1
|
Ibrahim E, Diab E, Hayek R, Hoyek K, Kourie H. Triple-Negative Breast Cancer: Tumor Immunogenicity and Beyond. Int J Breast Cancer 2024; 2024:2097920. [PMID: 39399414 PMCID: PMC11469932 DOI: 10.1155/2024/2097920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a breast malignancy with a poor prognosis and limited therapeutic options. Many studies show that TNBC exhibits heterogeneity across clinical, histopathological, and molecular levels. In this review, we discuss the immunogenic features of TNBC with a focus on immunotherapy and the current standard of care in the neoadjuvant, adjuvant, and metastatic setting. In addition, we address the ongoing research on immunotherapy, antibody-drug conjugates (ADCs), poly ADP-ribose polymerase (PARP) inhibitors, and future challenges in the treatment of this entity.
Collapse
Affiliation(s)
- Elio Ibrahim
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Ernest Diab
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Rony Hayek
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Karim Hoyek
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Hampig Kourie
- Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
2
|
Eckert KM, Hoskin TL, Olson CA, Goetz MP, Boughey JC. In-Breast Tumor Progression During Neoadjuvant Chemotherapy: Impact on and Factors Influencing Distant Recurrence-Free Survival. Ann Surg Oncol 2024:10.1245/s10434-024-16178-9. [PMID: 39266789 DOI: 10.1245/s10434-024-16178-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/27/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Most patients with breast cancer treated with neoadjuvant chemotherapy (NAC) experience clinical benefit, however, a small proportion progress. We aimed to characterize factors predicting in-breast tumor progression and impact on distant recurrence. PATIENTS AND METHODS We reviewed all patients with clinical stage I-III breast cancer treated with NAC in 2006-2021 at our institution. We compared in-breast progressive disease (PD), defined as ≥ 20% increase in tumor size, with stable disease (SD) or response. Distant recurrence-free survival (DRFS) was analyzed using the Kaplan-Meier method and Cox proportional hazards regression. RESULTS Of 1403 patients, 70 (5%) experienced in-breast PD, 243 (17%) SD, 560 (40%) partial response (PR), and 530 (38%) breast pathologic complete response (breast pCR, ypT0/Tis). The rate of PD varied by tumor subtype (8% in HR+/HER2-, 5% TNBC, 2% HER2+, p < 0.001). With median 48 months follow-up, the rates of DRFS were significantly different according to clinical breast response as follows: PD 56%, SD 68%, PR 82%, or breast pCR 93%, p < 0.001. In patients with PD on multivariable analysis, post-NAC grade (adjusted HR 2.9, p = 0.002) and ypT3-4 category (adjusted HR 2.4, p = 0.03) were the strongest predictors of DRFS. Combining these factors, 23% had neither, 44% had one, and 33% had both, which stratified outcome in PD with 3-year DRFS of 100%, 77%, and 30%, respectively (p < 0.001). CONCLUSIONS While in-breast PD during NAC is uncommon (5%), it predicts poor survival. Among patients with in-breast PD, post-NAC tumor grade and T category predict outcomes and may be useful to guide treatment escalation.
Collapse
Affiliation(s)
- Kathryn M Eckert
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Tanya L Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Carrie A Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Matthew P Goetz
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Judy C Boughey
- Division of Breast and Melanoma Surgical Oncology, Department of Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Zárate-Pérez A, Cruz-Cázares AP, Ordaz-Rosado D, García-Quiroz J, León-Del-Rio A, Avila E, Milo-Rocha E, Díaz L, García-Becerra R. The vitamin D analog EB1089 sensitizes triple-negative breast cancer cells to the antiproliferative effects of antiestrogens. Adv Med Sci 2024; 69:398-406. [PMID: 39233278 DOI: 10.1016/j.advms.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/12/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE Patients bearing estrogen receptor (ER)α-negative breast cancer tumors confront poor prognosis and are typically unresponsive to hormone therapy. Previous studies have shown that calcitriol, the active vitamin D metabolite, can induce ERα expression in ERα-negative cells. EB1089, a calcitriol analog with reduced calcemic effects, exhibits greater potency than calcitriol in inhibiting cancer cell growth. However, the impact of EB1089 on ERα expression in triple-negative breast cancer (TNBC) cells remains unexplored. This study aims to investigate whether EB1089 could induce functional ERα expression in TNBC cell lines, potentially enabling the antiproliferative effects of antiestrogens. MATERIALS AND METHODS TNBC cell lines HCC1806 and HCC1937 were treated with EB1089, and ERα expression was analyzed using real-time PCR and Western blots. The transcriptional activity of induced ERα was evaluated through a luciferase reporter assay. The antiproliferative effects of tamoxifen and fulvestrant antiestrogens were assessed using the sulforhodamine B assay in the EB1089-treated cells. RESULTS Our findings indicated that EB1089 significantly induced ERα mRNA and protein expression in TNBC cells. Moreover, EB1089-induced ERα exhibited transcriptional activity and effectively restored the inhibitory effects of antiestrogens, thereby suppressing cell proliferation in TNBC cells. CONCLUSION EB1089 induced the expression of functional ERα in TNBC cells, restoring the antiproliferative effects of antiestrogens. These results highlight the potential of using EB1089 as a promising strategy for re-establishment of the antiproliferative effect of antiestrogens as a possible management for TNBC. This research lays the foundation for potential advancements in TNBC treatment, offering new avenues for targeted and effective interventions.
Collapse
Affiliation(s)
- Adriana Zárate-Pérez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Alitzin Pamela Cruz-Cázares
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - David Ordaz-Rosado
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Janice García-Quiroz
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Alfonso León-Del-Rio
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Euclides Avila
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Edgar Milo-Rocha
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Lorenza Díaz
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico.
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
4
|
Telli ML, Litton JK, Beck JT, Jones JM, Andersen J, Mina LA, Brig R, Danso M, Yuan Y, Symmans WF, Hopkins JF, Albacker LA, Abbattista A, Noonan K, Mata M, Laird AD, Blum JL. Neoadjuvant talazoparib in patients with germline BRCA1/2 mutation-positive, early-stage triple-negative breast cancer: exploration of tumor BRCA mutational status. Breast Cancer 2024; 31:886-897. [PMID: 38869771 PMCID: PMC11341741 DOI: 10.1007/s12282-024-01603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Talazoparib monotherapy in patients with germline BRCA-mutated, early-stage triple-negative breast cancer (TNBC) showed activity in the neoadjuvant setting in the phase II NEOTALA study (NCT03499353). These biomarker analyses further assessed the mutational landscape of the patients enrolled in the NEOTALA study. METHODS Baseline tumor tissue from the NEOTALA study was tested retrospectively using FoundationOne®CDx. To further hypothesis-driven correlative analyses, agnostic heat-map visualizations of the FoundationOne®CDx tumor dataset were used to assess overall mutational landscape and identify additional candidate predictive biomarkers of response. RESULTS All patients enrolled (N = 61) had TNBC. In the biomarker analysis population, 75.0% (39/52) and 25.0% (13/52) of patients exhibited BRCA1 and BRCA2 mutations, respectively. Strong concordance (97.8%) was observed between tumor BRCA and germline BRCA mutations, and 90.5% (38/42) of patients with tumor BRCA mutations evaluable for somatic-germline-zygosity were predicted to exhibit BRCA loss of heterozygosity (LOH). No patients had non-BRCA germline DNA damage response (DDR) gene variants with known/likely pathogenicity, based on a panel of 14 non-BRCA DDR genes. Ninety-eight percent of patients had TP53 mutations. Genomic LOH, assessed continuously or categorically, was not associated with response. CONCLUSION The results from this exploratory biomarker analysis support the central role of BRCA and TP53 mutations in tumor pathobiology. Furthermore, these data support assessing germline BRCA mutational status for molecular eligibility for talazoparib in patients with TNBC.
Collapse
Affiliation(s)
- Melinda L Telli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Thaddeus Beck
- Department of Medical Oncology and Hematology, Highlands Oncology, Springdale, AR, USA
| | - Jason M Jones
- Avera Medical Group Oncology & Hematology, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Jay Andersen
- Medical Oncology, Compass Oncology, West Cancer Center, US Oncology Network, Tigard, OR, USA
| | - Lida A Mina
- Hematology Oncology Department, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Raymond Brig
- Medical Oncology, Brig Center for Cancer Care and Survivorship, Knoxville, TN, USA
| | - Michael Danso
- Medical Oncology, Virginia Oncology Associates, Norfolk, VA, USA
| | - Yuan Yuan
- Department of Medical Oncology & Therapeutics Research, Cedars-Sinai Cancer Center, West Hollywood, CA, USA
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Kay Noonan
- Clinical Oncology, Pfizer Inc., Groton, CT, USA
| | | | | | - Joanne L Blum
- Department of Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology Network, Dallas, TX, USA
| |
Collapse
|
5
|
Wang M, Pan B, Hu Y, Gao J, Hou L, Yu Z, Li M, Zhao Z. STIL facilitates the development and malignant progression of triple-negative breast cancer through activation of Fanconi anemia pathway via interacting with KLF16. Transl Oncol 2024; 46:102010. [PMID: 38823260 PMCID: PMC11177054 DOI: 10.1016/j.tranon.2024.102010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 05/08/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND STIL is an important cell cycle-regulating protein specifically recruited to the mitotic centrosome to promote the replication of centrioles in dividing cells. However, the potential role of STIL in the regulation of the biological functions of triple-negative breast cancer remains still unclear. METHODS We screened for differentially expressed STIL in the Cancer Genome Atlas database. The expression of STIL protein in 10 pairs of breast cancer tissues and adjacent normal tissues was further assessed by western blotting. Functionally, the knockdown and overexpression of STIL have been used to explore the effects of STIL on breast cancer cell proliferation, migration, and invasion. Mechanistically, RNA-seq, dual-luciferase reporter assay, chromatin immunoprecipitation assay, mass spectrometry, immunoprecipitation assay, and DNA pull-down assay were performed. RESULTS Breast cancer tissues and cells have higher STIL expression than normal tissues and cells. STIL knockdown impairs breast cancer cell growth, migration, and invasion, whereas STIL overexpression accelerates these processes. STIL promotes breast cancer progression by regulating FANCD2 expression, and exploration of its molecular mechanism demonstrated that STIL interacts with KLF16 to regulate the expression of FANCD2. CONCLUSIONS Collectively, our findings identified STIL as a critical promoter of breast cancer progression that interacts with KLF16 to regulate Fanconi anemia pathway protein FANCD2. In summary, STIL is a potential novel biomarker and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Meiling Wang
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Bo Pan
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Ye Hu
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Jiyue Gao
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Lu Hou
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Man Li
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China.
| | - Zuowei Zhao
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China.
| |
Collapse
|
6
|
Yang C, Liu H, Feng X, Shi H, Jiang Y, Li J, Tan J. Research hotspots and frontiers of neoadjuvant therapy in triple-negative breast cancer: a bibliometric analysis of publications between 2002 and 2023. Int J Surg 2024; 110:4976-4992. [PMID: 39143709 PMCID: PMC11326012 DOI: 10.1097/js9.0000000000001586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/25/2024] [Indexed: 08/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer with poor prognosis, and neoadjuvant therapy (NAT) has emerged as an important component in managing advanced-stage patients by providing surgical opportunities and improving survival outcomes. A search of publications on NAT for TNBC from 2002 to 2023 was conducted through the Web of Science core collection. A comprehensive bibliometric analysis was conducted on the data using CiteSpace, VOSviewer, and Bibliometrix. The analysis revealed a continuous and steady growth in the number of articles published in this field over the past 20 years. The United States has made significant contributions to this field, with The University of Texas MD Anderson Cancer Center publishing the most articles. Loibl, S. from Germany was found to be the most published author with 54 articles. Analysis of the journals showed that the Journal of Clinical Oncology is the most cited journal. Combined with the keyword co-occurrence analysis and clustering analysis, current research topic focuses on treatment regimens and disease prognosis. Dual-map overlay of the journals indicates that the research trend is gradually shifting from molecular biology and genetics to immunology and clinical research. Combination therapy, including immunotherapy, may be the future direction for NAT treatment of TNBC. Overall, this study provides valuable insights into the current research status, latest advancements, and emerging development trend of NAT for TNBC.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Hui Liu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University
| | - Xing Feng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
- Department of Hepatobiliary, Breast and Thyroid Surgery, The People's Hospital of Liangping District, Chongqing, China
| | - Han Shi
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Yuchan Jiang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Junfeng Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| | - Jinxiang Tan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University
| |
Collapse
|
7
|
Crespo B, Illera JC, Silvan G, Lopez-Plaza P, Herrera de la Muela M, de la Puente Yague M, Diaz del Arco C, de Andrés PJ, Illera MJ, Caceres S. Bicalutamide Enhances Conventional Chemotherapy in In Vitro and In Vivo Assays Using Human and Canine Inflammatory Mammary Cancer Cell Lines. Int J Mol Sci 2024; 25:7923. [PMID: 39063165 PMCID: PMC11276844 DOI: 10.3390/ijms25147923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Human inflammatory breast cancer (IBC) and canine inflammatory mammary cancer (IMC) are highly aggressive neoplastic diseases that share numerous characteristics. In IBC and IMC, chemotherapy produces a limited pathological response and anti-androgen therapies have been of interest for breast cancer treatment. Therefore, the aim was to evaluate the effect of a therapy based on bicalutamide, a non-steroidal anti-androgen, with doxorubicin and docetaxel chemotherapy on cell proliferation, migration, tumor growth, and steroid-hormone secretion. An IMC-TN cell line, IPC-366, and an IBC-TN cell line, SUM149, were used. In vitro assays revealed that SUM149 exhibited greater sensitivity, reducing cell viability and migration with all tested drugs. In contrast, IPC-366 exhibited only significant in vitro reductions with docetaxel as a single agent or in different combinations. Decreased estrogen levels reduced in vitro tumor growth in both IMC and IBC. Curiously, doxorubicin resulted in low efficacy, especially in IMC. In addition, all drugs reduced the tumor volume in IBC and IMC by increasing intratumoral testosterone (T) levels, which have been related with reduced tumor progression. In conclusion, the addition of bicalutamide to doxorubicin and docetaxel combinations may represent a potential treatment for IMC and IBC.
Collapse
Affiliation(s)
- Belen Crespo
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.C.); (J.C.I.); (P.L.-P.); (M.J.I.); (S.C.)
| | - Juan Carlos Illera
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.C.); (J.C.I.); (P.L.-P.); (M.J.I.); (S.C.)
| | - Gema Silvan
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.C.); (J.C.I.); (P.L.-P.); (M.J.I.); (S.C.)
| | - Paula Lopez-Plaza
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.C.); (J.C.I.); (P.L.-P.); (M.J.I.); (S.C.)
| | - María Herrera de la Muela
- Obstetrics and Gynecology Department, Hospital Clinico San Carlos, Instituto de Salud de la Mujer, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IsISSC), 28040 Madrid, Spain;
| | - Miriam de la Puente Yague
- Department of Public and Maternal Child Health University, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| | | | - Paloma Jimena de Andrés
- Department of Animal Medicine, Surgery and Pathology, Veterinary Medicine School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Maria Jose Illera
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.C.); (J.C.I.); (P.L.-P.); (M.J.I.); (S.C.)
| | - Sara Caceres
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.C.); (J.C.I.); (P.L.-P.); (M.J.I.); (S.C.)
| |
Collapse
|
8
|
Jia X, Wang K, Zhuo Q, Zhao Z, Li M. PARP Inhibitor for Neoadjuvant Therapy in HER2-Negative Breast Cancer: A Systematic Review and Meta-Analysis of Efficacy and Safety. Clin Breast Cancer 2024; 24:392-398.e3. [PMID: 38580572 DOI: 10.1016/j.clbc.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/23/2024] [Accepted: 02/25/2024] [Indexed: 04/07/2024]
Abstract
Poly-ADP ribose polymerase inhibitor (PARPi) is approved for HER2-negative advanced breast cancer with BRCA1/2 mutation. In recent years, many studies have explored the application of PARPi in neoadjuvant therapy, but failed to reach a unified conclusion. PubMed, Clinicaltrials.gov, Cochrane CENTRAL, Embase, and key oncological meetings for trials were searched for studies reporting neoadjuvant regimens with PARPi in HER2-negative breast cancer. Pathological complete response (pCR), residual cancer burden (RCB), breast-conservation surgery rate (BCSR), clinical response, and adverse events were extracted and pooled in a meta-analysis using the Mantel Haenszel random/fixed effects model. Subgroup analyses of pCR were conducted according to BRCA1/2 status, and hormone receptor (HR) status. Five studies (N = 1223) were included, the addition of PARPi to neoadjuvant regimens significantly increased pCR rates (HR 1.45, 95%CI 1.09-1.92, P = .01, I2 = 86%). In subgroup analysis, the addition of PARPi increased the pCR rate both in HR-positive (n = 383) and HR-negative (n = 431) subgroups, which showed a dominant effect of PARPi regardless of HR status (HR 2.07, 95%CI 1.33-3.23, P = .001, I2 = 0%; HR 1.85, 95%CI 1.39-2.26, P < .0001, I2 = 0%, respectively). However, when we performed a subgroup analysis based on the status of BRCA1/2, no further benefit for PARPi was found. Adverse reactions were generally tolerable. Other outcome indexes, including RCB, clinical response, BCSR, and PARPi did not show a clinical benefit. Regardless of BRCA1/2 status, PARPi in neoadjuvant therapy, can improve the pCR rate of HER2-negative breast cancer, especially in HR-positive patients. Thus, we should have performed larger randomized trials and provided a stronger evidence-based basis.
Collapse
Affiliation(s)
- Xiaomeng Jia
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kainan Wang
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiping Zhuo
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zuowei Zhao
- Department of Breast Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Man Li
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
9
|
Yang L, Hu Q, Huang T. Breast Cancer Treatment Strategies Targeting the Tumor Microenvironment: How to Convert "Cold" Tumors to "Hot" Tumors. Int J Mol Sci 2024; 25:7208. [PMID: 39000314 PMCID: PMC11241188 DOI: 10.3390/ijms25137208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Breast cancer characterized as "cold tumors" exhibit low levels of immune cell infiltration, which limits the efficacy of conventional immunotherapy. Recent studies have focused on strategies using nanotechnology combined with tumor microenvironment modulation to transform "cold tumors" into "hot tumors". This approach involves the use of functionalized nanoparticles that target and modify the tumor microenvironment to promote the infiltration and activation of antitumor immune cells. By delivering immune activators or blocking immunosuppressive signals, these nanoparticles activate otherwise dormant immune responses, enhancing tumor immunogenicity and the therapeutic response. These strategies not only promise to increase the response rate of breast cancer patients to existing immunotherapies but also may pave new therapeutic avenues, providing a new direction for the immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Liucui Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
10
|
Katkova SA, Bunev AS, Gasanov RE, Khochenkov DA, Kulsha AV, Ivashkevich OA, Serebryanskaya TV, Kinzhalov MA. Metal-(Acyclic Diaminocarbene) Complexes Demonstrate Nanomolar Antiproliferative Activity against Triple-Negative Breast Cancer. Chemistry 2024; 30:e202400101. [PMID: 38363795 DOI: 10.1002/chem.202400101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/18/2024]
Abstract
Hydrolytically stable PdII and PtII complexes supported by acyclic diaminocarbene ligands represent a novel class of structural organometallic anticancer agents exhibiting nanomolar antiproliferative activity in a panel of cancer cell lines (IC50 0.07-0.81 μM) and up to 300-fold selectivity for cancer cells over normal primary fibroblasts. The lead drug candidate was 300 times more potent than cisplatin in vitro and showed higher efficacy in reducing the growth of aggressive MDA-MB-231 xenograft tumors in mice.
Collapse
Affiliation(s)
- Svetlana A Katkova
- Saint Petersburg State University, 7/9 Universitetskaya Nab., St. Petersburg, 199034, Russian Federation
| | - Alexander S Bunev
- Medicinal Chemistry Center, Togliatti State University, Belorusskaya 14, Togliatti, 445020, Russian Federation
| | - Rovshan E Gasanov
- Medicinal Chemistry Center, Togliatti State University, Belorusskaya 14, Togliatti, 445020, Russian Federation
| | - Dmitry A Khochenkov
- Medicinal Chemistry Center, Togliatti State University, Belorusskaya 14, Togliatti, 445020, Russian Federation
- Blokhin National Medical Research Center of Oncology, Kashirskoe Shosse 24, 115478, Moscow, Russian Federation
| | - Andrey V Kulsha
- Department of Chemistry, Belarusian State University, Leningradskaya 14, 220006, Minsk, Belarus
| | - Oleg A Ivashkevich
- Research Institute for Physical Chemical Problems, Belarusian State University, Leningradskaya 14, 220006, Minsk, Belarus
| | - Tatiyana V Serebryanskaya
- Research Institute for Physical Chemical Problems, Belarusian State University, Leningradskaya 14, 220006, Minsk, Belarus
| | - Mikhail A Kinzhalov
- Saint Petersburg State University, 7/9 Universitetskaya Nab., St. Petersburg, 199034, Russian Federation
| |
Collapse
|
11
|
Zhao D, Wu T, Tan Z, Xu J, Lu Z. Role of non-coding RNAs mediated pyroptosis on cancer therapy: a review. Expert Rev Anticancer Ther 2024; 24:239-251. [PMID: 38594965 DOI: 10.1080/14737140.2024.2341737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
INTRODUCTION Non-coding RNAs (ncRNAs), which are incapable of encoding proteins, are involved in the progression of numerous tumors by altering transcriptional and post-transcriptional processing. Recent studies have revealed prominent features of ncRNAs in pyroptosis, a type of non-apoptotic programmed cellular destruction linked to an inflammatory reaction. Drug resistance has arisen gradually as a result of anti-apoptotic proteins, therefore strategies based on pyroptotic cell death have attracted increasing attention. We have observed that ncRNAs may exert significant influence on cancer therapy, chemotherapy, radio- therapy, targeted therapy and immunotherapy, by regulating pyroptosis. AREAS COVERED Literatures were searched (December 2023) for studies on cancer therapy for ncRNAs-mediated pyroptotic cell death. EXPERT OPINION The most universal mechanical strategy for ncRNAs to regulate target genes is competitive endogenous RNAs (ceRNA). Besides, certain ncRNAs could directly interact with proteins and modulate downstream genes to induce pyroptosis, resulting in tumor growth or inhibition. In this review, we aim to display that ncRNAs, predominantly long non-coding RNAs (lncRNAs), microRNAs (miRNAs) and circular RNAs (circRNAs), could function as potential biomarkers for diagnosis and prognosis and produce new insights into anti-cancer strategies modulated by pyroptosis for clinical applications.
Collapse
Affiliation(s)
- Dan Zhao
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tangwei Wu
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheqiong Tan
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Xu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongxin Lu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Research Institute of Wuhan, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Acs B, Hartman J, Sönmez D, Lindman H, Johansson AL, Fredriksson I. Real-world overall survival and characteristics of patients with ER-zero and ER-low HER2-negative breast cancer treated as triple-negative breast cancer: a Swedish population-based cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2024; 40:100886. [PMID: 38745990 PMCID: PMC11092884 DOI: 10.1016/j.lanepe.2024.100886] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 05/16/2024]
Abstract
Background Estrogen receptor-low (ER-low) HER2-negative breast cancer has similar pathological and molecular characteristics as triple-negative breast cancer (TNBC), and it is questionable whether it should be considered a separate entity. When the international guidelines lowered the cutoff for ER positivity to ≥1% in 2010, the ≥10% threshold was kept in Sweden. ER-low breast cancer (ER 1-9%) is thus in Sweden treated as TNBC. We aimed to describe patient and tumor characteristics, treatment patterns and overall survival in a Swedish population-based cohort of patients with ER-zero and ER-low HER2-negative breast cancer treated as TNBC. Methods All TNBC cases diagnosed in Sweden 2008-2020 were included in a population-based cohort study. Patient, tumor and treatment characteristics were analyzed by ER-status (ER 0% vs 1-9%), and associations between subgroups compared using χ2 test. Survival endpoint was overall survival (OS), and Kaplan-Meier curves were estimated. Cox proportional hazards models were used to estimate adjusted hazard ratios comparing ER-low to ER-zero. Findings Of the 5655 tumors, 90.1% had an ER expression of 0%, while 9.9% were ER-low. ER-low tumors were grade III in 69.4% (80.8% in ER-zero tumors, p-value = 0.001), with a median Ki67 of 60% (63% in ER-zero tumors, p-value = 0.005). There were no significant differences in given chemotherapy (p = 0.546). A pathological complete response (pCR) was achieved in 28.1% of ER-low tumors (25.1% in ER-zero tumors). In the unadjusted analysis of OS, women with ER-low disease had a borderline but not significantly better OS than those with ER-zero disease (HR 0.84 (95% CI 0.71-1.00), p = 0.052). ER-status 1-9% vs 0% was not associated with OS in the multivariable analysis (HR 1.11 (0.90-1.36)). Distant disease-free survival did not differ by ER-status 0% vs 1-9% (HR 0.97 for ER-zero vs ER-low (0.62-1.53), p = 0.905). After preoperative treatment, the impact of pCR for OS did not significantly differ between ER-zero or ER-low disease. Interpretation ER-low HER2-negative breast cancer has characteristics and prognosis similar to TNBC, when treated in the same way. Therefore, it seems reasonable to use a ≥10% threshold for ER positivity. This would provide patients with ER-low tumors the same treatment opportunities as patients with TNBC, within studies and within clinical routine. Funding This work was financially supported by Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA, in accordance with terms and conditions of a Master Collaboration Agreement between the company and Karolinska Institutet.
Collapse
Affiliation(s)
- Balazs Acs
- Dept of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Dept of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Hartman
- Dept of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Dept of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Demet Sönmez
- Center for Observational and Real-World Evidence, MSD, Stockholm, Sweden
| | - Henrik Lindman
- Dept of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Dept of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Anna L.V. Johansson
- Dept of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Irma Fredriksson
- Dept of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Dept of Breast-, Endocrine Tumors and Sarcoma, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Trabulus FDC, Nazli MA, Arslan E, Mermut O, Dal F, Akce B, Gursu RU, Talu ECK, Couteau JNA. Predictors of recurrence in breast cancer patients with pathological partial response. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20231215. [PMID: 38656005 DOI: 10.1590/1806-9282.20231215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 04/26/2024]
Abstract
OBJECTIVE Patients with residual disease after neoadjuvant chemotherapy have a relative risk of developing recurrence. This study investigates the risk factors for recurrence in locally advanced breast cancer patients with residual disease and evaluates survival analysis. METHODS This is a retrospective, single-center study. Breast cancer patients who failed to achieve a pathological complete response after neoadjuvant chemotherapy were included. Demographic, clinicopathological, and treatment characteristics were evaluated to identify predictive factors of recurrence and survival analysis. RESULTS We included 205 patients in this study. After a median of 31 months of follow-up, 10 patients died, and 20 developed distant metastasis. Disease-free survival and disease-specific survival were 73.8% and 83.1%, respectively. Lymphovascular invasion and non-luminal subtype were independent predictors of locoregional recurrence. In situ carcinoma, lymphovascular invasion, ypTIII stage, and non-luminal molecular subtypes were independent predictors of disease-free survival. The only independent factor affecting disease-specific survival was cNII-III. The number of involved lymph nodes was an independent predictor of disease-free survival in patients without complete axillary response. CONCLUSION Factors affecting disease-specific survival and disease-free survival were cNII-III and the number of involved lymph nodes, respectively. Patients with non-luminal, large residual tumors with in situ carcinoma, lymphovascular invasion, clinically positive axilla, and residual nodal involvement have a high relative risk for recurrence and may benefit from additional treatments.
Collapse
Affiliation(s)
- Fadime Didem Can Trabulus
- Bahçeşehir University, School of Medicine, Goztepe Medical Park Hospital, Department of General Surgery - İstanbul, Turkey
| | - Mehmet Ali Nazli
- University of Health Sciences, Istanbul Training and Education Hospital, Department of Radiology - İstanbul, Turkey
| | - Esra Arslan
- University of Health Sciences, Istanbul Training and Education Hospital, Department of Nuclear Medicine - İstanbul, Turkey
| | - Ozlem Mermut
- University of Health Sciences, Istanbul Training and Education Hospital, Department of Radiation Oncology - İstanbul, Turkey
| | - Fatih Dal
- University of Health Sciences, Istanbul Training and Education Hospital, Department of General Surgery - İstanbul, Turkey
| | - Bulent Akce
- University of Health Sciences, Istanbul Training and Education Hospital, Department of General Surgery - İstanbul, Turkey
| | - Riza Umar Gursu
- University of Health Sciences, Istanbul Training and Education Hospital, Department of Medical Oncology - İstanbul, Turkey
| | - Esra Canan Kelten Talu
- University of Health Sciences, İzmir Faculty of Medicine, Department of Pathology - İzmir, Turkey
| | | |
Collapse
|
14
|
Huang M, Fasching PA, Haiderali A, Xue W, Pan W, Karantza V, Yang F, Truscott J, Xin Y, O'Shaughnessy J. Association between event-free survival and overall survival in early-stage triple-negative breast cancer. Future Oncol 2024; 20:335-348. [PMID: 37602372 DOI: 10.2217/fon-2023-0315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023] Open
Abstract
Aim: This study evaluated event-free survival (EFS) as a surrogate outcome for overall survival (OS) in neoadjuvant therapy for early-stage triple-negative breast cancer (eTNBC). Methods: Meta-regression analyses based on a targeted literature review were used to evaluate the individual- and trial-level associations between EFS and OS. Results: In the individual-level analyses, 3-year EFS was a significant predictor of 5-year OS (p < 0.01; coefficient of determinations [R2]: 0.82 [95% CI: 0.68-0.91]). Additionally, there was a statistically significant association between the treatment effect on EFS and OS at the trial level (p < 0.001; R2: 0.64 [95% CI: 0.45-0.82]). Conclusion: This study demonstrates significant associations between EFS and OS and suggests that EFS is a valid surrogate for OS following neoadjuvant therapy for eTNBC.
Collapse
Affiliation(s)
| | - Peter A Fasching
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Department of Gynecology & Obstetrics, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology & US Oncology, Dallas, TX, USA
| |
Collapse
|
15
|
Bhattarai S, Saini G, Li H, Seth G, Fisher TB, Janssen EAM, Kiraz U, Kong J, Aneja R. Predicting Neoadjuvant Treatment Response in Triple-Negative Breast Cancer Using Machine Learning. Diagnostics (Basel) 2023; 14:74. [PMID: 38201383 PMCID: PMC10871101 DOI: 10.3390/diagnostics14010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) is the standard treatment for early-stage triple negative breast cancer (TNBC). The primary endpoint of NAC is a pathological complete response (pCR). NAC results in pCR in only 30-40% of TNBC patients. Tumor-infiltrating lymphocytes (TILs), Ki67 and phosphohistone H3 (pH3) are a few known biomarkers to predict NAC response. Currently, systematic evaluation of the combined value of these biomarkers in predicting NAC response is lacking. In this study, the predictive value of markers derived from H&E and IHC stained biopsy tissue was comprehensively evaluated using a supervised machine learning (ML)-based approach. Identifying predictive biomarkers could help guide therapeutic decisions by enabling precise stratification of TNBC patients into responders and partial or non-responders. METHODS Serial sections from core needle biopsies (n = 76) were stained with H&E and immunohistochemically for the Ki67 and pH3 markers, followed by whole-slide image (WSI) generation. The serial section stains in H&E stain, Ki67 and pH3 markers formed WSI triplets for each patient. The resulting WSI triplets were co-registered with H&E WSIs serving as the reference. Separate mask region-based CNN (MRCNN) models were trained with annotated H&E, Ki67 and pH3 images for detecting tumor cells, stromal and intratumoral TILs (sTILs and tTILs), Ki67+, and pH3+ cells. Top image patches with a high density of cells of interest were identified as hotspots. Best classifiers for NAC response prediction were identified by training multiple ML models and evaluating their performance by accuracy, area under curve, and confusion matrix analyses. RESULTS Highest prediction accuracy was achieved when hotspot regions were identified by tTIL counts and each hotspot was represented by measures of tTILs, sTILs, tumor cells, Ki67+, and pH3+ features. Regardless of the hotspot selection metric, a complementary use of multiple histological features (tTILs, sTILs) and molecular biomarkers (Ki67 and pH3) resulted in top ranked performance at the patient level. CONCLUSIONS Overall, our results emphasize that prediction models for NAC response should be based on biomarkers in combination rather than in isolation. Our study provides compelling evidence to support the use of ML-based models to predict NAC response in patients with TNBC.
Collapse
Affiliation(s)
- Shristi Bhattarai
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.B.); (G.S.); (G.S.)
| | - Geetanjali Saini
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.B.); (G.S.); (G.S.)
| | - Hongxiao Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30302, USA;
| | - Gaurav Seth
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.B.); (G.S.); (G.S.)
| | - Timothy B. Fisher
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA;
| | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.A.M.J.); (U.K.)
- Department of Chemistry, Bioscience and Environmental Engineering, Stavanger University, 4021 Stavanger, Norway
| | - Umay Kiraz
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.A.M.J.); (U.K.)
- Department of Chemistry, Bioscience and Environmental Engineering, Stavanger University, 4021 Stavanger, Norway
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30302, USA;
| | - Ritu Aneja
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.B.); (G.S.); (G.S.)
| |
Collapse
|
16
|
Haif SK, Al Kury LT, Talib WH. Combination of Thymoquinone and Intermittent Fasting as a Treatment for Breast Cancer Implanted in Mice. PLANTS (BASEL, SWITZERLAND) 2023; 13:35. [PMID: 38202341 PMCID: PMC10780740 DOI: 10.3390/plants13010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
Breast cancer stands out as a particularly challenging form of cancer to treat among various types. Traditional treatment methods have been longstanding approaches, yet their efficacy has diminished over time owing to heightened toxicity, adverse effects, and the emergence of multi-drug resistance. Nevertheless, a viable solution has emerged through the adoption of a complementary treatment strategy utilizing natural substances and the incorporation of intermittent fasting to enhance therapeutic outcomes. This study aimed to assess the anticancer activity of thymoquinone (TQ), intermittent fasting, and their combination using in vivo and in vitro methods. The anti-proliferative activity of TQ and fasting (glucose/serum restriction) were evaluated against the T47D, MDA-MB-231, and EMT6 cell lines and compared to normal cell lines (Vero) using the MTT colorimetric assay method. Additionally, this study aimed to determine the half-maximal inhibitory concentration (IC50) of TQ. For the in vivo experiment, the antitumor activity of TQ and intermittent fasting (IF) was assessed by measuring the tumor sizes using a digital caliper to determine the change in the tumor size and survival rates. At the molecular level, the serum levels of glucose, β-hydroxybutyrate (β-HB), leptin, and insulin growth factor-1 (IGF-1) were measured using standard kits. Additionally, the aspartate transaminase (AST), alanine transaminase (ALT), and creatinine serum levels were measured. The inhibition of the breast cancer cell lines was achieved by TQ. TQ and intermittent fasting both had an additional anticancer effect against breast tumors inoculated in mice. The combination therapy was evaluated and found to significantly reduce the tumor size, with a change in tumor size of -57.7%. Additionally, the combination of TQ and IF led to a decrease in the serum levels of glucose, IGF-1 (24.49 ng/mL) and leptin (1.77 ng/mL) while increasing β-hydroxybutyrate in the mice given combination therapy (200.86 nM) with no toxicity on the liver or kidneys. In the mice receiving combination therapy, TQ and IF treated breast cancer in an additive way without causing liver or kidney toxicity due to decreased levels of glucose, IGF-1, and leptin and increased levels of β-hydroxybutyrate. Further investigation is required to optimize the doses and determine the other possible mechanisms exhibited by the novel combination.
Collapse
Affiliation(s)
- Shatha Khaled Haif
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931-166, Jordan;
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates
| | - Wamidh H. Talib
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman 11931-166, Jordan
| |
Collapse
|
17
|
Barker VR, Naffouje SA, Mallory MA, Hoover SA, Laronga C. Surgical Management of the Axilla in HR+/HER2- Breast Cancer in the Z1071 Era: A Propensity Score-Matched Analysis of the National Cancer Database. Ann Surg Oncol 2023; 30:8371-8380. [PMID: 37610487 DOI: 10.1245/s10434-023-14029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/10/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Axillary management varies between sentinel lymph node biopsy (SLNB) and axillary lymph node dissection (ALND) for patients with clinical N1 (cN1), hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)/neu-negative (HER2-), infiltrative ductal carcinoma (IDC) who achieve a complete clinical response (cCR) to neoadjuvant systemic therapy (NAST). This study sought to evaluate clinical practice patterns and survival outcomes of SLNB versus ALND in this patient subset. METHODS Patients with cN1, HR+/HER2-, unilateral IDC demonstrating a cCR to NAST were identified from the 2012-2017 National Cancer Database (NCDB) and stratified based on final axillary surgery management (SLNB vs ALND). After propensity score-matching, overall survival (OS) was compared using a Kaplan-Meier analysis, and significant OS predictors were identified using Cox regression. RESULTS Of the 1676 patients selected for this study, 593 (35.4%) underwent SLNB and 1083 (64.6%) underwent ALND. Use of SLNB increased by 28 % between 2012 and 2017. Among a total of 584 matched patients, 461 matched ypN0 patients, and 108 matched ypN+ patients, mean OS did not differ between SLNB and ALND (all patients [92.1 ± 0.8 vs 90.2 ± 1.0 months; p = 0.157], ypN0 patients [92.4 ± 0.8 vs 89.9 ± 0.9 months; p = 0.105], ypN+ patients [83.5 ± 2.3 vs 91.7 ± 2.7 months; p ± 0.963). Cox regression identified age, Charlson score, clinical T stage, and pathologic nodal status as significant predictors of OS. CONCLUSION The final surgical management strategy used for cN1, HR+/HER2- IDC patients who achieved a cCR to NAST did not have a significant impact on survival outcomes in this analysis. Potential opportunities for de-escalation of axillary management among this patient subset exist, and validation studies are needed.
Collapse
Affiliation(s)
- Vayda R Barker
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Samer A Naffouje
- Department of Surgical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Melissa A Mallory
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Susan A Hoover
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Christine Laronga
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
18
|
Masci D, Naro C, Puxeddu M, Urbani A, Sette C, La Regina G, Silvestri R. Recent Advances in Drug Discovery for Triple-Negative Breast Cancer Treatment. Molecules 2023; 28:7513. [PMID: 38005235 PMCID: PMC10672974 DOI: 10.3390/molecules28227513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most heterogeneous and aggressive breast cancer subtypes with a high risk of death on recurrence. To date, TNBC is very difficult to treat due to the lack of an effective targeted therapy. However, recent advances in the molecular characterization of TNBC are encouraging the development of novel drugs and therapeutic combinations for its therapeutic management. In the present review, we will provide an overview of the currently available standard therapies and new emerging therapeutic strategies against TNBC, highlighting the promises that newly developed small molecules, repositioned drugs, and combination therapies have of improving treatment efficacy against these tumors.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Chiara Naro
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Claudio Sette
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| |
Collapse
|
19
|
Saleh GA, Batouty NM, Gamal A, Elnakib A, Hamdy O, Sharafeldeen A, Mahmoud A, Ghazal M, Yousaf J, Alhalabi M, AbouEleneen A, Tolba AE, Elmougy S, Contractor S, El-Baz A. Impact of Imaging Biomarkers and AI on Breast Cancer Management: A Brief Review. Cancers (Basel) 2023; 15:5216. [PMID: 37958390 PMCID: PMC10650187 DOI: 10.3390/cancers15215216] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/13/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer stands out as the most frequently identified malignancy, ranking as the fifth leading cause of global cancer-related deaths. The American College of Radiology (ACR) introduced the Breast Imaging Reporting and Data System (BI-RADS) as a standard terminology facilitating communication between radiologists and clinicians; however, an update is now imperative to encompass the latest imaging modalities developed subsequent to the 5th edition of BI-RADS. Within this review article, we provide a concise history of BI-RADS, delve into advanced mammography techniques, ultrasonography (US), magnetic resonance imaging (MRI), PET/CT images, and microwave breast imaging, and subsequently furnish comprehensive, updated insights into Molecular Breast Imaging (MBI), diagnostic imaging biomarkers, and the assessment of treatment responses. This endeavor aims to enhance radiologists' proficiency in catering to the personalized needs of breast cancer patients. Lastly, we explore the augmented benefits of artificial intelligence (AI), machine learning (ML), and deep learning (DL) applications in segmenting, detecting, and diagnosing breast cancer, as well as the early prediction of the response of tumors to neoadjuvant chemotherapy (NAC). By assimilating state-of-the-art computer algorithms capable of deciphering intricate imaging data and aiding radiologists in rendering precise and effective diagnoses, AI has profoundly revolutionized the landscape of breast cancer radiology. Its vast potential holds the promise of bolstering radiologists' capabilities and ameliorating patient outcomes in the realm of breast cancer management.
Collapse
Affiliation(s)
- Gehad A. Saleh
- Diagnostic and Interventional Radiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (G.A.S.)
| | - Nihal M. Batouty
- Diagnostic and Interventional Radiology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (G.A.S.)
| | - Abdelrahman Gamal
- Computer Science Department, Faculty of Computers and Information, Mansoura University, Mansoura 35516, Egypt (A.E.T.)
| | - Ahmed Elnakib
- Electrical and Computer Engineering Department, School of Engineering, Penn State Erie, The Behrend College, Erie, PA 16563, USA;
| | - Omar Hamdy
- Surgical Oncology Department, Oncology Centre, Mansoura University, Mansoura 35516, Egypt;
| | - Ahmed Sharafeldeen
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Ali Mahmoud
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Mohammed Ghazal
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates; (M.G.)
| | - Jawad Yousaf
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates; (M.G.)
| | - Marah Alhalabi
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates; (M.G.)
| | - Amal AbouEleneen
- Computer Science Department, Faculty of Computers and Information, Mansoura University, Mansoura 35516, Egypt (A.E.T.)
| | - Ahmed Elsaid Tolba
- Computer Science Department, Faculty of Computers and Information, Mansoura University, Mansoura 35516, Egypt (A.E.T.)
- The Higher Institute of Engineering and Automotive Technology and Energy, New Heliopolis, Cairo 11829, Egypt
| | - Samir Elmougy
- Computer Science Department, Faculty of Computers and Information, Mansoura University, Mansoura 35516, Egypt (A.E.T.)
| | - Sohail Contractor
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA
| | - Ayman El-Baz
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
20
|
Litton JK, Beck JT, Jones JM, Andersen J, Blum JL, Mina LA, Brig R, Danso M, Yuan Y, Abbattista A, Noonan K, Niyazov A, Chakrabarti J, Czibere A, Symmans WF, Telli ML. Neoadjuvant Talazoparib in Patients With Germline BRCA1/2 Mutation-Positive, Early-Stage Triple-Negative Breast Cancer: Results of a Phase II Study. Oncologist 2023; 28:845-855. [PMID: 37318349 PMCID: PMC10546823 DOI: 10.1093/oncolo/oyad139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/14/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND The undetermined efficacy of the current standard-of-care neoadjuvant treatment, anthracycline/platinum-based chemotherapy, in patients with early-stage triple-negative breast cancer (TNBC) and germline BRCA mutations emphasizes the need for biomarker-targeted treatment, such as poly(ADP-ribose) polymerase inhibitors, in this setting. This phase II, single-arm, open-label study evaluated the efficacy and safety of neoadjuvant talazoparib in patients with germline BRCA1/2-mutated early-stage TNBC. PATIENTS AND METHODS Patients with germline BRCA1/2-mutated early-stage TNBC received talazoparib 1 mg once daily for 24 weeks (0.75 mg for moderate renal impairment) followed by surgery. The primary endpoint was pathologic complete response (pCR) by independent central review (ICR). Secondary endpoints included residual cancer burden (RCB) by ICR. Safety and tolerability of talazoparib and patient-reported outcomes were assessed. RESULTS Of 61 patients, 48 received ≥80% talazoparib doses, underwent surgery, and were assessed for pCR or progressed before pCR assessment and considered nonresponders. pCR rate was 45.8% (95% confidence interval [CI], 32.0%-60.6%) and 49.2% (95% CI, 36.7%-61.6%) in the evaluable and intent-to-treat (ITT) population, respectively. RCB 0/I rate was 45.8% (95% CI, 29.4%-63.2%) and 50.8% (95% CI, 35.5%-66.0%) in the evaluable and ITT population, respectively. Treatment-related adverse events (TRAE) were reported in 58 (95.1%) patients. Most common grade 3 and 4 TRAEs were anemia (39.3%) and neutropenia (9.8%). There was no clinically meaningful detriment in quality of life. No deaths occurred during the reporting period; 2 deaths due to progressive disease occurred during long-term follow-up (>400 days after first dose). CONCLUSIONS Neoadjuvant talazoparib monotherapy was active despite pCR rates not meeting the prespecified threshold; these rates were comparable to those observed with combination anthracycline- and taxane-based chemotherapy regimens. Talazoparib was generally well tolerated. CLINICALTRIALS.GOV IDENTIFIER NCT03499353.
Collapse
Affiliation(s)
- Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Thaddeus Beck
- Department of Medical Oncology and Hematology, Highlands Oncology, Springdale, AR, USA
| | - Jason M Jones
- Avera Medical Group Oncology & Hematology, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Jay Andersen
- Medical Oncology, Compass Oncology, West Cancer Center, US Oncology Network, Tigard, OR, USA
| | - Joanne L Blum
- Department of Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology Network, Dallas, TX, USA
| | - Lida A Mina
- Hematology Oncology Department, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Raymond Brig
- Medical Oncology, Brig Center for Cancer Care and Survivorship, Knoxville, TN, USA
| | - Michael Danso
- Medical Oncology, Virginia Oncology Associates, Norfolk, VA, USA
| | - Yuan Yuan
- Department of Medical Oncology & Therapeutics Research, Cedars-Sinai Cancer Center, West Hollywood, CA, USA
| | | | - Kay Noonan
- Clinical Oncology, Pfizer Inc., Groton, CT, USA
| | | | | | - Akos Czibere
- Oncology Drug Development, Pfizer Inc., Cambridge, MA, USA
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melinda L Telli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
21
|
El Gazzar WB, Albakri KA, Hasan H, Badr AM, Farag AA, Saleh OM. Poly(ADP-ribose) polymerase inhibitors in the treatment landscape of triple-negative breast cancer (TNBC). J Oncol Pharm Pract 2023; 29:1467-1479. [PMID: 37559370 DOI: 10.1177/10781552231188903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
OBJECTIVE Chemotherapy is the mainstay for triple-negative breast cancer (TNBC) patients. Over the years, the use of chemotherapy for these patients has demonstrated many adversities, including toxicity and resistance, which suggested the need to develop novel alternative therapeutic options, such as poly(ADP-ribose) polymerase inhibitors (PARPi). Herein, we provide an overview on PARPi, mechanisms of action and the role of biomarkers in PARPi sensitivity trials, clinical advances in PARPi therapy for TNBC patients based on the most recent studies and findings of clinical trials, and challenges that prevent PARP inhibitors from achieving high efficacy such as resistance and overlapping toxicities with other chemotherapies. DATA SOURCES Searching for relevant articles was done using PubMed and Cochrane Library databases by using the keywords including TNBC; chemotherapy; PARPi; BRCA; homologous recombination repair (HRR). Studies had to be published in full-text in English in order to be considered. DATA SUMMARY Although PARPi have been used in the treatment of local/metastatic breast malignancies that are HER2 negative and has a germline BRCA mutation, several questions are still to be answered in order to maximize the clinical benefit of PARP inhibitors in TNBC treatment, such as questions related to the optimal use in the neoadjuvant and metastatic settings as well as the best combinations with various chemotherapies. CONCLUSIONS PARPi are emerging treatment options for patients with gBRCA1/2 mutations. Determining patients that are most likely to benefit from PARPi and identifying the optimal treatment combinations with high efficacy and fewer side effects are currently ongoing.
Collapse
Affiliation(s)
- Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha City, Egypt
| | | | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Amira M Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amina A Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha City, Egypt
| | | |
Collapse
|
22
|
Jeong KY, Park SY, Park MH, Kim HM. Suppressing Src-Mediated EGFR Signaling by Sustained Calcium Supply Targeting Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:13291. [PMID: 37686097 PMCID: PMC10488068 DOI: 10.3390/ijms241713291] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Src is emerging as a promising target in triple-negative breast cancer (TNBC) treatment because it activates survival signaling linked to the epidermal growth factor receptor. In this study, the effect of calcium supply on Src degradation was investigated to confirm underlying mechanisms and anticancer effects targeting TNBC. MDA-MB-231 cells, the TNBC cell line, were used. Calcium supply was feasible through lactate calcium salt (CaLac), and the applicable calcium concentration was decided by changes in the viability with different doses of CaLac. Expression of signaling molecules mediated by calcium-dependent Src degradation was observed by Western blot analysis and immunocytochemistry, and the recovery of the signaling molecules was confirmed following calpeptin treatment. The anticancer effect was investigated in the xenograft animal model. Significant suppression of Src was induced by calcium supply, followed by a successive decrease in the expression of epithelial growth factor receptor, RAS, extracellular signal-regulated kinase, and nuclear factor kappa B. Then, the suppression of cyclooxygenase-2 contributed to a significant deactivation of the prostaglandin E2 receptors. These results suggest that calcium supply has the potential to reduce the risk of TNBC. However, as this study is at an early stage to determine clinical applicability, close consideration is needed.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea; (S.Y.P.); (M.H.P.)
| | - Seon Young Park
- Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea; (S.Y.P.); (M.H.P.)
| | - Min Hee Park
- Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea; (S.Y.P.); (M.H.P.)
| | - Hwan Mook Kim
- MetiMedi Pharmaceuticals Co., 40, Imi-ro, Uiwang-si 16006, Republic of Korea
| |
Collapse
|
23
|
Li X, Chen Y, Wang T, Liu Z, Yin G, Wang Z, Sui C, Zhu L, Chen W. GPR81-mediated reprogramming of glucose metabolism contributes to the immune landscape in breast cancer. Discov Oncol 2023; 14:140. [PMID: 37500811 PMCID: PMC10374510 DOI: 10.1007/s12672-023-00709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/31/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Local tumor microenvironment (TME) plays a crucial role in immunotherapy for breast cancer (BC). Whereas, the molecular mechanism responsible for the crosstalk between BC cells and surrounding immune cells remains unclear. The present study aimed to determine the interplay between GPR81-mediated glucometabolic reprogramming of BC and the immune landscape in TME. MATERIALS AND METHODS Immunohistochemistry (IHC) assay was first performed to evaluate the association between GPR81 and the immune landscape. Then, several stable BC cell lines with down-regulated GPR81 expression were established to directly identify the role of GPR81 in glucometabolic reprogramming, and western blotting assay was used to detect the underlying molecular mechanism. Finally, a transwell co-culture system confirmed the crosstalk between glucometabolic regulation mediated by GPR81 in BC and induced immune attenuation. RESULTS IHC analysis demonstrated that the representation of infiltrating CD8+ T cells and FOXP3+ T cells were dramatically higher in BC with a triple negative (TN) subtype in comparison with that with a non-TN subtype (P < 0.001). Additionally, the ratio of infiltrating CD8+ to FOXP3+ T cells was significantly negatively associated with GPR81 expression in BC with a TN subtype (P < 0.001). Furthermore, GPR81 was found to be substantially correlated with the glycolytic capability (P < 0.001) of BC cells depending on a Hippo-YAP signaling pathway (P < 0.001). In the transwell co-culture system, GPR81-mediated reprogramming of glucose metabolism in BC significantly contributed to a decreased proportion of CD8+ T (P < 0.001) and an increased percentage of FOXP3+ T (P < 0.001) in the co-cultured lymphocytes. CONCLUSION Glucometabolic reprogramming through a GPR81-mediated Hippo-YAP signaling pathway was responsible for the distinct immune landscape in BC. GPR81 was a potential biomarker to stratify patients before immunotherapy to improve BC's clinical prospect.
Collapse
Affiliation(s)
- Xiaofeng Li
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yiwen Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zifan Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guotao Yin
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Chunxiao Sui
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lei Zhu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wei Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Molecular Imaging and Nuclear Medicine,Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
24
|
Foroughi A, Arefpour AM, Nikoofar A, Sanei M, Mahdavi SH, Javadinia SA. Total Neoadjuvant vs. Standard Perioperative Cisplatin/ Doxorubicin Chemotherapy in Patients with Extremities Osteosarcoma: A Multi-Center Cohort Study. Asian Pac J Cancer Prev 2023; 24:2369-2374. [PMID: 37505768 PMCID: PMC10676487 DOI: 10.31557/apjcp.2023.24.7.2369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
INTRODUCTION Despite improvements in survival of patients with high-grade osteosarcoma after the implementation of perioperative chemotherapy, osteosarcoma remains among the most lethal cancers. Prescription of all chemotherapy courses before the surgery may provide this opportunity to eliminate micrometastases more efficiently, increase the chances of pathologic complete response and organ preserving surgery. This study aimed to compare the outcomes of total neoadjuvant chemotherapy vs. standard perioperative chemotherapy with cisplatin/doxorubicin regimen in patients with extremities osteosarcoma. METHODS In this retrospective cohort, all patients with high-grade osteosarcoma admitted to oncologic centers affiliated to Iran University of Medical Sciences in Tehran, Iran from 2015 to 2021 were included. Organ preserving rates, pathologic responses, and survival of patients who received all six courses of cisplatin/doxorubicin regimen preoperatively were compared to those who received the regimen perioperatively. RESULTS Sixty-three patients were enrolled (total neoadjuvant chemotherapy: 32 patients and perioperative chemotherapy: 31 patients). In total neoadjuvant chemotherapy and perioperative chemotherapy groups, favorable pathology responses (necrosis>90%) were reported in 80.6% and 15.6% of patients, respectively (p<0.001). With a median follow-up of 24 months, mean overall survival of total neoadjuvant chemotherapy and perioperative chemotherapy groups were 21.29 months (95% CI; 19.3-23.27) and 23.46 months (95% CI; 22.7-24.1), respectively (p=0.2). The mean disease-free survival of patients in total neoadjuvant chemotherapy and perioperative chemotherapy groups were 19.54 months (95% CI; 17.0-22.0) and 21.37 months (95% CI; 19.4-23.2), respectively (p=0.2). CONCLUSION Our results showed that prescription of all courses of doxorubicin/cisplatin chemotherapy prior to surgery can increase favorable pathologic response rates, although this improvement is not translated into overall and disease-free survival benefits.
Collapse
Affiliation(s)
- Ahmad Foroughi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Amir Mohammad Arefpour
- Department of Radiotherapy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Alireza Nikoofar
- Department of Radiotherapy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mastaneh Sanei
- Department of Radiotherapy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Seyedeh Hoda Mahdavi
- Department of Radiotherapy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Seyed Alireza Javadinia
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| |
Collapse
|
25
|
Wilkinson AN. Demystifying breast cancer. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2023; 69:473-476. [PMID: 37451990 PMCID: PMC10348793 DOI: 10.46747/cfp.6907473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Affiliation(s)
- Anna N Wilkinson
- Associate Professor in the Department of Family Medicine at the University of Ottawa in Ontario, a family physician with the Ottawa Academic Family Health Team, a general practitioner oncologist at the Ottawa Hospital Cancer Centre, Program Director of PGY-3 FP-Oncology, and Regional Cancer Primary Care Lead for Champlain Region
| |
Collapse
|
26
|
Wilkinson AN. Démystifier le cancer du sein. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2023; 69:e149-e153. [PMID: 37451991 PMCID: PMC10348801 DOI: 10.46747/cfp.6907e149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Affiliation(s)
- Anna N Wilkinson
- Professeure agrégée au Département de médecine familiale de l'Université d'Ottawa (Ontario), médecin de famille dans l'Équipe universitaire de santé familiale d'Ottawa, omnipraticienne en oncologie au Centre de cancérologie de l'Hôpital d'Ottawa, directrice du programme de compétences avancées R3 en oncologie en MF et responsable régionale des soins primaires en cancérologie pour la région de Champlain
| |
Collapse
|
27
|
Markhulia J, Kekutia S, Mikelashvili V, Saneblidze L, Tsertsvadze T, Maisuradze N, Leladze N, Czigány Z, Almásy L. Synthesis, Characterization, and In Vitro Cytotoxicity Evaluation of Doxorubicin-Loaded Magnetite Nanoparticles on Triple-Negative Breast Cancer Cell Lines. Pharmaceutics 2023; 15:1758. [PMID: 37376206 DOI: 10.3390/pharmaceutics15061758] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
In this study, we investigated the cytotoxicity of doxorubicin (DOX)-loaded magnetic nanofluids on 4T1 mouse tumor epithelial cells and MDA-MB-468 human triple-negative breast cancer (TNBC) cells. Superparamagnetic iron oxide nanoparticles were synthesized using sonochemical coprecipitation by applying electrohydraulic discharge treatment (EHD) in an automated chemical reactor, modified with citric acid and loaded with DOX. The resulting magnetic nanofluids exhibited strong magnetic properties and maintained sedimentation stability in physiological pH conditions. The obtained samples were characterized using X-ray diffraction (XRD), transmission electron microscopy (TEM), Fourier-transform infrared spectroscopy, UV-spectrophotometry, dynamic light scattering (DLS), electrophoretic light scattering (ELS), vibrating sample magnetometry (VSM), and transmission electron microscopy (TEM). In vitro studies using the MTT method revealed a synergistic effect of the DOX-loaded citric-acid-modified magnetic nanoparticles on the inhibition of cancer cell growth and proliferation compared to treatment with pure DOX. The combination of the drug and magnetic nanosystem showed promising potential for targeted drug delivery, with the possibility of optimizing the dosage to reduce side-effects and enhance the cytotoxic effect on cancer cells. The nanoparticles' cytotoxic effects were attributed to the generation of reactive oxygen species and the enhancement of DOX-induced apoptosis. The findings suggest a novel approach for enhancing the therapeutic efficacy of anticancer drugs and reducing their associated side-effects. Overall, the results demonstrate the potential of DOX-loaded citric-acid-modified magnetic nanoparticles as a promising strategy in tumor therapy, and provide insights into their synergistic effects.
Collapse
Affiliation(s)
- Jano Markhulia
- Nanocomposites Laboratory, Vladimer Chavchanidze Institute of Cybernetics of the Georgian Technical University, Z. Anjafaridze Str. 5, 0186 Tbilisi, Georgia
| | - Shalva Kekutia
- Nanocomposites Laboratory, Vladimer Chavchanidze Institute of Cybernetics of the Georgian Technical University, Z. Anjafaridze Str. 5, 0186 Tbilisi, Georgia
| | - Vladimer Mikelashvili
- Nanocomposites Laboratory, Vladimer Chavchanidze Institute of Cybernetics of the Georgian Technical University, Z. Anjafaridze Str. 5, 0186 Tbilisi, Georgia
| | - Liana Saneblidze
- Nanocomposites Laboratory, Vladimer Chavchanidze Institute of Cybernetics of the Georgian Technical University, Z. Anjafaridze Str. 5, 0186 Tbilisi, Georgia
| | - Tamar Tsertsvadze
- Department of Biology Chair of Immunology and Microbiology, Faculty of Exact and Natural Sciences, Ivane Javakhishvili Tbilisi State University, 1, Ilia Tchavchavadze Ave., 0179 Tbilisi, Georgia
| | - Nino Maisuradze
- Nanocomposites Laboratory, Vladimer Chavchanidze Institute of Cybernetics of the Georgian Technical University, Z. Anjafaridze Str. 5, 0186 Tbilisi, Georgia
- Department of Biology Chair of Immunology and Microbiology, Faculty of Exact and Natural Sciences, Ivane Javakhishvili Tbilisi State University, 1, Ilia Tchavchavadze Ave., 0179 Tbilisi, Georgia
| | - Nino Leladze
- Department of Biology Chair of Immunology and Microbiology, Faculty of Exact and Natural Sciences, Ivane Javakhishvili Tbilisi State University, 1, Ilia Tchavchavadze Ave., 0179 Tbilisi, Georgia
| | - Zsolt Czigány
- Institute for Technical Physics and Materials Science, Centre for Energy Research, Konkoly Thege Miklós Str. 29-33, 1121 Budapest, Hungary
| | - László Almásy
- Institute for Energy Security and Environmental Safety, Centre for Energy Research, Konkoly Thege Miklós Str. 29-33, 1121 Budapest, Hungary
| |
Collapse
|
28
|
Wu L, Lin Y, Gao S, Wang Y, Pan H, Wang Z, Pozzolini M, Yang F, Zhang H, Yang Y, Xiao L, Xu Y. Luteolin inhibits triple-negative breast cancer by inducing apoptosis and autophagy through SGK1-FOXO3a-BNIP3 signaling. Front Pharmacol 2023; 14:1200843. [PMID: 37346292 PMCID: PMC10279868 DOI: 10.3389/fphar.2023.1200843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023] Open
Abstract
Background: Triple-negative breast cancer (TNBC) is one of the most prominent neoplasm disorders and lacks efficacious treatments yet. Luteolin (3',4',5,7-tetrahydroxyflavone), a natural flavonoid commonly presented in plants, has been reported to delay the progression of TNBC. However, the precise mechanism is still elusive. We aimed to elucidate the inhibition and molecular regulation mechanism of luteolin on TNBC. Methods: The effects of luteolin on the biological functions of TNBC cells were first evaluated using the corresponding assays for cell counting kit-8 assay, flow cytometry, wound-healing assay, and transwell migration assay, respectively. The mechanism of luteolin on TNBC cells was then analyzed by RNA sequencing and verified by RT-qPCR, Western blot, transmission electron microscopy, etc. Finally, in vivo mouse tumor models were constructed to further confirm the effects of luteolin on TNBC. Results: Luteolin dramatically suppressed cell proliferation, invasion, and migration while favoring cell apoptosis in a dose- and time-dependent manner. In TNBC cells treated with luteolin, SGK1 and AKT3 were significantly downregulated while their downstream gene BNIP3 was upregulated. According to the results of 3D modeling, the direct binding of luteolin to SGK1 was superior to that of AKT3. The inhibition of SGK1 promoted FOXO3a translocation into the nucleus and led to the transcription of BNIP3 both in vitro and in vivo, eventually facilitating the interaction between BNIP3 and apoptosis and autophagy protein. Furthermore, the upregulation of SGK1, induced by luteolin, attenuated the apoptosis and autophagy of the TNBC. Conclusion: Luteolin inhibits TNBC by inducing apoptosis and autophagy through SGK1-FOXO3a-BNIP3 signaling.
Collapse
Affiliation(s)
- Ling Wu
- Medical College of Yangzhou University, Yangzhou, China
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yingda Lin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Songyu Gao
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yongfang Wang
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Huiji Pan
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, China
| | - Zhaozhi Wang
- School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Marina Pozzolini
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Genova, Italy
| | - Fengling Yang
- Department of Healthcare, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Haiyan Zhang
- Department of Healthcare, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yi Yang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Liang Xiao
- Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yuan Xu
- Department of Pharmacy, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| |
Collapse
|
29
|
Bhattarai S, Saini G, Li H, Duanmu H, Seth G, Fisher TB, Janssen EAM, Kiraz U, Kong J, Aneja R. Predicting neoadjuvant treatment response in triple-negative breast cancer using machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.536459. [PMID: 37131688 PMCID: PMC10153161 DOI: 10.1101/2023.04.17.536459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background Neoadjuvant chemotherapy (NAC) is the standard treatment for early-stage triple negative breast cancer (TNBC). The primary endpoint of NAC is a pathological complete response (pCR). NAC results in pCR in only 30%â€"40% of TNBC patients. Tumor-infiltrating lymphocytes (TILs), Ki67 and phosphohistone H3 (pH3) are a few known biomarkers to predict NAC response. Currently, systematic evaluation of the combined value of these biomarkers in predicting NAC response is lacking. In this study, the predictive value of markers derived from H&E and IHC stained biopsy tissue was comprehensively evaluated using a supervised machine learning (ML)-based approach. Identifying predictive biomarkers could help guide therapeutic decisions by enabling precise stratification of TNBC patients into responders and partial or non-responders. Methods Serial sections from core needle biopsies (n=76) were stained with H&E, and immunohistochemically for the Ki67 and pH3 markers, followed by whole slide image (WSI) generation. The resulting WSI triplets were co-registered with H&E WSIs serving as the reference. Separate mask region-based CNN (MRCNN) models were trained with annotated H&E, Ki67 and pH3 images for detecting tumor cells, stromal and intratumoral TILs (sTILs and tTILs), Ki67 + , and pH3 + cells. Top image patches with a high density of cells of interest were identified as hotspots. Best classifiers for NAC response prediction were identified by training multiple ML models, and evaluating their performance by accuracy, area under curve, and confusion matrix analyses. Results Highest prediction accuracy was achieved when hotspot regions were identified by tTIL counts and each hotspot was represented by measures of tTILs, sTILs, tumor cells, Ki67 + , and pH3 + features. Regardless of the hotspot selection metric, a complementary use of multiple histological features (tTILs, sTILs) and molecular biomarkers (Ki67 and pH3) resulted in top ranked performance at the patient level. Conclusions Overall, our results emphasize that prediction models for NAC response should be based on biomarkers in combination rather than in isolation. Our study provides compelling evidence to support the use of ML-based models to predict NAC response in patients with TNBC.
Collapse
|
30
|
Assunção Ribeiro da Costa RE, Rocha de Oliveira FT, Nascimento Araújo AL, Vieira SC. Impact of Pathologic Complete Response on the Prognosis of Triple-Negative Breast Cancer Patients: A Cohort Study. Cureus 2023; 15:e37396. [PMID: 37182056 PMCID: PMC10171840 DOI: 10.7759/cureus.37396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is a molecular subtype in which estrogen (ER)/progesterone receptor (PR) and human epidermal growth receptor 2 (HER2) expression does not occur. The objective of this study was to analyze the impact of pathologic complete response (pCR) after neoadjuvant chemotherapy on the prognosis of triple-negative breast cancer (TNBC) patients. Methods This cohort study was conducted in a private-sector oncology clinic located in the city of Teresina, Brazil. Medical charts of 532 breast cancer patients treated from 2007 to 2020 were analyzed. Of these patients, 83 women with TNBC were selected (10 patients were excluded from the study). Univariate and multivariate analyses (Cox regression) were performed to evaluate the impact on patient survival, comparing patients with or without pCR. A significance level of 5% was set. Overall survival (OS) and disease-free survival (DFS) curves were constructed according to the Kaplan-Meier model. Results Angiolymphatic invasion and positive sentinel lymph node were associated with a lower OS and/or DFS in TNBC (p<0.05). The 10-year OS was 78% and 49%, and the 10-year DFS was 97% and 32% in patients with or without pCR, respectively. Conclusion pCR after neoadjuvant chemotherapy was associated with improvement in OS and DFS in TNBC patients.
Collapse
|
31
|
Sivina E, Blumberga L, Purkalne G, Irmejs A. Pathological complete response to neoadjuvant chemotherapy in triple negative breast cancer - single hospital experience. Hered Cancer Clin Pract 2023; 21:4. [PMID: 36922883 PMCID: PMC10018905 DOI: 10.1186/s13053-023-00249-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer is a heterogeneous molecular subtype of BC. Pathological complete response (pCR) is an important surrogate marker for recurrence-free and overall survival. AIM OF STUDY The aim of this study was to evaluate clinical and pathological factors that are associated with complete pathological response status in triple-negative breast cancer patients receiving neoadjuvant chemotherapy. MATERIALS AND METHODS Eighty triple-negative breast cancer patients who underwent neoadjuvant chemotherapy followed by surgery at Pauls Stradins Clinical University Hospital between January 2018 and January 2020 were retrospectively analysed. Twenty-six patients (32.5%) were BRCA1/2 pathogenic variant carriers. RESULTS A total of 32.5% (n = 26) of patients in all study groups and 57.7% (n = 15) of patients with BRCA1/2 pathogenic variants achieved pCR. Forty-seven patients received platinum-based neoadjuvant chemotherapy, and 19 patients (40.4%) achieved complete pathological response. Patients in the pCR group presented with significantly higher Ki-67 scores (p = 0.007), BRCA1/2 pathogenic variants (p = 0.001) and younger age (p = 0.02) than those in the non-pCR group. pCR did not significantly impact recurrence-free survival (RFS) or overall survival (OS). Multivariate analysis revealed that pretreatment N stage (clinical nodal status) was an independent prognostic factor for RFS and OS. CONCLUSIONS BRCA1 pathogenic variants, high Ki67 score and young age were predictors of pathological complete response, while clinical nodal status predicted survival outcomes in triple-negative breast cancer.
Collapse
Affiliation(s)
- Elina Sivina
- Institute of Oncology, Tumour Clinical Research Department, Riga Stradins University, Riga, Latvia. .,Clinic of Oncology, Pauls Stradins Clinical University Hospital, Riga, Latvia. .,Department of Internal Diseases, Riga Stradins University, Riga, Latvia.
| | - Lubova Blumberga
- Clinic of Oncology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Gunta Purkalne
- Clinic of Oncology, Pauls Stradins Clinical University Hospital, Riga, Latvia.,Department of Internal Diseases, Riga Stradins University, Riga, Latvia
| | - Arvids Irmejs
- Institute of Oncology, Tumour Clinical Research Department, Riga Stradins University, Riga, Latvia.,Breast Unit, Department of Surgery, Pauls Stradins Clinical University Hospital, Riga, Latvia.,Department of Surgery, Riga Stradins University, Riga, Latvia
| |
Collapse
|
32
|
van den Ende NS, Nguyen AH, Jager A, Kok M, Debets R, van Deurzen CHM. Triple-Negative Breast Cancer and Predictive Markers of Response to Neoadjuvant Chemotherapy: A Systematic Review. Int J Mol Sci 2023; 24:ijms24032969. [PMID: 36769287 PMCID: PMC9918290 DOI: 10.3390/ijms24032969] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Around 40-50% of all triple-negative breast cancer (TNBC) patients achieve a pathological complete response (pCR) after treatment with neoadjuvant chemotherapy (NAC). The identification of biomarkers predicting the response to NAC could be helpful for personalized treatment. This systematic review provides an overview of putative biomarkers at baseline that are predictive for a pCR following NAC. Embase, Medline and Web of Science were searched for articles published between January 2010 and August 2022. The articles had to meet the following criteria: patients with primary invasive TNBC without distant metastases and patients must have received NAC. In total, 2045 articles were screened by two reviewers resulting in the inclusion of 92 articles. Overall, the most frequently reported biomarkers associated with a pCR were a high expression of Ki-67, an expression of PD-L1 and the abundance of tumor-infiltrating lymphocytes, particularly CD8+ T cells, and corresponding immune gene signatures. In addition, our review reveals proteomic, genomic and transcriptomic markers that relate to cancer cells, the tumor microenvironment and the peripheral blood, which also affect chemo-sensitivity. We conclude that a prediction model based on a combination of tumor and immune markers is likely to better stratify TNBC patients with respect to NAC response.
Collapse
Affiliation(s)
- Nadine S. van den Ende
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
- Correspondence: ; Tel.: +31-640213383
| | - Anh H. Nguyen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Marleen Kok
- Department of Medical Oncology, Tumor Biology & Immunology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| | - Carolien H. M. van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Centre, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
33
|
Prediction of pathologic complete response to neoadjuvant systemic therapy in triple negative breast cancer using deep learning on multiparametric MRI. Sci Rep 2023; 13:1171. [PMID: 36670144 PMCID: PMC9859781 DOI: 10.1038/s41598-023-27518-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer. Neoadjuvant systemic therapy (NAST) followed by surgery are currently standard of care for TNBC with 50-60% of patients achieving pathologic complete response (pCR). We investigated ability of deep learning (DL) on dynamic contrast enhanced (DCE) MRI and diffusion weighted imaging acquired early during NAST to predict TNBC patients' pCR status in the breast. During the development phase using the images of 130 TNBC patients, the DL model achieved areas under the receiver operating characteristic curves (AUCs) of 0.97 ± 0.04 and 0.82 ± 0.10 for the training and the validation, respectively. The model achieved an AUC of 0.86 ± 0.03 when evaluated in the independent testing group of 32 patients. In an additional prospective blinded testing group of 48 patients, the model achieved an AUC of 0.83 ± 0.02. These results demonstrated that DL based on multiparametric MRI can potentially differentiate TNBC patients with pCR or non-pCR in the breast early during NAST.
Collapse
|
34
|
Prognostic Implications of the Residual Tumor Microenvironment after Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer Patients without Pathological Complete Response. Cancers (Basel) 2023; 15:cancers15030597. [PMID: 36765559 PMCID: PMC9913578 DOI: 10.3390/cancers15030597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
With a high risk of relapse and death, and a poor or absent response to therapeutics, the triple-negative breast cancer (TNBC) subtype is particularly challenging, especially in patients who cannot achieve a pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). Although the tumor microenvironment (TME) is known to influence disease progression and the effectiveness of therapeutics, its predictive and prognostic potential remains uncertain. This work aimed to define the residual TME profile after NAC of a retrospective cohort with 96 TNBC patients by immunohistochemical staining (cell markers) and chromogenic in situ hybridization (genetic markers). Kaplan-Meier curves were used to estimate the influence of the selected TME markers on five-year overall survival (OS) and relapse-free survival (RFS) probabilities. The risks of each variable being associated with relapse and death were determined through univariate and multivariate Cox analyses. We describe a unique tumor-infiltrating immune profile with high levels of lymphocytes (CD4, FOXP3) and dendritic cells (CD21, CD1a and CD83) that are valuable prognostic factors in post-NAC TNBC patients. Our study also demonstrates the value of considering not only cellular but also genetic TME markers such as MUC-1 and CXCL13 in routine clinical diagnosis to refine prognosis modelling.
Collapse
|
35
|
Identification and Validation of a Novel Glycolysis-Related Gene Signature for Predicting the Prognosis and Therapeutic Response in Triple-Negative Breast Cancer. Adv Ther 2023; 40:310-330. [PMID: 36316558 DOI: 10.1007/s12325-022-02330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/21/2022] [Indexed: 01/21/2023]
Abstract
INTRODUCTION A high malignancy rate and poor prognosis are common problems with triple-negative breast cancer (TNBC). There is increasing evidence that glycolysis plays vital roles in tumorigenesis, tumor invasion, immune evasion, chemoresistance, and metastasis. However, a comprehensive analysis of the diagnostic and prognostic significance of glycolysis in TNBC is lacking. METHODS Transcriptomic and clinical data of TNBC patients were obtained from The Cancer Genome Atlas (TCGA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) databases, respectively. Glycolysis-related genes (GRGs) were collected from the Molecular Signatures Database (MSigDB). Differential comparative analysis was performed to obtain the differentially expressed (DE)-GRGs associated with TNBC. Based on the DE-GRGs, a glycolysis-related risk signature was established using Least Absolute Shrinkage and Selector Operation (LASSO) and multivariable Cox regression analyses. The prognostic value, tumor microenvironment, mutation status, and chemotherapy response of different risk groups were analyzed. An independent cohort from the METABRIC database was used for external validation. Furthermore, the expression patterns of five genes derived from the prognostic model were validated by quantitative real-time polymerase chain reaction (RT-qPCR). RESULTS The glycolysis-related prognostic signature included five genes (IFNG, ACSS2, IRS2, GFUS, and GAL3ST1) and predicted the prognosis of TNBC patients independent of clinical factors (p < 0.05). Patients were divided into high- and low-risk groups based on the median risk score. Compared to low-risk TNBC patients, high-risk patients had significantly decreased overall survival (HR = 2.718, p < 0.001). Receiver operating characteristic and calibration curves demonstrated that the model had high performance in terms of predicting survival and risk stratification. The results remained consistent after external verification. Additionally, the tumor immune microenvironment significantly differed between the risk groups. Low-risk TNBC patients had a better immunotherapy response than high-risk patients. High-risk TNBC patients with a poor prognosis may benefit from targeted therapy. CONCLUSIONS This study developed a novel glycolysis and prognosis-related (GRP) signature based on GRGs to predict the prognosis of TNBC patients, and may aid clinical decision-making for these patients.
Collapse
|
36
|
Preoperative Radio(Chemo)Therapy in Breast Cancer: Time to Switch the Perspective? Curr Oncol 2022; 29:9767-9787. [PMID: 36547182 PMCID: PMC9777182 DOI: 10.3390/curroncol29120768] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
AIM Radiation therapy represents, together with surgery and systemic treatment, the triad on which the current management of patients with breast cancer is based, achieving high control and survival rates. In recent years we have witnessed a (r)evolution in the conception of breast cancer treatment. The classic scheme of surgery followed by systemic treatment and radiotherapy is being subverted and it is becoming more and more frequent to propose the primary administration of systemic treatment before surgery, seeking to maximize its effect and favoring not only the performance of more conservative surgeries but also, in selected cases, increasing the rates of disease-free survival and overall survival. Radiotherapy is also evolving toward a change in perspective: considering preoperative primary administration of radiotherapy may be useful in selected groups. Advances in radiobiological knowledge, together with technological improvements that are constantly being incorporated into clinical practice, support the administration of increasingly reliable, precise, and effective radiotherapy, as well as its safe combination with antitumor drugs or immunotherapy in the primary preoperative context. In this paper, we present a narrative review of the usefulness of preoperative radiotherapy for breast cancer patients and the possibilities for its combination with other therapies.
Collapse
|
37
|
Liu Z, Gao J, Gu R, Shi Y, Hu H, Liu J, Huang J, Zhong C, Zhou W, Yang Y, Gong C. Comprehensive Analysis of Transcriptomics and Genetic Alterations Identifies Potential Mechanisms Underlying Anthracycline Therapy Resistance in Breast Cancer. Biomolecules 2022; 12:biom12121834. [PMID: 36551262 PMCID: PMC9775906 DOI: 10.3390/biom12121834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Anthracycline is a mainstay of treatment for breast cancer patients because of its antitumor activity. However, anthracycline resistance is a critical barrier in treating breast cancer. Thus, it is of great importance to uncover the molecular mechanisms underlying anthracycline resistance in breast cancer. Herein, we integrated transcriptome data, genetic alterations data, and clinical data of The Cancer Genome Atlas (TCGA) to identify the molecular mechanisms involved in anthracycline resistance in breast cancer. Two hundred and four upregulated genes and 1376 downregulated genes were characterized between the anthracycline-sensitive and anthracycline-resistant groups. It was found that drug resistance-associated genes such as ABCB5, CYP1A1, and CYP4Z1 were significantly upregulated in the anthracycline-resistant group. The gene set enrichment analysis (GSEA) suggested that the P53 signaling pathway, DNA replication, cysteine, and methionine metabolism pathways were associated with anthracycline sensitivity. Somatic TP53 mutation was a common genetic abnormality observed in the anthracycline-sensitive group, while CDH1 mutation was presented in the anthracycline-resistant group. Immune infiltration patterns were extremely different between the anthracycline-sensitive and anthracycline-resistant groups. Immune-associated chemokines and cytokines, immune regulators, and human leukocyte antigen genes were significantly upregulated in the anthracycline-sensitive group. These results reveal potential molecular mechanisms associated with anthracycline resistance.
Collapse
Affiliation(s)
- Zihao Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Jingbo Gao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ran Gu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yu Shi
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Hong Hu
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Jianlan Liu
- Department of Pathology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Jiefeng Huang
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Caineng Zhong
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Wenbin Zhou
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Yaping Yang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Correspondence: (Y.Y.); or (C.G.)
| | - Chang Gong
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Correspondence: (Y.Y.); or (C.G.)
| |
Collapse
|
38
|
Lin YY, Gao HF, Yang X, Zhu T, Zheng XX, Ji F, Zhang LL, Yang CQ, Yang M, Li JQ, Cheng MY, Wang K. Neoadjuvant therapy in triple-negative breast cancer: A systematic review and network meta-analysis. Breast 2022; 66:126-135. [PMID: 36265208 PMCID: PMC9587342 DOI: 10.1016/j.breast.2022.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Evidence for the preferred neoadjuvant therapy regimen in triple-negative breast cancer (TNBC) is not yet established. METHODS Literature search was conducted from inception to February 12, 2022. Phase 2 and 3 randomized controlled trials (RCTs) investigating neoadjuvant therapy for TNBC were eligible. The primary outcome was pathologic complete response (pCR); the secondary outcomes were all-cause treatment discontinuation, disease-free survival or event-free survival (DFS/EFS), and overall survival. Odd ratios (OR) with 95% credible intervals (CrI) were used to estimate binary outcomes; hazard ratios (HR) with 95% CrI were used to estimate time-to-event outcomes. Bayesian network meta-analysis was implemented for each endpoint. Sensitivity analysis and network meta-regression were done. RESULTS 41 RCTs (N = 7109 TNBC patients) were eligible. Compared with anthracycline- and taxane-based chemotherapy (ChT), PD-1 inhibitor plus platinum plus anthracycline- and taxane-based ChT was associated with a significant increased pCR rate (OR 3.95; 95% CrI 1.81-9.44) and a higher risk of premature treatment discontinuation (3.25; 1.26-8.29). Compared with dose-dense anthracycline- and taxane-based ChT, the combined treatment was not associated with significantly improved pCR (OR 2.57; 95% CrI 0.69-9.92). In terms of time-to-event outcomes, PD-1 inhibitor plus platinum plus anthracycline- and taxane-based ChT was associated with significantly improved DFS/EFS (HR 0.42; 95% CrI 0.19-0.81). CONCLUSIONS PD-1 inhibitor plus platinum and anthracycline- and taxane-based ChT was currently the most efficacious regimen for pCR and DFS/EFS improvement in TNBC. The choice of chemotherapy backbone, optimization of patient selection with close follow-up and proactive symptomatic managements are essential to the antitumor activity of PD-1 inhibitor.
Collapse
Affiliation(s)
- Ying-Yi Lin
- Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Hong-Fei Gao
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Xin Yang
- Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Anesthesiology, the First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, 515041, Guangdong, China
| | - Teng Zhu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Xing-Xing Zheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China; Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Fei Ji
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Liu-Lu Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Ci-Qiu Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Mei Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Jie-Qing Li
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Min-Yi Cheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Kun Wang
- Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| |
Collapse
|
39
|
Azevedo-Barbosa H, Ferreira-Silva GÁ, do Vale BP, Hawkes JA, Ionta M, Carvalho DT. Synthesis and Structure-Activity Relationship Studies of Novel Aryl Sulfonamides and Their Activity against Human Breast Cancer Cell Lines. Chem Biodivers 2022; 19:e202200831. [PMID: 36305872 DOI: 10.1002/cbdv.202200831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/27/2022] [Indexed: 12/27/2022]
Abstract
A series of structural analogs of aryl sulfonamide hybrid compounds were synthesised and their cytotoxic activity was evaluated against three human breast cancer cell lines (MCF-7, MDA-MB-231 and Hs 578T). The compounds were designed through electronic, hydrophobic and steric modifications using the chemical structure of N-{4-[(2-hydroxy-3-methoxy-5-propylphenyl)sulfamoyl]phenyl}acetamide (referred to as compound 7) as a starting point to then assess a structure-activity relationship (SAR) study. From the data generated, we observed that compounds 9, 10 and 11 (which have modifications in the substituents of the aryl sulfonamide), efficiently reduced the cell viability of MCF-7 and MDA-MB-231 cell cultures. Based on initial data, we selected compounds 10 and 11 for further investigations into their antiproliferative and/or cytotoxic profile against MDA-MB-231 cells, and we noted that compound 10 was the most promising compound in the series. Compound 10 promoted morphological changes and altered the dynamics of cell cycle progression in MDA-MB-231 cells, inducing arrest in G1/S transition. Taken together, these results show that the dihydroeugenol-aryl-sulfonamide hybrid compound 10 (which has an electron withdrawing nitro group) displays promising antiproliferative activity against MDA-MB-231 cell lines.
Collapse
Affiliation(s)
- Helloana Azevedo-Barbosa
- LQFar - Laboratory of Pharmaceutical Chemistry Research, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, 700, Gabriel Monteiro da Silva, 37130-001, Alfenas, MG, Brazil
| | - Guilherme Álvaro Ferreira-Silva
- LABAInt - Laboratory of Integrative Animal Biology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Bianca Pereira do Vale
- LQFar - Laboratory of Pharmaceutical Chemistry Research, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, 700, Gabriel Monteiro da Silva, 37130-001, Alfenas, MG, Brazil
| | - Jamie Anthony Hawkes
- LQFar - Laboratory of Pharmaceutical Chemistry Research, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, 700, Gabriel Monteiro da Silva, 37130-001, Alfenas, MG, Brazil
| | - Marisa Ionta
- LABAInt - Laboratory of Integrative Animal Biology, Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Diogo Teixeira Carvalho
- LQFar - Laboratory of Pharmaceutical Chemistry Research, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, 700, Gabriel Monteiro da Silva, 37130-001, Alfenas, MG, Brazil
| |
Collapse
|
40
|
Wu C, Hormuth DA, Lorenzo G, Jarrett AM, Pineda F, Howard FM, Karczmar GS, Yankeelov TE. Towards Patient-Specific Optimization of Neoadjuvant Treatment Protocols for Breast Cancer Based on Image-Guided Fluid Dynamics. IEEE Trans Biomed Eng 2022; 69:3334-3344. [PMID: 35439121 PMCID: PMC9640301 DOI: 10.1109/tbme.2022.3168402] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE This study establishes a fluid dynamics model personalized with patient-specific imaging data to optimize neoadjuvant therapy (i.e., doxorubicin) protocols for breast cancers. METHODS Ten patients recruited at the University of Chicago were included in this study. Quantitative dynamic contrast-enhanced and diffusion weighted magnetic resonance imaging data are leveraged to estimate patient-specific hemodynamic properties, which are then used to constrain the mechanism-based drug delivery model. Then, computer simulations of this model yield the subsequent drug distribution throughout the breast. By systematically varying the dosing schedule, we identify an optimized regimen for each patient using the maximum safe therapeutic duration (MSTD), which is a metric balancing treatment efficacy and toxicity. RESULTS With an individually optimized dose (range = 12.11-15.11 mg/m2 per injection), a 3-week regimen consisting of a uniform daily injection significantly outperforms all other scheduling strategies (P < 0.001). In particular, the optimal protocol is predicted to significantly outperform the standard protocol (P < 0.001), improving the MSTD by an average factor of 9.93 (range = 6.63 to 14.17). CONCLUSION A clinical-mathematical framework was developed by integrating quantitative MRI data, advanced image processing, and computational fluid dynamics to predict the efficacy and toxicity of neoadjuvant therapy protocols, thus enabling the rational identification of an optimal therapeutic regimen on a patient-specific basis. SIGNIFICANCE Our clinical-computational approach has the potential to enable optimization of therapeutic regimens on a patient-specific basis and provide guidance for prospective clinical trials aimed at refining neoadjuvant therapy protocols for breast cancers.
Collapse
Affiliation(s)
- Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, the University of Texas at Austin, Austin TX 78712 USA
| | - David A. Hormuth
- Oden Institute for Computational Engineering and Sciences, and Livestrong Cancer Institutes, The University of Texas at Austin, USA
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, the University of Texas at Austin; Department of Civil Engineering and Architecture, University of Pavia, Italy
| | - Angela M. Jarrett
- Oden Institute for Computational Engineering and Sciences, and Livestrong Cancer Institutes, The University of Texas at Austin, USA
| | | | - Frederick M. Howard
- Section of Hematology/Oncology - Department of Medicine, The University of Chicago, USA
| | | | - Thomas E. Yankeelov
- Department of Biomedical Engineering, Department of Diagnostic Medicine, Department of Oncology, Oden Institute for Computational Engineering and Sciences, and Livestrong Cancer Institutes, The University of Texas at Austin; Department of Imaging Physics, MD Anderson Cancer Center, USA
| |
Collapse
|
41
|
Ciérvide R, Montero Á, García-Rico E, García-Aranda M, Herrero M, Skaarup J, Benassi L, Barrera MJ, Vega E, Rojas B, Bratos R, Luna A, Parras M, López M, Delgado A, Quevedo P, Castilla S, Feyjoo M, Higueras A, Prieto M, Suarez-Gauthier A, Garcia-Cañamaque L, Escolán N, Álvarez B, Chen X, Alonso R, López M, Hernando O, Valero J, Sánchez E, Ciruelos E, Rubio C. Primary Chemoradiotherapy Treatment (PCRT) for HER2+ and Triple Negative Breast Cancer Patients: A Feasible Combination. Cancers (Basel) 2022; 14:cancers14184531. [PMID: 36139688 PMCID: PMC9496977 DOI: 10.3390/cancers14184531] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 12/02/2022] Open
Abstract
Primary systemic treatment (PST) downsizes the tumor and improves pathological response. The aim of this study is to analyze the feasibility and tolerance of primary concurrent radio−chemotherapy (PCRT) in breast cancer patients. Patients with localized TN/HER2+ tumors were enrolled in this prospective study. Radiation was delivered concomitantly during the first 3 weeks of chemotherapy, and it was based on a 15 fractions scheme, 40.5 Gy/2.7 Gy per fraction to whole breast and nodal levels I-IV. Chemotherapy (CT) was based on Pertuzumab−Trastuzumab−Paclitaxel followed by anthracyclines in HER2+ and CBDCA-Paclitaxel followed by anthracyclines in TN breast cancers patients. A total of 58 patients were enrolled; 25 patients (43%) were TN and 33 patients HER2+ (57%). With a median follow-up of 24.2 months, 56 patients completed PCRT and surgery. A total of 35 patients (87.5%) achieved >90% loss of invasive carcinoma cells in the surgical specimen. The 70.8% and the 53.1% of patients with TN and HER-2+ subtype, respectively, achieved complete pathological response (pCR). This is the first study of concurrent neoadjuvant treatment in breast cancer in which three strategies were applied simultaneously: fractionation of RT (radiotherapy) in 15 sessions, adjustment of CT to tumor phenotype and local planning by PET. The pCR rates are encouraging.
Collapse
Affiliation(s)
- Raquel Ciérvide
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
- Correspondence: (R.C.); (E.G.-R.); Tel.: +34-669554042 (R.C.); +34-609165218 (E.G.-R.)
| | - Ángel Montero
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Eduardo García-Rico
- Department of Medical Oncology, HM Hospitales, 28050 Madrid, Spain
- Correspondence: (R.C.); (E.G.-R.); Tel.: +34-669554042 (R.C.); +34-609165218 (E.G.-R.)
| | | | - Mercedes Herrero
- Department of Gynecology and Obstetrics, HM Hospitales, 28050 Madrid, Spain
| | - Jessica Skaarup
- Department of Gynecology and Obstetrics, HM Hospitales, 28050 Madrid, Spain
| | - Leticia Benassi
- Department of Gynecology and Obstetrics, HM Hospitales, 28050 Madrid, Spain
| | - Maria José Barrera
- Department of Gynecology and Obstetrics, HM Hospitales, 28050 Madrid, Spain
| | - Estela Vega
- Department of Medical Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Beatriz Rojas
- Department of Medical Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Raquel Bratos
- Department of Medical Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Ana Luna
- Department of Medical Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Manuela Parras
- Department of Radiology, HM Hospitales, 28050 Madrid, Spain
| | - María López
- Department of Radiology, HM Hospitales, 28050 Madrid, Spain
| | - Ana Delgado
- Department of Radiology, HM Hospitales, 28050 Madrid, Spain
| | - Paloma Quevedo
- Department of Radiology, HM Hospitales, 28050 Madrid, Spain
| | | | - Margarita Feyjoo
- Department of Medical Oncology, Hospital Sanitas La Moraleja, 28050 Madrid, Spain
| | - Ana Higueras
- Department of Gynecology and Obstetrics, Hospital Sanitas La Moraleja, 28050 Madrid, Spain
| | - Mario Prieto
- Department of Pathology, HM Hospitales, 28050 Madrid, Spain
| | | | | | - Nieves Escolán
- Department of Plastic Surgery, HM Hospitales, 28050 Madrid, Spain
| | - Beatriz Álvarez
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Xin Chen
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Rosa Alonso
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Mercedes López
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Ovidio Hernando
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Jeannette Valero
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Emilio Sánchez
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Eva Ciruelos
- Department of Medical Oncology, HM Hospitales, 28050 Madrid, Spain
| | - Carmen Rubio
- Department of Radiation Oncology, HM Hospitales, 28050 Madrid, Spain
| |
Collapse
|
42
|
Zhang J, Xia Y, Zhou X, Yu H, Tan Y, Du Y, Zhang Q, Wu Y. Current landscape of personalized clinical treatments for triple-negative breast cancer. Front Pharmacol 2022; 13:977660. [PMID: 36188535 PMCID: PMC9523914 DOI: 10.3389/fphar.2022.977660] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly malignant subtype of breast cancer (BC) with vicious behaviors. TNBC is usually associated with relatively poor clinical outcomes, earlier recurrence, and high propensity for visceral metastases than other BC types. TNBC has been increasingly recognized to constitute a very molecular heterogeneous subtype, which may offer additional therapeutic opportunities due to newly discovered cancer-causing drivers and targets. At present, there are multiple novel targeted therapeutic drugs in preclinical researches, clinical trial designs, and clinical practices, such as platinum drugs, poly ADP-ribose polymerase (PARP) inhibitors, immunocheckpoint inhibitors, androgen receptor inhibitors as well as PI3K/AKT/mTOR targeted inhibitors. These personalized, single, or combinational therapies based on molecular heterogeneity are currently showing positive results. The scope of this review is to highlight the latest knowledge about these potential TNBC therapeutic drugs, which will provide comprehensive insights into the personalized therapeutic strategies and options for combating TNBC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Yu Xia
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomei Zhou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Honghao Yu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Li Y, Zhang H, Merkher Y, Chen L, Liu N, Leonov S, Chen Y. Recent advances in therapeutic strategies for triple-negative breast cancer. J Hematol Oncol 2022; 15:121. [PMID: 36038913 PMCID: PMC9422136 DOI: 10.1186/s13045-022-01341-0] [Citation(s) in RCA: 217] [Impact Index Per Article: 108.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 01/03/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer (BC) with a poor prognosis. Current treatment options are limited to surgery, adjuvant chemotherapy and radiotherapy; however, a proportion of patients have missed the surgical window at the time of diagnosis. TNBC is a highly heterogeneous cancer with specific mutations and aberrant activation of signaling pathways. Hence, targeted therapies, such as those targeting DNA repair pathways, androgen receptor signaling pathways, and kinases, represent promising treatment options against TNBC. In addition, immunotherapy has also been demonstrated to improve overall survival and response in TNBC. In this review, we summarize recent key advances in therapeutic strategies based on molecular subtypes in TNBC.
Collapse
Affiliation(s)
- Yun Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huajun Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700
| | - Lin Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700. .,Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia, 142290.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
44
|
An Integrative Clinical Model for the Prediction of Pathological Complete Response in Patients with Operable Stage II and Stage III Triple-Negative Breast Cancer Receiving Neoadjuvant Chemotherapy. Cancers (Basel) 2022; 14:cancers14174170. [PMID: 36077706 PMCID: PMC9454735 DOI: 10.3390/cancers14174170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Neoadjuvant chemotherapy (NAC) is widely used to treat stage II and III primary, operable triple-negative breast cancer (TNBC). The response to NAC critically affects the subsequent treatment plan, including not only curative surgical planning but also adjuvant therapy. There is no standard prediction model that accurately predicts NAC response. Therefore, the development of an easy-to-apply and cost-effective clinical prediction model for NAC treatment response would improve clinical practice. We propose an integrative clinical prediction model for the prediction of pathologically complete response in patients with operable stage II and stage III TNBC receiving NAC based on findings from tumor ultrasound and blood tests. All included parameters were readily available during and before NAC. This clinical prediction model could provide a reference to guide clinicians’ decisions in planning a patient’s NAC treatment as early as after the first cycle of NAC. Abstract Triple-negative breast cancer (TNBC) is treated with neoadjuvant chemotherapy (NAC). The response to NAC, particularly the probability of a complete pathological response (pCR), guides the surgical approach and adjuvant therapy. We developed a prediction model using a nomogram integrating blood tests and pre-treatment ultrasound findings for predicting pCR in patients with stage II or III operable TNBC receiving NAC. Clinical data before and after the first cycle of NAC collected from patients between 2012 and 2019 were analyzed using univariate and multivariate analyses to identify correlations with pCR. The coefficients of the significant parameters were calculated using logistic regression, and a nomogram was developed based on the logistic model to predict the probability of pCR. Eighty-eight patients were included. Five parameters correlated with the probability of pCR, including the neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte (PLR) ratio, percentage change in PLR, presence of echogenic halo, and tumor height-to-width ratio. The discrimination performance of the nomogram was indicated by an area under the curve of 87.7%, and internal validation showed that the chi-square value of the Hosmer–Lemeshow test was 7.67 (p = 0.363). Thus, the integrative prediction model using clinical data can predict the probability of pCR in patients with TNBC receiving NAC.
Collapse
|
45
|
Xu H, Wang Y, Han Y, Wu Y, Wang J, Xu B. CDK4/6 inhibitors versus PI3K/AKT/mTOR inhibitors in women with hormone receptor-positive, HER2-negative metastatic breast cancer: An updated systematic review and network meta-analysis of 28 randomized controlled trials. Front Oncol 2022; 12:956464. [PMID: 36091147 PMCID: PMC9449843 DOI: 10.3389/fonc.2022.956464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Updated evidence was required to compare the efficacy and safety of cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) inhibitors for patients with hormone receptor-positive and HER2-negative metastatic breast cancer. Methods A systematic review and network meta-analysis was conducted utilizing data from randomized controlled trials (RCTs) that contained interventions of CDK4/6 inhibitors or PI3K/AKT/mTOR inhibitors. Progression-free survival (PFS), overall survival (OS), and treatment-related adverse events (TRAEs) were primary outcomes of interest. Pooled hazard ratios (HRs) and odds ratios (ORs) with 95% credible intervals (CrIs) were used to assess the survival outcomes and safety profiles, respectively. Results A total of 28 RCTs with 12,129 participants were included. Pooled analysis showed that CDK4/6 inhibitors significantly prolonged PFS than PI3K/AKT/mTOR inhibitors (HR, 0.81; 95% CrI, 0.69–0.94), whereas no significant differences were detected regarding OS. After balancing the treatment lines and metastatic sites, the superiority of CDK4/6 inhibitors only appeared in the visceral and non-visceral subgroups. Among CDK4/6 inhibitors, abemaciclib was significantly better than others in ≥3 grade neutropenia (OR, 0.04; 95% CrI, 0.01–0.15). The incidence of stomatitis and digestive disorders was different among diverse kinds of PI3K/AKT/mTOR inhibitors. Discrepancies appeared regarding TRAEs of hepatotoxicity, diarrhea, and hyperglycemia among different interventions. Conclusions CDK4/6 inhibitors showed better efficacy in PFS, but the benefits disappeared when taking treatment line into consideration. Specific and discrepant safety profiles were found in two categories of agents. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42022321172.
Collapse
Affiliation(s)
| | | | | | | | - Jiayu Wang
- *Correspondence: Binghe Xu, ; Jiayu Wang,
| | - Binghe Xu
- *Correspondence: Binghe Xu, ; Jiayu Wang,
| |
Collapse
|
46
|
Wang H, Zhao C, Santa-Maria CA, Emens LA, Popel AS. Dynamics of tumor-associated macrophages in a quantitative systems pharmacology model of immunotherapy in triple-negative breast cancer. iScience 2022; 25:104702. [PMID: 35856032 PMCID: PMC9287616 DOI: 10.1016/j.isci.2022.104702] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/05/2022] [Accepted: 06/27/2022] [Indexed: 11/07/2022] Open
Abstract
Quantitative systems pharmacology (QSP) modeling is an emerging mechanistic computational approach that couples drug pharmacokinetics/pharmacodynamics and the course of disease progression. It has begun to play important roles in drug development for complex diseases such as cancer, including triple-negative breast cancer (TNBC). The combination of the anti-PD-L1 antibody atezolizumab and nab-paclitaxel has shown clinical activity in advanced TNBC with PD-L1-positive tumor-infiltrating immune cells. As tumor-associated macrophages (TAMs) serve as major contributors to the immuno-suppressive tumor microenvironment, we incorporated the dynamics of TAMs into our previously published QSP model to investigate their impact on cancer treatment. We show that through proper calibration, the model captures the macrophage heterogeneity in the tumor microenvironment while maintaining its predictive power of the trial results at the population level. Despite its high mechanistic complexity, the modularized QSP platform can be readily reproduced, expanded for new species of interest, and applied in clinical trial simulation. A mechanistic model of quantitative systems pharmacology in immuno-oncology Dynamics of tumor-associated macrophages are integrated into our previous work Conducting in silico clinical trials to predict clinical response to cancer therapy
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu211166, China
| | - Cesar A Santa-Maria
- Department of Oncology, the Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205, USA
| | - Leisha A Emens
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Oncology, the Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21205, USA
| |
Collapse
|
47
|
Xiao Y, Gao W. Therapeutic pattern and progress of neoadjuvant treatment for triple-negative breast cancer. Oncol Lett 2022; 24:219. [PMID: 35720488 PMCID: PMC9178680 DOI: 10.3892/ol.2022.13340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/03/2022] [Indexed: 11/23/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease, accounting for about 15.0-20.0% of all breast cancer cases. TNBC is associated with early recurrence and metastasis, strong invasiveness and a poor prognosis. Chemotherapy is currently the mainstay of treatment for TNBC, and achievement of a pathological complete response is closely associated with a long-term good prognosis. Improving the long-term prognosis in patients with TNBC is a challenge in breast cancer treatment, and more clinical evidence is needed to guide the choice of treatment strategies. The current study reviews the conventional treatment modality for TNBC and the selection of neoadjuvant chemotherapy (NACT) regimens available. The research progress on optimizing NACT regimens is also reviewed, and the uniqueness of the treatment of this breast cancer subtype is emphasized, in order to provide reference for the clinical practice and research with regard to TNBC treatment.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Oncology, Dongguan Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Wencheng Gao
- Department of General Surgery, Dongguan Houjie Town People's Hospital, Dongguan, Guangdong 523962, P.R. China
| |
Collapse
|
48
|
Silva D, Mesquita A. Evolving Evidence for the Optimization of Neoadjuvant Therapy in Triple-Negative Breast Cancer. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234221107580. [PMID: 35783596 PMCID: PMC9243491 DOI: 10.1177/11782234221107580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Representing 15% to 20% of all invasive breast cancers, adjuvant systemic
treatment for early-stage, high-risk triple-negative breast cancer (TNBC) is
preferentially done in the neoadjuvant setting based on a chemotherapy backbone
of anthracyclines and taxanes. Pathological complete response to neoadjuvant
treatment constitutes the main objective, regarding its correlation with
oncological outcomes. The optimal neoadjuvant regimen to achieve the highest
rates of pathological complete response is still under investigation, with the
increasing knowledge on the molecular pathways, genomic sequencing, and
immunological profile of TNBC allowing for the development of a wide array of
new therapeutic options. This review aims to summarize the current evidence and
ongoing clinical trials of new therapeutic options for the neoadjuvant treatment
of TNBC patients.
Collapse
Affiliation(s)
- Diogo Silva
- Department of Medical Oncology, Matosinhos Local Health Unity – Hospital Pedro Hispano, Porto, Portugal
| | - Alexandra Mesquita
- Department of Medical Oncology, Matosinhos Local Health Unity – Hospital Pedro Hispano, Porto, Portugal
| |
Collapse
|
49
|
Costa B, Vale N. Drug Metabolism for the Identification of Clinical Biomarkers in Breast Cancer. Int J Mol Sci 2022; 23:3181. [PMID: 35328602 PMCID: PMC8951384 DOI: 10.3390/ijms23063181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is classified into four major molecular subtypes, and is considered a heterogenous disease. The risk profiles and treatment of breast cancer differ according to these subtypes. Early detection dramatically improves the prospects of successful treatment, resulting in a reduction in overall mortality rates. However, almost 30% of women primarily diagnosed with the early-stage disease will eventually develop metastasis or resistance to chemotherapies. Immunotherapies are among the most promising cancer treatment options; however, long-term clinical benefit has only been observed in a small subset of responding patients. The current strategies for diagnosis and treatment rely heavily on histopathological examination and molecular diagnosis, disregarding the tumor microenvironment and microbiome involving cancer cells. In this review, we aim to praise the use of pharmacogenomics and pharmacomicrobiomics as a strategy to identify potential biomarkers for guiding and monitoring therapy in real-time. The finding of these biomarkers can be performed by studying the metabolism of drugs, more specifically, immunometabolism, and its relationship with the microbiome, without neglecting the information provided by genetics. A larger understanding of cancer biology has the potential to improve patient care, enable clinical decisions, and deliver personalized medicine.
Collapse
Affiliation(s)
- Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Associate Laboratory RISE-Health Research Network, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
50
|
Li J, Chen L, Tan W, Qi F, Zhang Y, Wang Z, Shao Z. Platinum is essential in neoadjuvant treatment of triple-negative breast cancer: a network meta-analysis. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0529. [PMID: 35170879 PMCID: PMC9196055 DOI: 10.20892/j.issn.2095-3941.2021.0529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE This study aimed to assess the efficacy and safety of various neoadjuvant regimens for patients diagnosed with early-stage or locally advanced triple-negative breast cancer (TNBC). METHODS Medline, EMBASE, Cochrane Library, and Web of Science were searched in May 2020 to identify randomized controlled trials (RCTs). Bayesian network meta-analysis (NMA) was performed (Registration: PROSPERO CRD42020223012). RESULTS A total of 35 RCTs involving 8,424 participants were reviewed, of which 22 RCTs with 5,203 patients were included in this NMA focusing on pathologic complete response (pCR). An anthracycline-taxane-based (AT) regimen combined with a platinum (ATPt) [odds ratio (OR) = 2.04, 95% credible interval (CrI): 1.69, 2.48] regimen, and a docetaxel regimen combined with a carboplatin (TCb; OR = 2.16, 95% CrI: 1.20, 3.91) regimen improved pCR beyond that with AT only. AT and ATPt combined with targeted therapy [including bevacizumab (Bev), veliparib, atezolizumab, or pembrolizumab] also improved pCR. Five RCTs included in this NMA reported serious adverse events (SAEs) or grade ≥ 3 AEs. TCb was associated with fewer grade ≥ 3 AEs than was AT (OR = 0.66, 95% CrI: 0.23, 1.72) alone. In contrast, ATPt, AT + Bev, ATPt + Bev, ATPt + veliparib, and ATPt + pembrolizumab were associated with more SAEs than was AT alone. CONCLUSIONS In patients with TNBC, platinum-based neoadjuvant regimens ATPt and TCb increase pCR beyond that with AT alone, but TCb appears to be better tolerated than either AT or ATPt. Platinum-based regimens combined with targeted therapies (Bev, PARPi, and PD-1/PD-L1 inhibitor) also improve the pCR rate beyond that with AT alone, but this benefit is accompanied by greater toxicity.
Collapse
Affiliation(s)
- Junjie Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Li Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wei Tan
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham NG7 2RD, UK
| | - Fang Qi
- Academic Department, Systematic Review Solutions Ltd, Shanghai 201400, China
| | - Yang Zhang
- Academic Department, Systematic Review Solutions Ltd, Shanghai 201400, China
| | - Zhonghua Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhimin Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|