Erleada - Mechanism of Action

Erleada - Mechanism of Action

Erleada, also known as apalutamide, is a medication that belongs to a class of drugs called androgen receptor inhibitors. This medication is recommended for grown-up individuals who have been diagnosed with prostate cancer that has not spread to other parts of the body (non-metastatic castration-resistant prostate cancer) or has spread to other regions (metastatic castration-sensitive prostate cancer). Erleada works by blocking androgen receptors, which are proteins found in prostate cells that are essential for the growth and survival of cancer cells. By preventing the binding of androgens like testosterone, Erleada slows down the growth and progression of prostate cancer. Erleada's mechanism of action disrupts the androgen signaling pathway, further impairing the growth and survival of cancer cells. This innovative treatment option has shown promising results in clinical trials and offers improved outcomes for patients with prostate cancer.

Mechanism of Action

Inhibition of Androgen Receptors

Erleada operates through the androgen signaling pathway, an essential process in prostate cancer development and progression. By attaching to the androgen receptor, Erleada suppresses the receptor's activation by androgenic hormones like testosterone. This action obstructs the translocation of the androgen receptor into the nucleus and its binding to DNA, vital steps for gene expression involved in prostate cancer cell growth and survival. Furthermore, Erleada curbs the production of androgens by hindering CYP17's enzymatic activity, a crucial contributing factor to androgen synthesis. In inhibiting both the androgen receptor and androgen production, Erleada effectively disrupts the signaling pathway, suppressing the survival and proliferation of prostate cancer cells.

Disruption of Androgen Signaling Pathway

Erleada, through its defined mechanism, directly impacts prostate cancer cells. By blocking androgen receptors, it prevents the connection between androgen hormones such as testosterone and these receptors. This interference disrupts the critical androgen signaling pathway, crucial in sustaining prostate cancer cells and their growth. Consequently, this action by Erleada curtails the growth of these cancer cells and instigates their mortality - a process known as apoptosis. The impact on prostate cancer cells considerably contributes to Erleada's overall therapeutic potential in treating prostate cancer.

erleada mechanism of action

Effects on Prostate Cancer Cells

In clinical trials, Erleada has shown considerable effects in slowing down disease progression. Studies involving patients with a diagnosis of non-metastatic castration-resistant prostate cancer indicate that the drug markedly delayed the metastasis timeframe or mortality when compared to placebo subjects. This result implies that patients undergoing Erleada treatment had cancer spread at a slower rate, and their life expectancy increased. Additionally, when co-administered with the standard Androgen Deprivation Therapy (ADT) to patients with metastatic castration-sensitive prostate cancer, Erleada demonstrated significant improvement in radiographic progression-free survival, outperforming the placebo plus ADT. These positive outcomes suggest that Erleada does not merely delay disease progression; it also enhances survival rates. These scientific findings exemplify the efficacy of Erleada in decelerating prostate cancer progression, providing patients an extended duration of disease control and an improved quality of life.

Impact on Disease Progression

Erleada's direct effects on prostate cancer cells through its mechanism of action have demonstrated specific impacts on cancer cells. The inhibition of androgen receptors by Erleada impairs the docking of androgen hormones, such as testosterone, to these receptors. This suppression disrupts the crucial androgen signaling pathway that is essential for the growth and maintenance of prostate cancer cells.This results in the reduction of cancer cell proliferation and the promotion of their apoptotic demise. These observed effects on prostate cancer cells underline the collective therapeutic effectiveness of Erleada in managing prostate cancer.

Clinical Applications

Use in Metastatic Castration-Sensitive Prostate Cancer

Its role has been further highlighted when used in combination with androgen deprivation therapy (ADT). It not only enhances the potency of ADT but alters the patient outcomes, giving a new treatment paradigm for people afflicted with metastatic castration-sensitive prostate cancer. Another dimension to Erleada's application is its approval for non-metastatic castration-resistant prostate cancer (nmCRPC) where it serves as an oral androgen receptor inhibitor.

Use in Non-Metastatic Castration-Resistant Prostate Cancer

In nmCRPC, the drug effectively curbs the activation of signalling pathways downstream that promote the survival and evolution of the tumor, despite effective surgical or medical castration, when PSA levels persistently rise, indicating disease progression. The drug's mechanism of disrupting androgen signalling delays the progression of nmCRPC, reducing time to metastasis and mitigating risk of skeletal events that are symptomatic. Through this, it delivers a significant period of disease control and a better life quality for patients, creating a viable treatment opportunity for people suffering with this advanced prostate cancer form.

Combination Therapy with Other Agents

Erleada has reflected positive results when combined with other agents for treating prostate cancer, with improved survival rates and progression-free survival as compared to ADT alone. Various clinical studies have tested the effectiveness of Erleada in combination with other drugs like abiraterone acetate and prednisone, enzalutamide, and docetaxel. The synergistic outcomes have augmented its overall effectiveness in suppressing androgen receptor signalling and inhibiting prostate cancer cell growth. Further, the combination of Erleada can potentiate its action by delaying resistance development to treatment, thus improving therapeutic outcomes in metastatic castration-resistant prostate cancer patients. Research is ongoing to explore more about this combined therapy involving Erleada and other agents that could escalate treatment response and boost patient outcomes in diverse prostate cancer stages.

Bibliography

  1. Chung, C., & Abboud, K. (2022). Targeting the androgen receptor signaling pathway in advanced prostate cancer. American Journal of Health-System Pharmacy, 79(15), 1224-1235. (https://www.researchgate.net/profile/Clement-Chung/publication/359805449_Targeting_the_androgen_receptor_signaling_pathway_in_advanced_prostate_cancer/links/627dd136b1ad9f66c8b9783c/Targeting-the-androgen-receptor-signaling-pathway-in-advanced-prostate-cancer.pdf)

  2. Rajaram, P., Rivera, A., Muthima, K., Olveda, N., Muchalski, H., & Chen, Q. H. (2020). Second-generation androgen receptor antagonists as hormonal therapeutics for three forms of prostate cancer. Molecules, 25(10), 2448. (https://www.mdpi.com/1420-3049/25/10/2448/pdf)

  3. Prutianu, I., CĂRUNTU, I. D., Manole, M. B., GIUȘCĂ, S. E., Profire, B., Timofte, A. D., ... & Gafton, B. (2021). DEVELOPMENTS IN PROSTATE CANCER THERAPY: FROM THE ANDROGEN RECEPTOR TO ANTIANDROGENS AND BEYOND. Farmacia, 69(3). (https://farmaciajournal.com/wp-content/uploads/art-01-Prutianu_Caruntu_Gafton_389-398.pdf)

  4. Zhao, J., Ning, S., Lou, W., Yang, J. C., Armstrong, C. M., Lombard, A. P., ... & Liu, C. (2020). Cross-resistance among next-generation antiandrogen drugs through the AKR1C3/AR-V7 axis in advanced prostate cancer. Molecular cancer therapeutics, 19(8), 1708-1718. (https://aacrjournals.org/mct/article/19/8/1708/92824)

  5. Potdar, R., Gartrell, B. A., Given, R., Karsh, L., Frankel, J., Nenno, K., ... & Pieczonka, C. (2022). Concomitant use of oral anticoagulants in patients with advanced prostate cancer receiving apalutamide: A post-hoc analysis of TITAN and SPARTAN studies. American Journal of Cancer Research, 12(1), 445. (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8822273/)

  6. Zhang, J., Sun, J., Bakht, S., & Hassan, W. (2022). Recent Development and Future Prospects of Molecular Targeted Therapy in Prostate Cancer. Current Molecular Pharmacology, 15(1), 159-169. (https://www.ingentaconnect.com/content/ben/cmp/2022/00000015/00000001/art00010)

  7. Duran, I., Carles, J., Bulat, I., Hellemans, P., Mitselos, A., Ward, P., ... & Chien, C. (2020). Pharmacokinetic drug--drug interaction of apalutamide, part 1: clinical studies in healthy men and patients with castration-resistant prostate cancer. Clinical pharmacokinetics, 59, 1135-1148. (https://link.springer.com/article/10.1007/s40262-020-00882-2)

  8. Christenson, M., Song, C. S., Liu, Y. G., & Chatterjee, B. (2022). Precision targets for intercepting the lethal progression of prostate cancer: Potential avenues for personalized therapy. Cancers. (https://www.mdpi.com/2072-6694/14/4/892)

  9. Miller, D. R., Ingersoll, M. A., Teply, B. A., & Lin, M. F. (2021). Combination treatment options for castration-resistant prostate cancer. Exon Publications. (https://exonpublications.com/index.php/exon/article/download/298/625)

  10. Eberli, D., Kranzbühler, B., Mortezavi, A., Sulser, T., & Salemi, S. (2020, August). Apalutamide in combination with autophagy inhibitors improves treatment effects in prostate cancer cells. In Urologic Oncology: Seminars and Original Investigations (Vol. 38, No. 8, pp. 683-e19). Elsevier. (https://www.zora.uzh.ch/id/eprint/191325/1/\~2158265.pdf)