Next Article in Journal
Significance of LIF/LIFR Signaling in the Progression of Obesity-Driven Triple-Negative Breast Cancer
Previous Article in Journal
Targeting Cancer Metabolism as a New Strategy to Enhance Treatment Efficacy and Overcome Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

State of the Art and New Perspectives in Lung Cancer Therapeutics

by
Kostas A. Papavassiliou
1,
Amalia A. Sofianidi
2,
Vassiliki A. Gogou
1 and
Athanasios G. Papavassiliou
2,*
1
First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(21), 3631; https://doi.org/10.3390/cancers16213631
Submission received: 24 October 2024 / Accepted: 25 October 2024 / Published: 28 October 2024
(This article belongs to the Section Tumor Microenvironment)
Cisplatin became a first-line chemotherapy regimen for lung cancer in the mid-1980s, marking a pivotal advance in lung cancer treatment [1,2]. Since then, the therapeutic landscape has evolved significantly, with major breakthroughs in surgical techniques, chemotherapy regimens, precision medicine, immunotherapy, and targeted therapies. However, despite these innovations, the overall 5-year survival rate across all stages of lung cancer remains low, at just 19% [3]. Alarmingly, 57% of patients are diagnosed at an advanced stage, where the disease has already metastasized throughout the body [4]. These realities underscore an urgent need for cutting-edge therapies to combat this formidable disease.
Targeted therapies aim to counteract the molecular alterations driving lung cancer. While third-generation tyrosine kinase inhibitors (TKIs) have been effective, resistance inevitably develops over time [5]. BBT-176, a novel fourth-generation TKI currently undergoing clinical trials, has shown promising preliminary efficacy against the epidermal growth factor receptor (EGFR) C797S mutation—a key factor in the development of such resistance [6]. Recent progress has led to the development of small-molecule inhibitors like sotorasib that target Kirsten rat sarcoma (KRAS) mutations, previously considered “undruggable” [7]. Ongoing research aims to refine these drugs and expand direct or indirect treatment options for patients with other KRAS alterations [8]. Regarding anaplastic lymphoma kinase (ALK) inhibitors, SAF-189s is an innovative ALK inhibitor that has demonstrated potential in preclinical studies to overcome most known resistance mutations linked to ALK [9]. The latest additions to the armamentarium against lung cancer include a rapidly emerging class of therapeutics called antibody–drug conjugates, such as trastuzumab deruxtecan, which targets the human epidermal growth factor receptor 2 (HER2) [10]. The promise of targeted therapy lies in combining these molecules with each other and integrating them with other therapeutic approaches.
In the field of immunotherapy, the future of lung cancer treatment is being shaped by pioneering approaches such as lung cancer vaccines, adoptive cell transfer (ACT), and oncolytic viruses. Preventative vaccines for high-risk groups are anticipated in the coming years, aiming to reduce the incidence of the disease. Notably, this past August, the first lung cancer patient received an mRNA vaccine, heralding the beginning of a new era in therapeutic vaccines. ACT includes chimeric antigen receptor T (CAR-T) cells and tumor-infiltrating lymphocyte (TIL) therapy, with both currently being evaluated in clinical trials [11]. Dendritic cell (DC) vaccination has the potential to transform the tumor microenvironment (TME) in lung cancer patients, promoting sustained anti-tumor immune activity [12]. This approach may also offer the advantage of fewer adverse events, such as cytokine release syndrome (CRS), which is often associated with CAR-T cell therapy [13]. Oncolytic viruses operate through a dual mechanism: they selectively replicate within and destroy tumor cells that lack effective antiviral defenses, thereby stimulating a stronger antitumor adaptive immune response. Simultaneously, when oncolytic viruses infect healthy cells, they trigger antiviral reactions that help recruit immune cells to the TME, further enhancing the immune attack against tumor cells [14].
Recent advancements in radiation therapy have significantly improved its precision, efficacy, and safety, paving the way toward innovative future applications. Intensity-modulated radiotherapy (IMRT) and volumetric-modulated arc therapy (VMAT) are sophisticated radiation treatment techniques that enhance dosimetric results in lung cancer care by precisely focusing on tumors while protecting surrounding healthy tissue [15]. Stereotactic body radiotherapy (SBRT) administers high doses of radiation with remarkable accuracy to small, well-defined tumors [16]. Additionally, the highly promising boron neutron capture therapy (BNCT) leverages the selective accumulation of boron-10 compounds within cancer cells, allowing for the targeted destruction of these malignant cells [17]. Building on this, artificial intelligence (AI) offers a powerful tool for fostering cancer care. AI can improve the accuracy of tumor identification, optimize radiation dosing, and even predict patient responses to treatment. By exploiting machine learning algorithms to analyze extensive patient data, personalized treatment plans can be developed, leading to better outcomes and fewer side effects. Notably, radiomics—through the analysis of imaging features—offers deeper insights into tumor biology and behavior [18], heralding a new era of tailored and effective radiation therapies.
Various types of nanoparticles have recently emerged as powerful tools for enhancing cancer drug delivery in both preclinical studies and clinical trials, offering the promise of controlled and sustained release [19]. Lipid-based nanoparticles, such as liposomes, solid lipid nanoparticles, and nanostructured lipid carriers, are known for their biocompatibility and capacity to encapsulate both hydrophilic and hydrophobic drugs. Polymeric nanoparticles, including micelles, dendrimers, and nanogels, provide versatility in drug loading and sustained release. Inorganic nanoparticles, like gold, silver, and iron oxide nanoparticles, as well as quantum dots, are valued for their stability and potential use in targeted therapies. Additionally, hybrid nanoparticles, which combine organic and inorganic components, offer enhanced functionality and precision, making them promising candidates for more effective and targeted lung cancer treatments [8,19].
In conclusion, the therapeutic landscape of lung cancer treatment is moving from a one-size-fits-all approach to a more personalized one. The development of predictive biomarkers, combined with the valuable support of AI, has the potential to provide lung cancer patients with more tailored and effective treatments, ultimately improving their quality of life. Nevertheless, it is crucial to address the risk of widening disparities in cancer care, as personalized approaches could come with higher costs, potentially limiting access for some patients.

Author Contributions

Conceptualization, K.A.P., A.A.S. and A.G.P.; writing—original draft preparation, K.A.P., A.A.S. and V.A.G.; literature search and preparation of all references, A.A.S. and V.A.G.; supervision, A.G.P.; writing—review and editing, K.A.P. and A.G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Evans, W.K.; Shepherd, F.A.; Feld, R.; Osoba, D.; DeBoer, G. First-Line Therapy with VP-16 and Cisplatin for Small-Cell Lung Cancer. Semin. Oncol. 1986, 13, 17–23. [Google Scholar] [CrossRef] [PubMed]
  2. Evans, W.K.; Shepherd, F.A.; Feld, R.; Osoba, D.; Dang, P.; Deboer, G. VP-16 and Cisplatin as First-Line Therapy for Small-Cell Lung Cancer. J. Clin. Oncol. 1985, 3, 1471–1477. [Google Scholar] [CrossRef] [PubMed]
  3. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  4. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  5. He, J.; Huang, Z.; Han, L.; Gong, Y.; Xie, C. Mechanisms and Management of 3rd-generation EGFR-TKI Resistance in Advanced Non-small Cell Lung Cancer (Review). Int. J. Oncol. 2021, 59, 90. [Google Scholar] [CrossRef] [PubMed]
  6. Lim, S.M.; Ahn, J.S.; Hong, M.-H.; Kim, T.M.; Jung, H.-A.; Jung, H.-A.; Ou, S.-H.I.; Jeong, S.; Lee, Y.-H.; Yim, E.; et al. MA07.09 BBT-176, a 4th Generation EGFR TKI, for Progressed NSCLC after EGFR TKI Therapy: PK, Safety and Efficacy from Phase 1 Study. J. Thorac. Oncol. 2022, 17, S70–S71. [Google Scholar] [CrossRef]
  7. Skoulidis, F.; Li, B.T.; Dy, G.K.; Price, T.J.; Falchook, G.S.; Wolf, J.; Italiano, A.; Schuler, M.; Borghaei, H.; Barlesi, F.; et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N. Engl. J. Med. 2021, 384, 2371–2381. [Google Scholar] [CrossRef] [PubMed]
  8. La’ah, A.S.; Chiou, S.-H. Cutting-Edge Therapies for Lung Cancer. Cells 2024, 13, 436. [Google Scholar] [CrossRef] [PubMed]
  9. Yang, J.-J.; Zhou, J.; Cheng, Y.; Li, M.; Zhao, Q.; Zhang, Z.; Zang, A.; Fan, Y.; Hui, A.-M.; Zhou, Y.; et al. SAF-189s in Advanced, ALK-Positive, Non–Small Cell Lung Cancer: Results from a First-in-Human Phase 1/2, Multicenter Study. J. Clin. Oncol. 2022, 40, 9076. [Google Scholar] [CrossRef]
  10. Li, B.T.; Smit, E.F.; Goto, Y.; Nakagawa, K.; Udagawa, H.; Mazières, J.; Nagasaka, M.; Bazhenova, L.; Saltos, A.N.; Felip, E.; et al. Trastuzumab Deruxtecan in HER2 -Mutant Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2022, 386, 241–251. [Google Scholar] [CrossRef] [PubMed]
  11. Katiyar, V.; Chesney, J.; Kloecker, G. Cellular Therapy for Lung Cancer: Focusing on Chimeric Antigen Receptor T (CAR T) Cells and Tumor-Infiltrating Lymphocyte (TIL) Therapy. Cancers 2023, 15, 3733. [Google Scholar] [CrossRef] [PubMed]
  12. Abascal, J.; Oh, M.S.; Liclican, E.L.; Dubinett, S.M.; Salehi-Rad, R.; Liu, B. Dendritic Cell Vaccination in Non-Small Cell Lung Cancer: Remodeling the Tumor Immune Microenvironment. Cells 2023, 12, 2404. [Google Scholar] [CrossRef] [PubMed]
  13. Shah, D.; Soper, B.; Shopland, L. Cytokine Release Syndrome and Cancer Immunotherapies–Historical Challenges and Promising Futures. Front. Immunol. 2023, 14, 1190379. [Google Scholar] [CrossRef] [PubMed]
  14. Nistal-Villan, E.; Rius-Rocabert, S.; Llinares-Pinel, F. Oncolytic Virotherapy in Lung Cancer. In International Review of Cell and Molecular Biology; Elsevier: Amsterdam, The Netherlands, 2023; Volume 379, pp. 221–239. ISBN 978-0-443-19249-4. [Google Scholar]
  15. Ko, Y.E.; Ahn, S.D.; Je, H.U. Usability and Necessity of a Novel Hybrid Radiation Therapy Technique Based on Volumetric Modulated Arc Therapy (VMAT) in Stage III Lung Cancer Treatment. Radiat. Phys. Chem. 2022, 195, 110054. [Google Scholar] [CrossRef]
  16. Buchberger, D.S.; Videtic, G.M.M. Stereotactic Body Radiotherapy for the Management of Early-Stage Non–Small-Cell Lung Cancer: A Clinical Overview. JCO Oncol. Pract. 2023, 19, 239–249. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, S.; Zhang, Z.; Miao, L.; Li, Y. Boron Neutron Capture Therapy: Current Status and Challenges. Front. Oncol. 2022, 12, 788770. [Google Scholar] [CrossRef] [PubMed]
  18. Van Timmeren, J.E.; Cester, D.; Tanadini-Lang, S.; Alkadhi, H.; Baessler, B. Radiomics in Medical Imaging—“How-to” Guide and Critical Reflection. Insights Imaging 2020, 11, 91. [Google Scholar] [CrossRef] [PubMed]
  19. Carrasco-Esteban, E.; Domínguez-Rullán, J.A.; Barrionuevo-Castillo, P.; Pelari-Mici, L.; Leaman, O.; Sastre-Gallego, S.; López-Campos, F. Current Role of Nanoparticles in the Treatment of Lung Cancer. J. Clin. Transl. Res. 2021, 7, 140–155. [Google Scholar] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Papavassiliou, K.A.; Sofianidi, A.A.; Gogou, V.A.; Papavassiliou, A.G. State of the Art and New Perspectives in Lung Cancer Therapeutics. Cancers 2024, 16, 3631. https://doi.org/10.3390/cancers16213631

AMA Style

Papavassiliou KA, Sofianidi AA, Gogou VA, Papavassiliou AG. State of the Art and New Perspectives in Lung Cancer Therapeutics. Cancers. 2024; 16(21):3631. https://doi.org/10.3390/cancers16213631

Chicago/Turabian Style

Papavassiliou, Kostas A., Amalia A. Sofianidi, Vassiliki A. Gogou, and Athanasios G. Papavassiliou. 2024. "State of the Art and New Perspectives in Lung Cancer Therapeutics" Cancers 16, no. 21: 3631. https://doi.org/10.3390/cancers16213631

APA Style

Papavassiliou, K. A., Sofianidi, A. A., Gogou, V. A., & Papavassiliou, A. G. (2024). State of the Art and New Perspectives in Lung Cancer Therapeutics. Cancers, 16(21), 3631. https://doi.org/10.3390/cancers16213631

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop