Next Article in Journal
The Significance of FDG PET/CT–Derived Parameters in Determining Prognosis of Cases with Pancreatic Adenocarcinoma: A Prospective Study
Next Article in Special Issue
Molecular Pathways Related to Sulforaphane as Adjuvant Treatment: A Nanomedicine Perspective in Breast Cancer
Previous Article in Journal
Plants, Plants, and More Plants: Plant-Derived Nutrients and Their Protective Roles in Cognitive Function, Alzheimer’s Disease, and Other Dementias
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

On the Frontiers of Breast Cancer Diagnosis and Treatment: Current and Future Directions in a Rapidly Changing Field

1
VA Cooperative Studies Program Coordinating Center, Boston, MA 02130, USA
2
Department of Radiation Oncology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
3
Harvard Medical School, Harvard University, Boston, MA 02115, USA
4
Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
*
Author to whom correspondence should be addressed.
Medicina 2022, 58(8), 1026; https://doi.org/10.3390/medicina58081026
Submission received: 24 July 2022 / Accepted: 28 July 2022 / Published: 31 July 2022
(This article belongs to the Collection Frontiers in Breast Cancer Diagnosis and Treatment)
Breast cancer (BCa) represents a medically heterogeneous group of malignancies, with differing biological and genetic makeups [1,2]. This malignancy unequally affects women of different races, ethnicities, and economic backgrounds [3,4]. Because of its diverse prevalence and subtypes, BCa often poses diagnostic and treatment challenges. In previous generations, guidance tended to lack specificity and direction in the management of BCa [5].
The landscape of BCa today has moved well beyond its historic limitations, with the advent of encouraging innovations not only in precision diagnosis and treatment, but most importantly in understanding the biology of the disease. Novel developments in circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) technology (i.e., liquid biopsies) has led to a less-expensive and non-invasive means for predicting disease severity and survival potential in BCa patients [6,7,8]. The refinement of tumor profiling using artificial intelligence and machine learning tools has likewise enabled the prediction of drug response with selected miRNA isoforms [9].
Phenotype and genotype, with the aid of tissue microarray data, currently classifies BCa into distinct subgroups. This reflects the 8th edition of the AJCC cancer staging manual for BCa where, in addition to classic anatomic Tumor, Node and Metastasis (TMN) categories, prognostic groups now incorporate information on grade (i.e., Nottingham histological score), receptor expression (i.e., ER, PR, and HER2 status), and results from a multigene panel assay [10]. With the adoption of these guidelines, BCa staging more accurately reflects the underlying biology and inherent tumor aggressiveness than previously possible.
Inroads in pharmaceutical therapies for BCa have been equally impressive, with the well-tested and currently available inhibitors of CDK4/6 and AKT. Furthermore, recent regulatory approvals include: (1) TRODELVY (sacituzumab govitecan) for triple negative BCa (TNBC); (2) ENHERTU (trastuzumab deruxtecan) for unresectable or metastatic HER2-positive BCa; (3) TUKYSA (tucatinib) for advanced and metastatic HER2-positive BCa, including patients with brain metastases; and (4) PIQRAY (alpelisib) for HR+/HER2 advanced or metastatic BCa with PIK3CA mutations who have progressed after initial aromatase inhibitors [11,12].
Cellular immune function also plays an important but intricate role in BCa. On the treatment side, both immune checkpoint inhibitors (targeting programmed death protein-1/programmed death protein-ligand 1) and tumor-infiltrating lymphocytes have demonstrated clinical benefit in BCa, especially TNBC disease [13,14]. As a potential predictive marker for BCa, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4/CD152), a member of the Immunoglobulin superfamily and negative regulator of T-cell activation, is at the forefront of targeted compounds receiving research attention. Specifically, in a meta-analysis based on 52 case-referent studies and stratified by ethnicity, the single nucleotide polymorphisms CTLA-4 +49 A/G has been associated with a statistically decreased risk of BCa [15].
Another exciting area of progress has been in polymeric nanotechnology [16,17]. Multiple drug resistance and the presentation of unwanted side effects, attributable to inadequate drug concentrations at the tumor site, has limited the effectiveness of various BCa drugs. Nanoscale-based pharmaceuticals, focusing on the tumor microenvironment and tumor disseminate cells, have shown promise for overcoming these limitations. For example, the combined incorporation of quercetin (a naturally occurring flavonoid manifesting antioxidant, anticarcinogenic, anti-inflammatory, antiallergic, and antiviral properties), scorpion venom peptides (known to induce apoptosis), and the proprietary compound Phospholipon 90H into a nano-based delivery system has displayed antiproliferative efficacy in a cell line derived from human BCa MCF-7 cells [18]. This optimized formula resulted in significant cell cycle arrest at the S phase and increased levels of caspase-9, Bax, Bcl-2, and p53 mRNA expression.
Therapeutic advances extend beyond the above-mentioned drugs involving local therapy. Intraoperative radiation therapy (IORT), delivering a single dose of radiation during breast conserving surgery (BCS), has become an accepted alternative to whole breast irradiation (WBI) among patient with early stage BCa [19,20]. Additionally, hypo- and ultra-fractionated radiation after surgery has now become standard of care, without decreasing local control or increasing long term late toxicities [21]. In fact, most modern trials have shown the risk of local failure remains extremely low with a breast conserving approach. There are several ongoing trials testing whether the best prognostic group of BCa patients can avoid adjuvant radiation altogether (NCT03878342) [22], NCT02889874 [23]).
On the surgical frontline, autologous and implant-based breast reconstructions are innovations currently available for the rebuilding of the breast, typically on the same day as surgery [24]. Indeed, oncoplastic breast surgery has become a hallmark in the post-surgical management of BCa patients [25]. Additionally, sentinel lymph node biopsy has replaced morbid full axillary nodal dissection in most cases of early BCa, thereby reducing long-term complications, without decreasing local control.
BCa continues to be the most commonly diagnosed malignant tumor among women worldwide [26]. In spite of advances in the field, the death rate remains high for advanced and metastatic disease. Many questions persist regarding the diagnosis and treatment of this cancer, especially given its intricate molecular subtypes and varying pathologies [27]. In the search for solutions, we expect that research will progress forward with the discovery and implementation of novel targeted compounds and other breakthrough therapies. Early BCa diagnosis will also benefit from research involving innovative imaging techniques and efforts to reduce the cost of (and correspondingly, increase the access to) these methods. The ongoing transition from traditional localized therapy to systemic-based approaches, based on a patient’s molecular and genetic profile, holds promise for future advances that ideally will improve patient survival and quality of life [28].

Author Contributions

Conceptualization, J.T.E. and T.B.; writing—original draft preparation, J.T.E.; writing—review and editing, J.T.E., C.J. and T.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Januškevičienė, I.; Petrikaitė, V. Heterogeneity of breast cancer: The importance of interaction between different tumor cell populations. Life Sci. 2019, 239, 117009. [Google Scholar] [CrossRef]
  2. Krug, K.; Jaehnig, E.J.; Satpathy, S.; Blumenberg, L.; Karpova, A.; Anurag, M.; Miles, G.; Mertins, P.; Geffen, Y.; Tang, L.C.; et al. Proteogenomic Landscape of Breast Cancer Tumorigenesis and Targeted Therapy. Cell 2020, 183, 1436–1456.e1431. [Google Scholar] [CrossRef] [PubMed]
  3. Yedjou, C.G.; Sims, J.N.; Miele, L.; Noubissi, F.; Lowe, L.; Fonseca, D.D.; Alo, R.A.; Payton, M.; Tchounwou, P.B. Health and Racial Disparity in Breast Cancer. Adv. Exp. Med. Biol. 2019, 1152, 31–49. [Google Scholar] [CrossRef] [PubMed]
  4. Prakash, O.; Hossain, F.; Danos, D.; Lassak, A.; Scribner, R.; Miele, L. Racial Disparities in Triple Negative Breast Cancer: A Review of the Role of Biologic and Non-biologic Factors. Front. Public Health 2020, 8, 576964. [Google Scholar] [CrossRef] [PubMed]
  5. Yersal, O.; Barutca, S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014, 5, 412–424. [Google Scholar] [CrossRef]
  6. Davis, A.A.; Zhang, Q.; Gerratana, L.; Shah, A.N.; Zhan, Y.; Qiang, W.; Finkelman, B.S.; Flaum, L.; Behdad, A.; Gradishar, W.J.; et al. Association of a novel circulating tumor DNA next-generating sequencing platform with circulating tumor cells (CTCs) and CTC clusters in metastatic breast cancer. Breast Cancer Res. 2019, 21, 137. [Google Scholar] [CrossRef] [PubMed]
  7. Alix-Panabières, C.; Pantel, K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016, 6, 479–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Kong, S.L.; Liu, X.; Tan, S.J.; Tai, J.A.; Phua, L.Y.; Poh, H.M.; Yeo, T.; Chua, Y.W.; Haw, Y.X.; Ling, W.H.; et al. Complementary Sequential Circulating Tumor Cell (CTC) and Cell-Free Tumor DNA (ctDNA) Profiling Reveals Metastatic Heterogeneity and Genomic Changes in Lung Cancer and Breast Cancer. Front. Oncol. 2021, 11, 698551. [Google Scholar] [CrossRef] [PubMed]
  9. Ogunleye, A.Z.; Piyawajanusorn, C.; Gonçalves, A.; Ghislat, G.; Ballester, P.J. Interpretable Machine Learning Models to Predict the Resistance of Breast Cancer Patients to Doxorubicin from Their microRNA Profiles. Adv. Sci. 2022, e2201501. [Google Scholar] [CrossRef]
  10. Giuliano, A.E.; Edge, S.B.; Hortobagyi, G.N. Eighth Edition of the AJCC Cancer Staging Manual: Breast Cancer. Ann. Surg. Oncol. 2018, 25, 1783–1785. [Google Scholar] [CrossRef]
  11. Wilhoit, T.; Patrick, J.M.; May, M.B. Alpelisib: A Novel Therapy for Patients With PIK3CA-Mutated Metastatic Breast Cancer. J. Adv. Pract. Oncol. 2020, 11, 768–775. [Google Scholar] [CrossRef] [PubMed]
  12. McNulty, R. 2020 Brings 3 Drugs: How the Breast Cancer Treatment Landscape Has Evolved. Available online: https://www.ajmc.com/view/how-the-breast-cancer-treatment-landscape-has-evolved (accessed on 24 July 2022).
  13. Qi, Y.; Zhang, L.; Wang, Z.; Kong, X.; Zhai, J.; Fang, Y.; Wang, J. Efficacy and Safety of Anti-PD-1/ PD-L1 Monotherapy for Metastatic Breast Cancer: Clinical Evidence. Front. Pharmacol. 2021, 12, 653521. [Google Scholar] [CrossRef] [PubMed]
  14. Stanton, S.E.; Disis, M.L. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J. Immunother. Cancer 2016, 4, 59. [Google Scholar] [CrossRef] [Green Version]
  15. Wang, L.; Jiang, Z.; Qiu, H.; Tang, W.; Duan, T.; Wang, L. Associations between CTLA-4 +49 A/G (rs231775) polymorphism and cancer risk: A meta-analysis based on 52 case-control studies. Int. J. Clin. Exp. Med. 2015, 8, 6835–6851. [Google Scholar] [PubMed]
  16. Sartaj, A.; Qamar, Z.; Qizilbash, F.F.; Annu, S.; Alhakamy, N.A.; Baboota, S.; Ali, J. Polymeric Nanoparticles: Exploring the Current Drug Development and Therapeutic Insight of Breast Cancer Treatment and Recommendations. Polymers 2021, 13, 4400. [Google Scholar] [CrossRef]
  17. Rodríguez, F.; Caruana, P.; De la Fuente, N.; Español, P.; Gámez, M.; Balart, J.; Lurba, E.; Rovira, R.; Ruiz, R.; Martín-Lorente, C.; et al. Nano-Based Approved Pharmaceuticals for Cancer Treatment: Present and Future Challenges. Biomolecules 2022, 12, 784. [Google Scholar] [CrossRef]
  18. Alhakamy, N.A.; Fahmy, U.A.; Eldin, S.M.B.; Ahmed, O.A.A.; Aldawsari, H.M.; Okbazghi, S.Z.; Alfaleh, M.A.; Abdulaal, W.H.; Alamoudi, A.J.; Mady, F.M. Scorpion Venom-Functionalized Quercetin Phytosomes for Breast Cancer Management: In Vitro Response Surface Optimization and Anticancer Activity against MCF-7 Cells. Polymers 2021, 14, 93. [Google Scholar] [CrossRef] [PubMed]
  19. Brown, A.; Buss, E.J.; Chin, C.; Liu, G.; Lee, S.; Rao, R.; Taback, B.; Wiechmann, L.; Horowitz, D.; Choi, J.C.; et al. Targeted Intraoperative Radiotherapy (TARGIT-IORT) for Early-Stage Invasive Breast Cancer: A Single Institution Experience. Front. Oncol. 2022, 12, 788213. [Google Scholar] [CrossRef]
  20. Lei, J.; Wang, Y.; Bi, Z.; Xue, S.; Ou, B.; Liu, K. Intraoperative radiotherapy (IORT) versus whole-breast external beam radiotherapy (EBRT) in early stage breast cancer: Results from SEER database. JPN J. Radiol. 2020, 38, 85–92. [Google Scholar] [CrossRef] [PubMed]
  21. Nair, V.J.; Caudrelier, J.M. Hypofractionated radiotherapy for elderly breast cancer patients: From early stages disease to local palliation for unresectable disease. Transl Cancer Res. 2020, 9, S189–S196. [Google Scholar] [CrossRef]
  22. Radiotherapy Omission in Low Risk Ductal in Situ Carcinoma Breast (ROMANCE). Available online: https://clinicaltrials.gov/ct2/show/NCT03878342 (accessed on 24 July 2022).
  23. Examining Personalised Radiation Therapy for Low-Risk Early Breast Cancer (EXPERT). Available online: https://clinicaltrials.gov/ct2/show/NCT02889874 (accessed on 24 July 2022).
  24. Citgez, B.; Yigit, B.; Bas, S. Oncoplastic and Reconstructive Breast Surgery: A Comprehensive Review. Cureus 2022, 14, e21763. [Google Scholar] [CrossRef] [PubMed]
  25. Gilmour, A.; Cutress, R.; Gandhi, A.; Harcourt, D.; Little, K.; Mansell, J.; Murphy, J.; Pennery, E.; Tillett, R.; Vidya, R.; et al. Oncoplastic breast surgery: A guide to good practice. Eur. J. Surg. Oncol. 2021, 47, 2272–2285. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, Y.L.; Ma, Y.; Zeng, Y.Q.; Liu, Y.; He, E.P.; Liu, Y.T.; Qiao, F.L.; Yu, R.; Wang, Y.S.; Wu, X.Y.; et al. A narrative review of research progress on FoxM1 in breast cancer carcinogenesis and therapeutics. Ann. Transl. Med. 2021, 9, 1704. [Google Scholar] [CrossRef] [PubMed]
  27. Li, H.; Li, H. A narrative review of the current landscape and future perspectives of HER2-targeting antibody drug conjugates for advanced breast cancer. Transl. Breast Cancer Res. 2021, 2, 29. [Google Scholar] [CrossRef]
  28. Zhang, L.; Zhang, S.; Yuan, Y.; Li, J.; Li, F.; Geng, C.; Jiang, Z. More than 14 years’ progress free survival of neratinib monotherapy for human epidermal growth factor receptor 2 (HER2) positive advanced breast cancer: A case report. Transl. Breast Cancer Res. 2021, 2, 32. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Efird, J.T.; Jindal, C.; Biswas, T. On the Frontiers of Breast Cancer Diagnosis and Treatment: Current and Future Directions in a Rapidly Changing Field. Medicina 2022, 58, 1026. https://doi.org/10.3390/medicina58081026

AMA Style

Efird JT, Jindal C, Biswas T. On the Frontiers of Breast Cancer Diagnosis and Treatment: Current and Future Directions in a Rapidly Changing Field. Medicina. 2022; 58(8):1026. https://doi.org/10.3390/medicina58081026

Chicago/Turabian Style

Efird, Jimmy T., Charulata Jindal, and Tithi Biswas. 2022. "On the Frontiers of Breast Cancer Diagnosis and Treatment: Current and Future Directions in a Rapidly Changing Field" Medicina 58, no. 8: 1026. https://doi.org/10.3390/medicina58081026

Article Metrics

Back to TopTop