Page 229 - EJMO-9-3
P. 229

Eurasian Journal of
            Medicine and Oncology                                              FN3K–Nrf2 axis inhibition in breast cancer



            for breast cancer. However, several critical avenues remain   and capivasertib – that significantly inhibited FN3K
            to be explored to fully establish its clinical relevance. First,   expression and function. This inhibition was corresponded
            while the study establishes the inhibitory effects of selected   with suppression of Nrf2-mediated antioxidant signaling
            FN3K inhibitors on breast cancer cell lines, further in-depth   and reduced  cancer  cell  viability,  supporting the  role  of
            mechanistic investigations are required to elucidate the   FN3K in tumor progression and redox adaptation.
            precise molecular role of FN3K in cancer progression.   Importantly, all  three  compounds  demonstrated
            These should include studies on FN3K-mediated metabolic   strong selectivity toward breast cancer cells with minimal
            adaptations and its impact on oxidative stress pathways
            beyond Nrf2 signaling; as such investigations would   cytotoxicity in normal cells, indicating a favorable
            provide  deeper  insights into its therapeutic  potential.   therapeutic index. These findings collectively support the
            Secondly, in vivo  studies and pharmacokinetics are   FN3K–Nrf2 axis as a promising molecular target in redox-
            critical; although initial  in vivo validation demonstrated   driven breast cancer subtypes.
            significant tumor growth suppression, comprehensive   While the present findings are encouraging, we
            pharmacokinetic and toxicological evaluations in animal   acknowledge the limitation that in vivo validation was not
            models  are  essential.  Determining  the  bioavailability,   included in this phase of the study. To address this, future
            metabolic stability, and potential off-target effects of FN3K   experiments are already planned to assess the efficacy and
            inhibitors will be crucial before progressing to clinical   safety  of  FN3K  inhibition  in  xenograft  tumor  models.
            trials. In addition, the development of selective FN3K   These will include evaluation of tumor growth suppression,
            inhibitors  from  existing  FDA-approved  drugs,  future   pharmacokinetic profiles, and modulation of the FN3K–
            efforts should focus on rational drug design to develop   Nrf2 axis in excised tumor tissues. In addition, downstream
            more selective inhibitors with minimal off-target effects.   molecular pathway mapping, ADME optimization, and
            Computational approaches, such as MD simulations and   combinatorial strategies with standard therapeutics will be
            free energy perturbation studies can aid in optimizing   explored to facilitate translational relevance.
            lead compounds for enhanced specificity and efficacy.
            Furthermore, exploration of combination therapies is   Overall,  this  work  establishes  FN3K  inhibition
            warranted. Given the role of FN3K in modulating oxidative   as  a viable therapeutic  strategy for  breast  cancer  and
            stress responses, investigating its inhibition in combination   provides a strong foundation for future  in vivo and
            with chemotherapeutic agents or targeted therapies could   clinical investigations. Based on the present stage of
            provide synergistic effects. Evaluating the impact of FN3K   validation, oxaliplatin, lansoprazole, and capivasertib
            inhibitors on drug resistance mechanisms may reveal novel   should be regarded as promising pre-clinical candidates
            combination strategies to enhance treatment efficacy. For   for FN3K-targeted intervention, rather than as definitive
            clinical translation and biomarker development, studies   therapeutic options. Further  in vivo and combination
            should assess FN3K expression patterns in patient-derived   therapy studies are required to fully determine their
            tumor samples and correlate them with disease progression   clinical applicability.
            and treatment response. Identifying reliable biomarkers
            associated with FN3K inhibition will be instrumental in   Acknowledgments
            patient stratification and personalized therapy approaches.   The  authors  acknowledge  the  JSS  Academy  of  Higher
            Finally, CRISPR/Cas9-based gene editing approaches,   Education and Research and the JSS College of Pharmacy
            including knockout or knockdown studies in breast cancer   for providing institutional support. The Department of
            models, can help validate the direct contribution of FN3K   Computer Aided Drug Design is gratefully acknowledged
            to tumorigenesis. Functional studies in FN3K-deficient   for providing the computational resources essential for
            models will clarify its essentiality in breast cancer survival   the in silico studies. The authors also express their sincere
            and progression.                                   thanks to Dr. Subramanyam P, Muppandal Genomics and
              By addressing these critical aspects, FN3K-targeted   Immunologicals Pvt. Ltd., Hyderabad, for his technical
            therapy can move closer to clinical application, offering a   assistance in conducting the in vitro biological experiments
            promising avenue for breast cancer treatment.      and supporting data validation.
            6. Conclusion                                      Funding

            The present study underscores the therapeutic potential   The work was supported by the Council of Scientific and
            of targeting FN3K in breast cancer management. By   Industrial Research, Human Resource Development
            integrating in silico virtual screening with in vitro validation,   Group (CSIR-HRDG), New Delhi, India (Grant No.111-
            we identified lead compounds – oxaliplatin, lansoprazole,   5634- 11759/2023/1 dated February 19 , 2024).
                                                                                              th

            Volume 9 Issue 3 (2025)                        221                         doi: 10.36922/EJMO025150114
   224   225   226   227   228   229   230   231   232   233   234