Journal of Cancer Immunology & Therapy

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.
Reach Us +1 (202) 780-3397

Mini Review - Journal of Cancer Immunology & Therapy (2024) Volume 7, Issue 1

Unraveling the Role of Hormones in Breast Cancer: Insights into Development and Treatment.

Fiona Crane *

School of Psychology and Public Health, La Trobe University, Australia

*Corresponding Author:
Fiona Crane
School of Psychology and Public Health, La Trobe University, Australia
E-mail: f.crane@latrobe.edu.au

Received: 02-Feb -2024, Manuscript No. AAJCIT-24-129827; Editor assigned: 03-Feb-2024, PreQC No. AAJCIT-24-129827 (PQ); Reviewed:17-Feb-2024, QC No. AAJCIT-24-129827; Revised:22-Feb-2024, Manuscript No. AAJCIT-24-129827 (R); Published:29-Feb-2024, DOI:10.35841/aara-7.1.192

Citation: Crane F., Unraveling the role of hormones in breast cancer: Insights into development and treatment. J Cancer Immunol Ther. 2024;7(1):192

Visit for more related articles at Journal of Cancer Immunology & Therapy

Introduction

Breast cancer remains one of the most prevalent malignancies affecting women worldwide, with hormonal factors playing a pivotal role in its development and progression. The intricate interplay between hormones and breast cancer has been the subject of extensive research, leading to significant advancements in our understanding of the disease. Additionally, it discusses the implications of hormonal therapies in breast cancer management, highlighting the importance of personalized approaches tailored to individual hormone receptor status [1].

Estrogen receptors (ER) and progesterone receptors (PR) expressed on breast cells mediate the effects of estrogen and progesterone, respectively, regulating gene expression and cellular proliferation. Hormonal fluctuations during puberty, menstrual cycles, pregnancy, and menopause influence breast tissue dynamics, highlighting the dynamic interplay between hormones and breast physiology [2].

Dysregulation of hormonal signaling pathways plays a central role in the pathogenesis of hormone receptor-positive breast cancer, which accounts for approximately 75% of all breast cancer cases. In estrogen receptor-positive (ER+) breast cancer, aberrant estrogen signaling drives tumor growth and progression. Elevated estrogen levels, either endogenously produced or exogenously administered, promote cell proliferation, inhibit apoptosis, and induce genomic instability, fostering the development of ER+ breast tumors. Similarly, progesterone signaling contributes to tumor progression in hormone receptor-positive breast cancer, albeit to a lesser extent than estrogen [3].

The assessment of hormone receptor status, particularly estrogen and progesterone receptor expression, plays a crucial role in guiding treatment decisions and predicting prognosis in breast cancer patients. Hormone receptor-positive tumors are typically associated with a favorable prognosis and are more likely to respond to endocrine therapies, such as selective estrogen receptor modulators (SERMs), aromatase inhibitors (AIs), and selective estrogen receptor degraders (SERDs). In contrast, hormone receptor-negative (ER-/PR-) tumors are less responsive to hormonal therapies and often require alternative treatment approaches, such as chemotherapy or targeted therapies [4].

Selective estrogen receptor modulators (SERMs), such as tamoxifen and raloxifene, block estrogen binding to the ER, thereby inhibiting estrogen-dependent tumor proliferation. Aromatase inhibitors (AIs), including letrozole, anastrozole, and exemestane, suppress estrogen synthesis by inhibiting aromatase, the enzyme responsible for converting androgens into estrogens in postmenopausal women. Additionally, selective estrogen receptor degraders (SERDs), such as fulvestrant, promote degradation of the ER, leading to inhibition of estrogen signaling and tumor regression [5].

Despite the success of endocrine therapies, resistance to hormonal treatments remains a significant clinical challenge in breast cancer management. Ongoing research efforts are focused on identifying novel therapeutic targets and developing innovative treatment strategies to overcome resistance mechanisms [6].

Furthermore, precision medicine approaches, guided by genomic profiling and molecular subtyping, enable the identification of specific tumor vulnerabilities and the selection of tailored treatment regimens based on individual tumor characteristics [7].

Endocrine therapies represent a cornerstone of treatment for hormone receptor-positive breast cancer, aiming to disrupt estrogen signaling and inhibit tumor growth. This article explores the multifaceted role of hormones in breast cancer, encompassing their contributions to tumor initiation, growth, and response to treatment [8].

The normal development and function of the breast are intricately regulated by hormonal signals, primarily estrogen and progesterone. These hormones orchestrate the growth, differentiation, and maintenance of breast epithelial cells through complex signaling pathways [9].

Combination therapies, such as combining endocrine agents with targeted therapies or immunotherapies, hold promise for improving treatment outcomes and delaying disease progression in hormone receptor-positive breast cancer [10].

conclusion

The role of hormones in breast cancer development and treatment is multifaceted and dynamic, reflecting the intricate interplay between hormonal signaling pathways and tumor biology. Understanding the complex interactions between hormones and breast cancer is essential for guiding treatment decisions, predicting prognosis, and optimizing therapeutic outcomes. By leveraging insights from basic research and clinical trials, healthcare providers can deliver personalized and targeted therapies that effectively modulate hormonal pathways and improve outcomes for patients with hormone receptor-positive breast cancer.

References

  1. Clarke, R. B. Steroid receptors and proliferation in the human breast. Cancer Sur., 2013; 18, 57-68.
  2. Indexed at, Google Scholar, Cross Ref

  3. Early Breast Cancer Trialists' Collaborative Group. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet. 2011;378(9793), 771-784.
  4. Indexed at, Google Scholar, Cross Ref

  5. Higgins, M. J., & Baselga, J. Targeted therapies for breast cancer. J Clin Invest. 2011; 121(10), 3797-3803.
  6. Indexed at, Google Scholar

  7. Osborne, C. K., Schiff, R., & Fuqua, S. A. Estrogen receptor: Current understanding of its activation and modulation. Clin Cancer Res. 2001; 7(12 Supplement), 4338s-4342s.
  8. Indexed at, Google Scholar

  9. Anderson, W. F., Chatterjee, N., Ershler, W. B., & Brawley, O. W. Estrogen receptor breast cancer phenotypes in the Surveillance, Epidemiology, and End Results database. Breast Cancer Res Treat, 2002;76(1), 27-36.
  10. Indexed at, Google Scholar, Cross Ref

  11. Kaasa S, Loge JH, Aapro M, Albreht T, Anderson R, Bruera E, Brunelli C, Caraceni A, Cervantes A, Currow DC, Deliens L. Integration of oncology and palliative care: a Lancet Oncology Commission. Lancet OncolO. 2018;19(11):e588-653.
  12. Indexed at, Google Scholar, Cross Ref

  13. Schwartzberg L, Kim ES, Liu D, Schrag D. Precision oncology: who, how, what, when, and when not?. Am Soc Clin Oncol Educ Book. 2017;37:160-9.
  14. Indexed at, Google Scholar, Cross Ref

  15. Ma CX, Reinert T, Chmielewska I, Ellis MJ. Mechanisms of aromatase inhibitor resistance. Nat Rev Cancer. 2015;15(5):261-75.
  16. Indexed at,Google Scholar, Cross Ref

  17. Miller WR. Aromatase inhibitors: mechanism of action and role in the treatment of breast cancer. InSemin 2003;Vol. 30, pp. 3-11. WB Saunders.
  18. Indexed at, Google Scholar

  19. Turner, N. C., & Reis-Filho, J. S. Basal-like breast cancer and the BRCA1 phenotype. Oncogene. 2013;25(43), 5846-5853.
  20. Indexed at, Google Scholar

Get the App