Skip to main content

EDITORIAL article

Front. Pharmacol., 20 September 2024
Sec. Translational Pharmacology
This article is part of the Research Topic Molecular Targets in Oncological and Hematological Disease Management: Innovations in Precision Medicine View all 10 articles

Editorial: Molecular targets in oncological and hematological disease management: innovations in precision medicine

  • 1Department of Translational Medicine, Institute of Medical Research and Life Sciences - MedFuture, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
  • 2Department of Chemistry and Biochemistry, Seton Hall University, South Orange, NJ, United States
  • 3Department of Enzymology, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania

Targeted therapies represent a significant advancement in medicine, particularly in treating complex diseases such as cancer. These therapies, designed to specifically target biological mechanisms driving disease, can alter pathways that exacerbate disease progression, effectively slowing or halting tumor growth. Unlike traditional therapies that broadly affect both healthy and cancerous cells, targeted therapies minimize collateral damage by focusing on specific molecular triggers. This precision leads to superior outcomes while reducing adverse effects and toxicity (Buongervino et al., 2021; Fu et al., 2022; Zatovicova et al., 2022).

By providing a more specific and focused intervention, targeted therapies lead to improved treatment outcomes while reducing the risks of adverse effects and morbidity. Traditional medicines tend to affect a broader range of systems, often leading to more negative side effects (Canova et al., 2023; Disselhorst and Baas, 2020; Lohiya et al., 2023; Pralea et al., 2024). In contrast, targeted therapies focus on disease-related molecules, keeping healthy cells unexposed and minimizing patient toxicity.

Recent advances in precision medicine have accelerated the development of therapies targeting key genetic mutations or molecular abnormalities driving tumor growth (Zafar et al., 2024; Zhang et al., 2024). Personalized treatments designed according to each tumor’s unique DNA profile offer improved efficacy and reduced toxicity (Dailah et al., 2024; Guo et al., 2024; Liu et al., 2024; Tomuleasa et al., 2024). This Research Topic explores therapeutic agents that target disease-associated molecules, investigating strategies to alter molecular pathways responsible for disease progression and resistance to treatment.

Shafiei et al. investigated silibinin-loaded magnetic niosomal nanoparticles (MNNPs) for colorectal cancer treatment. Their findings demonstrated a significant increase in the cytotoxic effects of silibinin on cancer cells while sparing healthy cells. This study illustrates the growing importance of nanoparticle-based drug delivery systems in precision oncology, improving drug bioavailability and reducing side effects. The innovation presented in this study opens the door for broader applications of nanotechnology in cancer treatment.

Liu et al. reviewed diverse experimental models used in colorectal cancer (CRC) research, offering an in-depth overview of in vitro and in vivo models. Their work emphasizes the need to refine models to better replicate human CRC, providing a framework for testing targeted therapies like Shafiei’s MNNPs in ways that closely mimic human disease.

In hematological diseases, Li et al. examined the role of hemopexin (HPX) in mitigating heme toxicity during hemolysis, which commonly occurs in diseases such as sickle cell anemia and transfusion-induced hemolysis. While HPX protects against free heme, under certain conditions, it may exacerbate disease progression. This dual role emphasizes the need for more nuanced therapeutic approaches in hematology, similar to the precision seen in CRC treatments. Both studies demonstrate the complex dynamics of molecular precision in reducing harm and mitigating disease.

Zhang et al. presented a case study of a patient with severe combined immunodeficiency (SCID) who developed colon lymphoma due to a novel DCLRE1C mutation. It illustrates the intersection of immune system failure and cancer, highlighting the need for precision in both diagnostics and treatment. Early genetic diagnosis, as underscored by Li’s research on biomarkers, is critical for timely intervention, further emphasizing the importance of molecular precision in preventing and managing complex disease profiles.

Iacobescu et al. explored proteomics-based biomarkers for predicting and managing graft-versus-host disease (GVHD) in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). Their review delves into both single protein markers and protein panels, offering a comprehensive look at how these biomarkers can be used to stratify risk and diagnose GVHD. The authors emphasize how non-invasive biospecimens, such as blood or saliva, could be harnessed to enhance the precision of GVHD management.

Mocan et al. performed a proteomic exploration to identify novel serum biomarkers for intrahepatic cholangiocarcinoma (iCCA). Using high-throughput mass spectrometry, they identified several potential biomarkers, including S100A9, haptoglobin (HP), and serum amyloid A (SAA), which could distinguish iCCA from other liver conditions, such as cirrhosis and hepatocellular carcinoma. Their findings underscore the importance of proteomics in early cancer diagnosis and open the door to developing non-invasive diagnostic tools that could improve survival rates by enabling earlier detection and treatment.

Zhang et al. conducted a pan-cancer analysis of the TET family genes, particularly TET3, uncovering its role in tumor progression and drug sensitivity. Their bioinformatics analysis revealed that TET3 is involved in several cancer-related pathways, including cell cycle regulation, DNA damage response, and immune modulation. Targeting TET3 could inhibit tumor growth and increase chemotherapy sensitivity, positioning TET3 as a potential therapeutic target in a variety of cancers.

Chomczyk et al. provided an in-depth analysis of TP53 mutations in acute myeloid leukemia (AML) and their implications for disease progression. The review discussed the metabolic rewiring and immune evasion mechanisms driven by TP53 mutations, which are present in a subset of AML cases and associated with poor prognosis. Despite advances in targeted therapies, TP53-mutated AML remains a challenge due to its resistance to chemotherapy. This review called for further research into therapies targeting the metabolic vulnerabilities of TP53-mutated AML.

Munteanu et al. advanced the conversation around targeted therapies with their in vivo imaging system (IVIS) therapeutic assessment of tyrosine kinase inhibitor (TKI)-loaded gold nanoparticles for treating acute myeloid leukemia (AML). By using IVIS, the authors were able to track and assess the therapeutic effects of FLT3 inhibitors loaded into gold nanoparticles. Their preclinical study demonstrated that these nanocarriers improved drug delivery and bioavailability, offering enhanced tumor inhibition in FLT3-mutated AML models. This work reinforces the potential of nanomedicine in boosting the efficacy of precision therapies while minimizing systemic toxicity.

In conclusion, the studies featured in this Research Topic provide a comprehensive view of the current landscape of molecular targets in oncology and hematology. From novel drug delivery systems to biomarker identification, these contributions highlight the critical role of precision medicine in advancing treatment by targeting disease mechanisms, improving patient outcomes, and reducing the burden of traditional therapies.

Author contributions

AT: Writing–review and editing, Writing–original draft. GW: Writing–review and editing, Writing–original draft. SS: Writing–review and editing, Writing–original draft.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Acknowledgments

We would like to express our gratitude to all the authors who contributed to this Research Topic, as well as to the reviewers and invited editors who have played a significant role in enhancing its quality.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Buongervino, S., Lane, M. V., Garrigan, E., Zhelev, D. V., Dimitrov, D. S., and Bosse, K. R. (2021). Antibody-drug conjugate efficacy in neuroblastoma: role of payload, resistance mechanisms, target density, and antibody internalization. Mol. Cancer Ther. 20 (11), 2228–2239. doi:10.1158/1535-7163.MCT-20-1034

PubMed Abstract | CrossRef Full Text | Google Scholar

Canova, S., Trevisan, B., Abbate, M. I., Colonese, F., Sala, L., Baggi, A., et al. (2023). Novel therapeutic options for small cell lung cancer. Curr. Oncol. Rep. 25 (11), 1277–1294. doi:10.1007/s11912-023-01465-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Dailah, H. G., Hommdi, A. A., Koriri, M. D., Algathlan, E. M., and Mohan, S. (2024). Potential role of immunotherapy and targeted therapy in the treatment of cancer: a contemporary nursing practice. Heliyon 10 (2), e24559. doi:10.1016/j.heliyon.2024.e24559

PubMed Abstract | CrossRef Full Text | Google Scholar

Disselhorst, M. J., and Baas, P. (2020). Chemotherapy options versus “novel” therapies: how should we treat patients with malignant pleural mesothelioma. Transl. Lung Cancer Res. 9 (Suppl. 1), S77-S85–S85. doi:10.21037/tlcr.2020.01.16

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, Z., Li, S., Han, S., Shi, C., and Zhang, Y. (2022). Antibody drug conjugate: the “biological missile” for targeted cancer therapy. Signal Transduct. Target Ther. 7 (1), 93. doi:10.1038/s41392-022-00947-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, M., Sun, Y., Wei, Y., Xu, J., and Zhang, C. (2024). Advances in targeted therapy and biomarker research in thyroid cancer. Front. Endocrinol. (Lausanne) 15, 1372553. doi:10.3389/fendo.2024.1372553

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, B., Zhou, H., Tan, L., Siu, K. T. H., and Guan, X. Y. (2024). Exploring treatment options in cancer: tumor treatment strategies. Signal Transduct. Target Ther. 9 (1), 175. doi:10.1038/s41392-024-01856-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Lohiya, D. V., Mehendale, A. M., Lohiya, D. V., Lahoti, H. S., and Agrawal, V. N. (2023). Novel chemotherapy modalities for different cancers. Cureus 15 (9), e45474. doi:10.7759/cureus.45474

PubMed Abstract | CrossRef Full Text | Google Scholar

Pralea, I. E., Moldovan, R. C., Tigu, A. B., Moldovan, C. S., Fischer-Fodor, E., and Iuga, C. A. (2024). Cellular responses induced by NCT-503 treatment on triple-negativebreast cancer cell lines: a proteomics approach. Biomedicines 12 (5), 1087. doi:10.3390/biomedicines12051087

PubMed Abstract | CrossRef Full Text | Google Scholar

Tomuleasa, C., Tigu, A. B., Munteanu, R., Moldovan, C. S., Kegyes, D., Onaciu, A., et al. (2024). Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct. Target Ther. 9 (1), 201. doi:10.1038/s41392-024-01899-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Zafar, A., Khan, M. J., Abu, J., and Naeem, A. (2024). Revolutionizing cancer care strategies: immunotherapy, gene therapy, and molecular targeted therapy. Mol. Biol. Rep. 51 (1), 219. doi:10.1007/s11033-023-09096-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Zatovicova, M., Kajanova, I., Barathova, M., Takacova, M., Labudova, M., Csaderova, L., et al. (2022). Novel humanized monoclonal antibodies for targeting hypoxic human tumors via two distinct extracellular domains of carbonic anhydrase IX. Cancer Metab. 10 (1), 3. doi:10.1186/s40170-022-00279-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S., Xiao, X., Yi, Y., Wang, X., Zhu, L., Shen, Y., et al. (2024). Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct. Target Ther. 9 (1), 149. doi:10.1038/s41392-024-01848-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: targeted therapy, precision medicine, molecular diagnosis, innovations, nanomedicine

Citation: Tigu AB, Wiedman G and Szedlacsek SE (2024) Editorial: Molecular targets in oncological and hematological disease management: innovations in precision medicine. Front. Pharmacol. 15:1494396. doi: 10.3389/fphar.2024.1494396

Received: 10 September 2024; Accepted: 16 September 2024;
Published: 20 September 2024.

Edited and reviewed by:

Alastair George Stewart, The University of Melbourne, Australia

Copyright © 2024 Tigu, Wiedman and Szedlacsek. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Adrian Bogdan Tigu, tiguadrianbogdan@yahoo.com; Gregory Wiedman, gregory.wiedman@shu.edu; Stefan Eugen Szedlacsek, szedlacs@yahoo.co.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.