor adherence, treatment interruptions, or dose reductions due to adverse events. Most institutions lack standard monitoring protocols for toxicities from these targeted agents. This review describes important adverse events observed in clinical trials and reported by the U.S. Food and Drug Administration for both currently approved and upcoming promising therapies in the treatment of NSCLC. These agents cause a range of toxicities, including dermatologic, gastroenteric, pulmonary, and cardiac toxicities. This review proposes protocols for routine monitoring of these adverse events, both prior to initiation of therapy and while on treatment. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
21 pages, 2199 KiB  
Review
Overview on Therapeutic Options in Uncommon EGFR Mutant Non-Small Cell Lung Cancer (NSCLC): New Lights for an Unmet Medical Need
by Giulia Pretelli, Calogera Claudia Spagnolo, Giuliana Ciappina, Mariacarmela Santarpia and Giulia Pasello
Int. J. Mol. Sci. 2023, 24(10), 8878; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24108878 - 17 May 2023
Cited by 2 | Viewed by 2071
Abstract
The majority of epidermal growth factor receptor (EGFR) mutations (85–90%) are exon 19 deletions and L858R point mutations of exon 21, characterized by high sensitivity to EGFR-tyrosine kinase inhibitors (TKIs). Less is known about uncommon mutations (10–15% of EGFR mutations). The predominant mutation [...] Read more.
The majority of epidermal growth factor receptor (EGFR) mutations (85–90%) are exon 19 deletions and L858R point mutations of exon 21, characterized by high sensitivity to EGFR-tyrosine kinase inhibitors (TKIs). Less is known about uncommon mutations (10–15% of EGFR mutations). The predominant mutation types in this category include exon 18 point mutations, exon 21 L861X, exon 20 insertions, and exon 20 S768I. This group shows a heterogeneous prevalence, partly due to different testing methods and to the presence of compound mutations, which in some cases can lead to shorter overall survival and different sensitivity to different TKIs compared to simple mutations. Additionally, EGFR-TKI sensitivity may also vary depending on the specific mutation and the tertiary structure of the protein. The best strategy remains uncertain, and the data of EGFR-TKIs efficacy are based on few prospective and some retrospective series. Newer investigational agents are still under study, and there are no other approved specific treatments targeting uncommon EGFR mutations. Defining the best treatment option for this patient population remains an unmet medical need. The objective of this review is to evaluate existing data on the outcomes, epidemiology, and clinical characteristics of lung cancer patients with rare EGFR mutations, with a focus on intracranial activity and response to immunotherapy. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
Show Figures

Figure 1

Other

Jump to: Research, Review

14 pages, 1723 KiB  
Brief Report
Membrane Localization and Phosphorylation of Indoleamine 2,3-Dioxygenase 2 (IDO2) in A549 Human Lung Adenocarcinoma Cells: First Steps in Exploring Its Signaling Function
by Chiara Suvieri, Francesca De Marchis, Martina Mandarano, Sara Ambrosino, Sofia Rossini, Giada Mondanelli, Marco Gargaro, Eleonora Panfili, Ciriana Orabona, Maria Teresa Pallotta, Maria Laura Belladonna and Claudia Volpi
Int. J. Mol. Sci. 2023, 24(22), 16236; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242216236 - 12 Nov 2023
Cited by 1 | Viewed by 1126
Abstract
Indoleamine 2,3-dioxygenase 2 (IDO2) is a paralog of Indoleamine 2,3-dioxygenase 1 (IDO1), a tryptophan-degrading enzyme producing immunomodulatory molecules. However, the two proteins are unlikely to carry out the same functions. IDO2 shows little or no tryptophan catabolic activity and exerts contrasting immunomodulatory roles [...] Read more.
Indoleamine 2,3-dioxygenase 2 (IDO2) is a paralog of Indoleamine 2,3-dioxygenase 1 (IDO1), a tryptophan-degrading enzyme producing immunomodulatory molecules. However, the two proteins are unlikely to carry out the same functions. IDO2 shows little or no tryptophan catabolic activity and exerts contrasting immunomodulatory roles in a context-dependent manner in cancer and autoimmune diseases. The recently described potential non-enzymatic activity of IDO2 has suggested its possible involvement in alternative pathways, resulting in either pro- or anti-inflammatory effects in different models. In a previous study on non-small cell lung cancer (NSCLC) tissues, we found that IDO2 expression revealed at the plasma membrane level of tumor cells was significantly associated with poor prognosis. In this study, the A549 human cell line, basally expressing IDO2, was used as an in vitro model of human lung adenocarcinoma to gain more insights into a possible alternative function of IDO2 different from the catalytic one. In these cells, immunocytochemistry and isopycnic sucrose gradient analyses confirmed the IDO2 protein localization in the cell membrane compartment, and the immunoprecipitation of tyrosine-phosphorylated proteins revealed that kinase activities can target IDO2. The different localization from the cytosolic one and the phosphorylation state are the first indications for the signaling function of IDO2, suggesting that the IDO2 non-enzymatic role in cancer cells is worthy of deeper understanding. Full article
(This article belongs to the Special Issue Lung Cancer: From Molecular Mechanisms to Novel Therapeutics)
Show Figures

Figure 1

Back to TopTop