GET THE APP

Immunotherapy and Targeted Therapies in Breast Cancer Treatment
..

Journal of Cancer Clinical Trials

ISSN: 2577-0535

Open Access

Commentary - (2023) Volume 8, Issue 3

Immunotherapy and Targeted Therapies in Breast Cancer Treatment

Sawicki Tomasz*
*Correspondence: Sawicki Tomasz, Department of Medicine & Advanced Technology, Central Michigan University, Mt Pleasant, USA, Email:
Department of Medicine & Advanced Technology, Central Michigan University, Mt Pleasant, USA

Received: 01-Jun-2023, Manuscript No. jcct-23-118153; Editor assigned: 03-Jun-2023, Pre QC No. P-118153; Reviewed: 17-Jun-2023, QC No. Q-118153; Revised: 22-Jun-2023, Manuscript No. R-118153; Published: 29-Jun-2023 , DOI: 10.37421/2577-0535.2023.8.223
Citation: Tomasz, Sawicki. “Immunotherapy and Targeted Therapies in Breast Cancer Treatment.” J Cancer Clin Trails 8 (2023): 223.
Copyright: © 2023 Tomasz S. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Breast cancer is a complex and heterogeneous disease that affects millions of women worldwide. While traditional treatment modalities such as surgery, chemotherapy and radiation therapy have been cornerstones in the fight against breast cancer, the landscape of breast cancer treatment is evolving rapidly. Two promising avenues in this evolution are immunotherapy and targeted therapies. These innovative approaches have shown great promise in improving treatment outcomes, reducing side effects and offering new hope to breast cancer patients. Immunotherapy is a groundbreaking approach that harnesses the body's own immune system to target and destroy cancer cells. In breast cancer, immunotherapy has gained significant attention, particularly in cases that are resistant to conventional treatments. The future of immunotherapy in breast cancer looks promising. Ongoing research aims to refine patient selection, develop novel combination therapies and optimize treatment regimens. Advancements in understanding the tumor microenvironment, the role of the immune system and the genetics of breast cancer will continue to drive progress in this field.

Keywords

Immunotherapy • Breast cancer • Immune checkpoint inhibitors

Introduction

Immune checkpoint inhibitors, such as pembrolizumab and atezolizumab, have shown promise in treating Triple-Negative Breast Cancer (TNBC). These inhibitors release the brakes on the immune system, enabling it to recognize and attack cancer cells. Clinical trials have demonstrated significant response rates in TNBC patients, offering new hope for this aggressive subtype. Immune checkpoint inhibitors are a type of immunotherapy that has gained significant attention in breast cancer treatment, particularly in the case of triple-negative breast cancer. Drugs like pembrolizumab and atezolizumab work by blocking certain proteins that prevent immune cells from attacking cancer cells. By releasing these "brakes," the immune system can recognize and attack cancer cells more effectively.

Chimeric Antigen Receptor T-cell (CAR-T) therapy is a personalized treatment that involves modifying a patient's own T-cells to target specific proteins expressed on breast cancer cells. Clinical trials are underway to explore the potential of CAR-T therapy in breast cancer, with a focus on HER2-positive tumors [1]. Chimeric antigen receptor T-cell therapy is a groundbreaking approach that modifies a patient's own T-cells to target specific proteins expressed on the surface of breast cancer cells. While this therapy has primarily been associated with hematological cancers, research is ongoing to adapt CAR-T therapy for breast cancer, especially HER2-positive tumors.

Description

Cancer vaccines, like the HER2-targeted NeuVax, are designed to stimulate the immune system's response against breast cancer cells. They offer the potential to prevent disease recurrence and are currently undergoing clinical trials. Vaccines have long been used for infectious diseases, but they are now being explored in the context of cancer. In breast cancer, vaccines are designed to stimulate the immune system's response against cancer cells. NeuVax, a vaccine targeting HER2, is currently in clinical trials, showing potential in preventing disease recurrence. Immunotherapy in breast cancer has demonstrated significant potential. It can lead to durable responses, potentially with fewer side effects than traditional treatments. For example, immunotherapies may be better tolerated by patients as they specifically target cancer cells, sparing healthy tissue.

However, there are challenges to overcome. Not all breast cancer subtypes respond equally to immunotherapy, making patient selection crucial. Identifying predictive biomarkers to determine who will benefit the most from these treatments is an ongoing area of research. Moreover, the management of immune-related adverse events, where the immune system mistakenly attacks healthy tissues, is an important consideration in immunotherapy. While side effects can often be managed, they require close monitoring and intervention [2,3]. Targeted therapies are drugs designed to interfere with specific molecules or pathways involved in the growth and spread of cancer cells. These therapies aim to disrupt the unique characteristics of cancer cells, leaving healthy cells unharmed. HER2 (human epidermal growth factor receptor 2) is a protein that is overexpressed in about 20% of breast cancers. Drugs like trastuzumab, pertuzumab and ado-trastuzumab emtansine (TDM1) specifically target HER2-positive breast cancer, leading to significant improvements in treatment outcomes. Cyclin-dependent kinase 4/6 inhibitors, such as palbociclib, ribociclib and abemaciclib, have transformed the treatment of Hormone Receptor-positive (HR+) metastatic breast cancer. These drugs work by blocking proteins that promote cell division, slowing down cancer growth.

Phosphoinositide 3-Kinase (PI3K) inhibitors, like alpelisib, are used in combination with hormonal therapy to treat certain HR+ breast cancers with PI3K mutations. These targeted therapies help overcome resistance to hormonal treatment. Poly (ADP-ribose) Polymerase (PARP) inhibitors, such as olaparib and talazoparib, are used in the treatment of breast cancer associated with BRCA gene mutations. These drugs interfere with DNA repair mechanisms, leading to cancer cell death. While immunotherapy and targeted therapies hold great promise in the treatment of breast cancer, challenges remain. Not all breast cancer subtypes benefit equally from these treatments and identifying the right patients for these therapies is crucial. Additionally, managing potential side effects and optimizing treatment regimens are ongoing concerns. Future research in breast cancer treatment aims to refine patient selection, develop novel combination therapies and enhance the effectiveness of immunotherapy and targeted treatments [4,5]. Advancements in understanding the tumor microenvironment, immune response and the genetics of breast cancer will drive progress in personalized treatment strategies.

Conclusion

Immunotherapy and targeted therapies have introduced a new era in breast cancer treatment. These innovative approaches offer improved outcomes and reduced side effects compared to traditional treatments, giving hope to patients with advanced and aggressive forms of the disease. As research continues to evolve and refine these therapies, the future holds great promise for breast cancer patients, offering them a brighter prognosis and an enhanced quality of life. In conclusion, immunotherapy is an exciting and evolving approach in the treatment of breast cancer. While there are challenges to overcome, the potential for durable responses and improved quality of life for breast cancer patients is driving ongoing research and clinical trials. With each breakthrough, immunotherapy brings us one step closer to more effective and targeted treatment strategies for this challenging disease.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Lopez-Vilchez, Irene, Maribel Diaz-Ricart, James G. White and Gines Escolar, et al. "Serotonin enhances platelet procoagulant properties and their activation induced during platelet tissue factor uptake." Cardiovasc Res 84 (2009): 309-316.

    Google Scholar, Crossref, Indexed at

  2. Zhao, Aiping, Joseph F. Urban Jr, Motoko Morimoto and Justin E. Elfrey, et al. "Contribution of 5-HT2A receptor in nematode infection-induced murine intestinal smooth muscle hypercontractility." Gastroenterology 131 (2006): 568-578.

    Google Scholar, Crossref, Indexed at

  3. Bhattacharyya, Samarjit, Sapna Puri, Ricardo Miledi and Mitradas M. Panicker. "Internalization and recycling of 5-HT2A receptors activated by serotonin and protein kinase C-mediated mechanisms." Proc Natl Acad Sci99 (2002): 14470-14475.

    Google Scholar, Crossref

  4. Hussain, Mustafa I., Tera L. Reynolds and Kai Zheng. "Medication safety alert fatigue may be reduced via interaction design and clinical role tailoring: A systematic review." J Am Med Inform Assoc 26 (2019): 1141-1149.

    Google Scholar, Crossref, Indexed at

  5. Bazrafshani, Maliheh Sadat, Abbas Pardakhty, Behjat Kalantari Khandani and Naser Nasiri, et al. "The prevalence of polypharmacy and potential drug–drug interactions among Iranian cancer patients: Which patients were at risk?." Cancer Investig 40 (2022): 457-472.

    Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 95

Journal of Cancer Clinical Trials received 95 citations as per Google Scholar report

Journal of Cancer Clinical Trials peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward