1
|
Srivastava TP, Dhar R, Karmakar S. Looking beyond the ER, PR, and HER2: what's new in the ARsenal for combating breast cancer? Reprod Biol Endocrinol 2025; 23:9. [PMID: 39833837 PMCID: PMC11744844 DOI: 10.1186/s12958-024-01338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BrCa) is a complex and heterogeneous disease with diverse molecular subtypes, leading to varied clinical outcomes and posing significant treatment challenges. The increasing global burden of BrCa, particularly in low- and middle-income countries, underscores the urgent need for more effective therapeutic strategies. The androgen receptor (AR), expressed in a substantial proportion of breast cancer cases, has emerged as a potential biomarker and therapeutic target. In breast cancer, AR exhibits diverse functions across subtypes, often interacting with other hormone receptors, thereby influencing tumor progression and treatment responses. This intricate interplay is further complicated by the presence of constitutively expressed AR splice variants (AR-Vs) that drive resistance to AR-targeting therapies through structural rearrangements in the domains and activation of aberrant signaling pathways. Although AR-targeting drugs, initially developed for prostate cancer (PCa), have shown promise in AR-positive breast cancer, significant gaps remain in understanding AR's precise functions and therapeutic potential. The systemic management of breast cancer is guided primarily by theranostic biomarkers; ER, PR, HER2, and Ki67 which also dictate the breast cancer classification. The ubiquitous expression of AR in BrCa and the emergence of AR-Vs can assist the management of disease complementing the standard of care. This article provides a comprehensive overview of AR and its splice variants in the context of breast cancer, highlighting their prognostic and predictive value across different subtypes looking beyond the conventional ER, PR, and HER2 status. This review also raises the possibility of using AR splice variants in predicting tumor aggressiveness. From the settings of developing nations, this may provide useful insight by integrating recent advances in AR-targeted therapies and exploring their translational potential, emphasizing the critical need for further research to optimize AR-based therapeutic strategies for breast cancer management.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Female
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
2
|
Liu X, Ma Z, Jing X, Wang G, Zhao L, Zhao X, Zhang Y. The deubiquitinase OTUD5 stabilizes SLC7A11 to promote progression and reduce paclitaxel sensitivity in triple-negative breast cancer. Cancer Lett 2024; 604:217232. [PMID: 39276913 DOI: 10.1016/j.canlet.2024.217232] [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: 06/03/2024] [Revised: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
Ferroptosis is a newly defined form of programmed cell death characterized by iron-dependent lipid peroxide accumulation and is associated with the progression of cancer. Solute carrier family 7 member 11 (SLC7A11), a key component of cystine/glutamate antiporter, has been characterized as a critical regulator of ferroptosis. Although many studies have established the transcriptional regulation of SLC7A11, it remains largely unknown how the stability of SLC7A11 is regulated in cancers, especially in triple-negative breast cancer (TNBC). Here we demonstrated that ovarian tumor domain-containing protein 5 (OTUD5), which deubiquitinated and stabilized SLC7A11, played a key role in TNBC progression and paclitaxel chemosensitivity through modulating ferroptosis. The clinical data analysis showed OTUD5 was higher expressed in TNBC, which positively correlated with SLC7A11 level. Mechanistically, OTUD5 interacted with SLC7A11 and cleaved K48-linked polyubiquitin chains from SLC7A11 to enhance the stability of SLC7A11. Taken together, these findings uncover a functional and mechanistic role of OTUD5 in TNBC progression and paclitaxel sensitivity, indicating OTUD5 could be a potential target for TNBC treatment.
Collapse
Affiliation(s)
- Xizhi Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Xi'an, China
| | - Xin Jing
- Department of Pathology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Guanying Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lin Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinhan Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yujiao Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
3
|
Mishra A, Mishra SK, Sharanappa V, Krishnani N, Kumari N, Agarwal G. Incidence and Prognostic Significance of Androgen Receptors (AR) in Indian Triple-Negative Breast Cancer (TNBC). Indian J Surg Oncol 2024; 15:250-257. [PMID: 38741650 PMCID: PMC11088609 DOI: 10.1007/s13193-024-01877-2] [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: 02/13/2023] [Accepted: 01/07/2024] [Indexed: 05/16/2024] Open
Abstract
Molecular sub-characterization of triple-negative breast cancer (TNBC) has great therapeutic and possibly prognostic implications. The primary aim of this study was to investigate the incidence of luminal androgen receptor (LAR) subtype of TNBC and secondary aims were sub-categorization and clinico-pathologic correlation of LAR breast cancers. Retrospective study (January 2008 and 31st of December 2018) consisting of 157 TNBC patients. Androgen receptor (AR) expression was measured by immunohistochemical analysis. One percent cutoff was set as a positive expression. Sub-categorization was done on the basis of EGFR (> 15% of tumor cells) and Ki-67 expression (low- < 11%, intermediate- 11-20%, and high- > 21%). AR expression was correlated with various clinico-pathologic features and outcomes of the patients. The incidence of AR expression in TNBC was 24.8%. Considering different thresholds of > 5%, > 10%, and > 20% immunostaining, the incidence of AR positivity was 18.4, 15.2, and 11.5% respectively. The incidence of Ki-67 (p = 0.89) and EGFR (p = 0.643) expression did not differ significantly in AR-positive and -negative TNBC. Based on EGFR expression 19, 67 and 14% patients were categorized as low, intermediate, and high risk respectively. Low-risk (p ≤ 0.001) and low-grade (p = 0.014) tumors were more likely to have > 10% AR expression. Clinico-pathological profile, response to neoadjuvant chemotherapy, disease-free survival (p = 0.458), and overall survival (p = 0.806) did not significantly differ between AR expressing and negative TNBC. On multivariate analysis, only tumor staging was a significant predictor of survival (p = 0.012) and AR expression of > 10% revealed a trend towards improved survival (p = 0.07). When considering only AR-positive TNBC, AR expression of > 10% (p = 0.038), distant metastases (p = 0.003), and EGFR status (p = 0.024) were significantly associated with survival. AR expression does not seem to very strongly correlate with prognosis in TNBC and further studies could focus more on its predictive role in deciding anti-androgen therapy.
Collapse
Affiliation(s)
- Anjali Mishra
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Shravan Kumar Mishra
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Vikram Sharanappa
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Niraj Kumari
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Gaurav Agarwal
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| |
Collapse
|
4
|
Alrumaihi F. Chemoinformatics and machine learning techniques to identify novel inhibitors of the lemur tyrosine kinase-3 receptor involved in breast cancer. Front Mol Biosci 2024; 11:1366763. [PMID: 38638686 PMCID: PMC11025642 DOI: 10.3389/fmolb.2024.1366763] [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: 01/07/2024] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Breast cancer is still the largest cause of cancer death in women, and around 70% of primary breast cancer patients are estrogen receptor (ER)-positive, which is the most frequent kind of breast cancer. The lemur tyrosine kinase-3 (LMTK3) receptor has been linked to estrogen responsiveness in breast cancer. However, the function of LMTK3 in reaction to cytotoxic chemotherapy has yet to be studied. Breast cancer therapy research remains tricky due to a paucity of structural investigations on LMTK3. We performed structural investigations on LMTK3 using molecular docking and molecular dynamics (MD) simulations of the LMTK3 receptor in complex with the top three inhibitor molecules along with a control inhibitor. Analysis revealed the top three compounds show the best binding affinities during docking simulations. Interactive analysis of hydrogen bonds inferred hotspot residues Tyr163, Asn138, Asp133, Tyr56, Glu52, Ser132, Asp313, and Asp151. Some other residues in the 5-Å region determined strong alkyl bonds and conventional hydrogen bond linkages. Furthermore, protein dynamics analysis revealed significant modifications among the top complexes and the control system. There was a transition from a loop to a-helix conformation in the protein-top1 complex, and in contrast, in complexes top2 and top3, the formation of a stabilizing sheet in the C chain was observed, which limited significant mobility and increased complex stability. Significant structural alterations were observed in the protein-top complexes, including a shorter helix region and the creation of some loop regions in comparison to the control system. Interestingly, binding free energies, including MMGB/PBSA WaterSwap analysis estimation, reveals that the top1 complex system was more stable than other systems, especially in comparison to the control inhibitor complex system. These results suggest a the plausible mode of action for the novel inhibitors. Therefore, the current investigation contributes to understanding the mechanism of action, serving as a basis for future experimental studies.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
5
|
Xavier PL, Marção M, Simões RL, Job MEG, de Francisco Strefezzi R, Fukumasu H, Malta TM. Machine learning determines stemness associated with simple and basal-like canine mammary carcinomas. Heliyon 2024; 10:e26714. [PMID: 38439848 PMCID: PMC10909659 DOI: 10.1016/j.heliyon.2024.e26714] [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: 01/19/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Simple and complex carcinomas are the most common type of malignant Canine Mammary Tumors (CMTs), with simple carcinomas exhibiting aggressive behavior and poorer prognostic. Stemness is an ability associated with cancer initiation, malignancy, and therapeutic resistance, but is still few elucidated in canine mammary tumor subtypes. Here, we first validated, using CMT samples, a previously published canine one-class logistic regression machine learning algorithm (OCLR) to predict stemness (mRNAsi) in canine cancer cells. Then, using the canine mRNAsi, we observed that simple carcinomas exhibit higher stemness than complex carcinomas and other histological subtypes. Also, we confirmed that stemness is higher and associated with basal-like CMTs and with NMF2 metagene signature, a tumor-specific DNA-repair metagene signature. Using correlation analysis, we selected the top 50 genes correlated with higher stemness, and the top 50 genes correlated with lower stemness and further performed a gene set enrichment analysis to observe the biological processes enriched for these genes. Finally, we suggested two promise stemness-associated targets in CMTs, POLA2 and APEX1, especially in simple carcinomas. Thus, our work elucidates stemness as a potential mechanism behind the aggressiveness and development of canine mammary tumors, especially in simple carcinomas, describing evidence of a promising strategy to target this disease.
Collapse
Affiliation(s)
- Pedro L.P. Xavier
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
| | - Maycon Marção
- Cancer Epigenomics Laboratory, Department of Clinical Analysis, Toxicology and Food Sciences, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renan L.S. Simões
- Cancer Epigenomics Laboratory, Department of Clinical Analysis, Toxicology and Food Sciences, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Eduarda G. Job
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
| | - Ricardo de Francisco Strefezzi
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
| | - Heidge Fukumasu
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, São Paulo, Brazil
| | - Tathiane M. Malta
- Cancer Epigenomics Laboratory, Department of Clinical Analysis, Toxicology and Food Sciences, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
6
|
Basho RK, Zhao L, White JB, Huo L, Bassett RL, Mittendorf EA, Thompson A, Litton JK, Ueno N, Arun B, Lim B, Valero V, Tripathy D, Zhang J, Adrada BE, Santiago L, Ravenberg E, Seth S, Yam C, Moulder SL, Damodaran S. Comprehensive Analysis Identifies Variability in PI3K Pathway Alterations in Triple-Negative Breast Cancer Subtypes. JCO Precis Oncol 2024; 8:e2300124. [PMID: 38484209 PMCID: PMC10954064 DOI: 10.1200/po.23.00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 03/19/2024] Open
Abstract
PURPOSE The PI3K pathway is frequently altered in triple-negative breast cancer (TNBC). Limited cell line and human data suggest that TNBC tumors characterized as mesenchymal (M) and luminal androgen receptor (LAR) subtypes have increased incidence of alterations in the PI3K pathway. The impact of PI3K pathway alterations across TNBC subtypes is poorly understood. METHODS Pretreatment tumor was evaluated from operable TNBC patients enrolled on a clinical trial of neoadjuvant therapy (NAT; A Robust TNBC Evaluation fraMework to Improve Survival [ClinicalTrials.gov identifier: NCT02276443]). Tumors were characterized into seven TNBC subtypes per Pietenpol criteria (basal-like 1, basal-like 2, immunomodulatory, M, mesenchymal stem-like, LAR, and unstable). Using whole-exome sequencing, RNA sequencing, and immunohistochemistry for PTEN, alterations were identified in 32 genes known to activate the PI3K pathway. Alterations in each subtype were associated with pathologic response to NAT. RESULTS In evaluated patients (N = 177), there was a significant difference in the incidence of PI3K pathway alterations across TNBC subtypes (P < .01). The highest incidence of alterations was seen in LAR (81%), BL2 (79%), and M (62%) subtypes. The odds ratio for pathologic complete response (pCR) in the presence of PIK3CA mutation, PTEN mutation, and/or PTEN loss was highest in the LAR subtype and lowest in the M subtype, but these findings did not reach statistical significance. Presence of PIK3CA mutation was associated with pCR in the LAR subtype (P = .02). CONCLUSION PI3K pathway alteration can affect response to NAT in TNBC, and targeted agents may improve outcomes, particularly in patients with M and LAR TNBC.
Collapse
Affiliation(s)
| | - Li Zhao
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason B. White
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lei Huo
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | - Naoto Ueno
- University of Texas MD Anderson Cancer Center, Houston, TX
- University of Hawaii Cancer Center, Honolulu, HI
| | - Banu Arun
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bora Lim
- Baylor College of Medicine, Houston, TX
| | - Vicente Valero
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Debu Tripathy
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jianhua Zhang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Sahil Seth
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Clinton Yam
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stacy L. Moulder
- University of Texas MD Anderson Cancer Center, Houston, TX
- Eli Lilly and Company, Indianapolis, IN
| | | |
Collapse
|
7
|
Cao T, Huang M, Huang X, Tang T. Research and experimental verification on the mechanisms of cellular senescence in triple-negative breast cancer. PeerJ 2024; 12:e16935. [PMID: 38435998 PMCID: PMC10909353 DOI: 10.7717/peerj.16935] [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: 11/17/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with high heterogeneity, poor prognosis, and a low 10-year survival rate of less than 50%. Although cellular senescence displays extensive effects on cancer, the comprehensions of cellular senescence-related characteristics in TNBC patients remains obscure. Method Single-cell RNA sequencing (scRNA-seq) data were analyzed by Seurat package. Scores for cellular senescence-related pathways were computed by single-sample gene set enrichment analysis (ssGSEA). Subsequently, unsupervised consensus clustering was performed for molecular cluster identification. Immune scores of patients in The Cancer Genome Atlas (TCGA) dataset and associated immune cell scores were calculated using Estimation of STromal and Immune cells in MAlignantTumours using Expression data (ESTIMATE) and Microenvironment Cell Populations-counter (MCP-counter), Tumor Immune Estimation Resource (TIMER) and Estimating the Proportion of Immune and Cancer cells (EPIC) methods, respectively. Immunotherapy scores were assessed using TIDE. Furthermore, feature genes were identified by univariate Cox and Least Absolute Shrinkage and Selection Operator (LASSO) regression analyses; these were used to construct a risk model. Additionally, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and transwell assay were conducted for in vitro validation of hub genes. Result TNBC was classified into three subtypes based on cellular senescence-related pathways as clusters 1, 2, and 3. Specifically, cluster 1 showed the best prognosis, followed by cluster 2 and cluster 3. The levels of gene expression in cluster 2 were the lowest, whereas these were the highest in cluster 3. Moreover, clusters 1 and 3 showed a high degree of immune infiltration. TIDE scores were higher for cluster 3, suggesting that immune escape was more likely in patients with the cluster 3 subtype who were less likely to benefit from immunotherapy. Next, the TNBC risk model was constructed and validated. RT-qPCR revealed that prognostic risk genes (MMP28, ACP5 and KRT6A) were up-regulated while protective genes (CT83) were down-regulated in TNBC cell lines, validating the results of the bioinformatics analysis. Meanwhile, cellular experiments revealed that ACP5 could promote the migration and invasion abilities in two TNBC cell lines. Finally, we evaluated the validity of prognostic models for assessing TME characteristics and TNBC chemotherapy response. Conclusion In conclusion, these findings help to assess the efficacy of targeted therapies in patients with different molecular subtypes, have practical applications for subtype-specific treatment of TNBC patients, and provide information on prognostic factors, as well as guidance for the revelation of the molecular mechanisms by which senescence-associated genes influence TNBC progression.
Collapse
Affiliation(s)
- Tengfei Cao
- Department of Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengjie Huang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinyue Huang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Tang
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Hoogenboezem EN, Patel SS, Lo JH, Cavnar AB, Babb LM, Francini N, Gbur EF, Patil P, Colazo JM, Michell DL, Sanchez VM, McCune JT, Ma J, DeJulius CR, Lee LH, Rosch JC, Allen RM, Stokes LD, Hill JL, Vickers KC, Cook RS, Duvall CL. Structural optimization of siRNA conjugates for albumin binding achieves effective MCL1-directed cancer therapy. Nat Commun 2024; 15:1581. [PMID: 38383524 PMCID: PMC10881965 DOI: 10.1038/s41467-024-45609-0] [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: 03/14/2023] [Accepted: 01/29/2024] [Indexed: 02/23/2024] Open
Abstract
The high potential of siRNAs to silence oncogenic drivers remains largely untapped due to the challenges of tumor cell delivery. Here, divalent lipid-conjugated siRNAs are optimized for in situ binding to albumin to improve pharmacokinetics and tumor delivery. Systematic variation of the siRNA conjugate structure reveals that the location of the linker branching site dictates tendency toward albumin association versus self-assembly, while the lipid hydrophobicity and reversibility of albumin binding also contribute to siRNA intracellular delivery. The lead structure increases tumor siRNA accumulation 12-fold in orthotopic triple negative breast cancer (TNBC) tumors over the parent siRNA. This structure achieves approximately 80% silencing of the anti-apoptotic oncogene MCL1 and yields better survival outcomes in three TNBC models than an MCL-1 small molecule inhibitor. These studies provide new structure-function insights on siRNA-lipid conjugate structures that are intravenously injected, associate in situ with serum albumin, and improve pharmacokinetics and tumor treatment efficacy.
Collapse
Affiliation(s)
- Ella N Hoogenboezem
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shrusti S Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Justin H Lo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley B Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren M Babb
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Eva F Gbur
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danielle L Michell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Violeta M Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua T McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Linus H Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonah C Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ryan M Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Larry D Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jordan L Hill
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Rebecca S Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
9
|
Chang YJ, Prince GMSH, Wei PL, Batzorig U, Huang CY, Hung CS, Chang TC. The role of thrombomodulin in modulating ITGB3 expression and its implications for triple-negative breast cancer progression. Cell Biol Int 2024; 48:216-228. [PMID: 38081783 DOI: 10.1002/cbin.12104] [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: 09/05/2023] [Revised: 10/24/2023] [Accepted: 11/18/2023] [Indexed: 01/18/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer (BC) compared to other BC subtypes in clinical settings. Currently, there are no effective therapeutic strategies for TNBC treatment. Therefore, there is an urgent need to identify suitable biomarkers or therapeutic targets for TNBC patients. Thrombomodulin (TM) plays a role in cancer progression and metastasis in many different cancers. However, the role of TM in TNBC is not yet fully understood. First, silenced-TM in MDA-MB-231 cells caused an increase in proliferative and metastatic activity. In contrast, overexpression of TM in Hs578T cells caused a reduction in proliferation, invasion, and migration rate. Using RNA-seq analysis, we found that Integrin beta 3 (ITGB3) expression may be a downstream target of TM. Furthermore, we found an increase in ITGB3 levels in TM-KD cells by QPCR and western blot analysis but a decrease in ITGB3 levels in TM-overexpressing cells. We found phospho-smad2/3 levels were increased in TM-KD cells but decreased in TM-overexpressing cells. This implies that TM negatively regulates ITGB3 levels through the activation of the smad2/3 pathway. Silencing ITGB3 in TM-KD cells caused a decrease in proliferation and migration. Finally, we found that higher ITGB3 levels were correlated with poor overall survival and relapse-free survival in patients with TNBC. Our results indicated a novel regulatory relationship between TM and ITGB3 in TNBC.
Collapse
Affiliation(s)
- Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Cancer Research Center and Translational Laboratory, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | | | - Po-Li Wei
- Department of Medical Research, Cancer Research Center and Translational Laboratory, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, Division of Colorectal Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, College of Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Uyanga Batzorig
- Department of Dermatology, University of California, San Diego, La Jolla, California, USA
| | - Chien-Yu Huang
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chin-Sheng Hung
- Department of Surgery, Division of Colorectal Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tung-Cheng Chang
- Department of Surgery, Division of Colorectal Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Shuang-Ho Hospital, New Taipei City, Taiwan
- Division of Colorectal Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
Cheng M, Wang L, Xuan Y, Zhai Z. Identification of genes and pathways associated with menopausal status in breast cancer patients using two algorithms. BMC Womens Health 2024; 24:4. [PMID: 38166892 PMCID: PMC10763477 DOI: 10.1186/s12905-023-02846-7] [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: 08/14/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Menopausal status has a known relationship with the levels of estrogen, progesterone, and other sex hormones, potentially influencing the activity of ER, PR, and many other signaling pathways involved in the initiation and progression of breast cancer. However, the differences between premenopausal and postmenopausal breast cancer patients at the molecular level are unclear. METHODS We retrieved eight datasets from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) associated with menopausal status in breast cancer patients were identified using the MAMA and LIMMA methods. Based on these validated DEGs, we performed Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Protein-protein interaction (PPI) networks were constructed. We used DrugBank data to investigate which of these validated DEGs are targetable. Survival analysis was performed to explore the influence of these genes on breast cancer patient prognosis. RESULTS We identified 762 DEGs associated with menopausal status in breast cancer patients. PPI network analysis indicated that these genes are primarily involved in pathways such as the cell cycle, oocyte meiosis and progesterone-mediated oocyte maturation pathways. Notably, several genes played roles in multiple signaling pathways and were associated with patient survival. These genes were also observed to be targetable according to the DrugBank database. CONCLUSION We identified DEGs associated with menopausal status in breast cancer patients. The association of these genes with several key pathways may promote understanding of the complex characterizations of breast cancer. Our findings offer valuable insights for developing new therapeutic strategies tailored to the menopausal status of breast cancer patients.
Collapse
Affiliation(s)
- Minzhang Cheng
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, the Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Lingchen Wang
- School of Public Health, University of Nevada, Reno, Reno, Nevada, 89557, USA
| | - Yanlu Xuan
- Jiangxi Clinical Research Center for Respiratory Diseases, Jiangxi Institute of Respiratory Disease, the Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhenyu Zhai
- Jiangxi Key Laboratory of Molecular Diagnostics and Precision Medicine, Center for Experimental Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
11
|
Capuozzo M, Celotto V, Santorsola M, Fabozzi A, Landi L, Ferrara F, Borzacchiello A, Granata V, Sabbatino F, Savarese G, Cascella M, Perri F, Ottaiano A. Emerging treatment approaches for triple-negative breast cancer. Med Oncol 2023; 41:5. [PMID: 38038783 DOI: 10.1007/s12032-023-02257-6] [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: 07/21/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
Approximately, 15% of global breast cancer cases are diagnosed as triple-negative breast cancer (TNBC), identified as the most aggressive subtype due to the simultaneous absence of estrogen receptor, progesterone receptor, and HER2. This characteristic renders TNBC highly aggressive and challenging to treat, as it excludes the use of effective drugs such as hormone therapy and anti-HER2 agents. In this review, we explore standard therapies and recent emerging approaches for TNBC, including PARP inhibitors, immune checkpoint inhibitors, PI3K/AKT pathway inhibitors, and cytotoxin-conjugated antibodies. The mechanism of action of these drugs and their utilization in clinical practice is explained in a pragmatic and prospective manner, contextualized within the current landscape of standard therapies for this pathology. These advancements present a promising frontier for tailored interventions with the potential to significantly improve outcomes for TNBC patients. Interestingly, while TNBC poses a complex challenge, it also serves as a paradigm and an opportunity for translational research and innovative therapies in the field of oncology.
Collapse
Affiliation(s)
- Maurizio Capuozzo
- Pharmaceutical Department, ASL Napoli 3, Ercolano, 80056, Naples, Italy
| | - Venere Celotto
- Pharmaceutical Department, ASL Napoli 3, Ercolano, 80056, Naples, Italy
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Antonio Fabozzi
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Loris Landi
- Sanitary District, Ds. 58 ASL Napoli 3, Pompei, 80045, Naples, Italy
| | - Francesco Ferrara
- Pharmaceutical Department, ASL Napoli 3, Via Dell'amicizia 22, Nola, 80035, Naples, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Naples, Italy
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, 84081, Salerno, Italy
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale Srl, Via Padre Carmine Fico 24, Casalnuovo Di, 80013, Naples, Italy
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", via M. Semmola, 80131, Naples, Italy.
| |
Collapse
|
12
|
Guha A, Goswami KK, Sultana J, Ganguly N, Choudhury PR, Chakravarti M, Bhuniya A, Sarkar A, Bera S, Dhar S, Das J, Das T, Baral R, Bose A, Banerjee S. Cancer stem cell-immune cell crosstalk in breast tumor microenvironment: a determinant of therapeutic facet. Front Immunol 2023; 14:1245421. [PMID: 38090567 PMCID: PMC10711058 DOI: 10.3389/fimmu.2023.1245421] [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: 06/23/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC) is globally one of the leading killers among women. Within a breast tumor, a minor population of transformed cells accountable for drug resistance, survival, and metastasis is known as breast cancer stem cells (BCSCs). Several experimental lines of evidence have indicated that BCSCs influence the functionality of immune cells. They evade immune surveillance by altering the characteristics of immune cells and modulate the tumor landscape to an immune-suppressive type. They are proficient in switching from a quiescent phase (slowly cycling) to an actively proliferating phenotype with a high degree of plasticity. This review confers the relevance and impact of crosstalk between immune cells and BCSCs as a fate determinant for BC prognosis. It also focuses on current strategies for targeting these aberrant BCSCs that could open avenues for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Aishwarya Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | | | - Jasmine Sultana
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Pritha Roy Choudhury
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Mohona Chakravarti
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anirban Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Saurav Bera
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sukanya Dhar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Juhina Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Tapasi Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anamika Bose
- Department of Pharmaceutical Technology Biotechnology National Institute of Pharmaceutical Education and Research (NIPER) Sahibzada Ajit Singh (S.A.S.) Nagar, Mohali, Punjab, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
13
|
Yan G, Dai M, Poulet S, Wang N, Boudreault J, Daliah G, Ali S, Lebrun JJ. Combined in vitro/in vivo genome-wide CRISPR screens in triple negative breast cancer identify cancer stemness regulators in paclitaxel resistance. Oncogenesis 2023; 12:51. [PMID: 37932309 PMCID: PMC10628277 DOI: 10.1038/s41389-023-00497-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is defined as lacking the expressions of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). TNBC patients exhibit relatively poor clinical outcomes due to lack of molecular markers for targeted therapies. As such chemotherapy often remains the only systemic treatment option for these patients. While chemotherapy can initially help shrink TNBC tumor size, patients eventually develop resistance to drug, leading to tumor recurrence. We report a combined in vitro/in vivo genome-wide CRISPR synthetic lethality screening approach in a relevant TNBC cell line model to identify several targets responsible for the chemotherapy drug, paclitaxel resistance. Computational analysis integrating in vitro and in vivo data identified a set of genes, for which specific loss-of-function deletion enhanced paclitaxel resistance in TNBC. We found that several of these genes (ATP8B3, FOXR2, FRG2, HIST1H4A) act as cancer stemness negative regulators. Finally, using in vivo orthotopic transplantation TNBC models we showed that FRG2 gene deletion reduced paclitaxel efficacy and promoted tumor metastasis, while increasing FRG2 expression by means of CRISPR activation efficiently sensitized TNBC tumors to paclitaxel treatment and inhibited their metastatic abilities. In summary, the combined in vitro/in vivo genome-wide CRISPR screening approach proved effective as a tool to identify novel regulators of paclitaxel resistance/sensitivity and highlight the FRG2 gene as a potential therapeutical target overcoming paclitaxel resistance in TNBC.
Collapse
Affiliation(s)
- Gang Yan
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Meiou Dai
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Sophie Poulet
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Ni Wang
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Julien Boudreault
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Girija Daliah
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Suhad Ali
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Jean-Jacques Lebrun
- Department of Medicine, Cancer Research Program, McGill University Health Center, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
14
|
Ezeana CF, He T, Patel TA, Kaklamani V, Elmi M, Brigmon E, Otto PM, Kist KA, Speck H, Wang L, Ensor J, Shih YCT, Kim B, Pan IW, Cohen AL, Kelley K, Spak D, Yang WT, Chang JC, Wong STC. A Deep Learning Decision Support Tool to Improve Risk Stratification and Reduce Unnecessary Biopsies in BI-RADS 4 Mammograms. Radiol Artif Intell 2023; 5:e220259. [PMID: 38074778 PMCID: PMC10698614 DOI: 10.1148/ryai.220259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 06/08/2023] [Accepted: 07/07/2023] [Indexed: 01/31/2024]
Abstract
Purpose To evaluate the performance of a biopsy decision support algorithmic model, the intelligent-augmented breast cancer risk calculator (iBRISK), on a multicenter patient dataset. Materials and Methods iBRISK was previously developed by applying deep learning to clinical risk factors and mammographic descriptors from 9700 patient records at the primary institution and validated using another 1078 patients. All patients were seen from March 2006 to December 2016. In this multicenter study, iBRISK was further assessed on an independent, retrospective dataset (January 2015-June 2019) from three major health care institutions in Texas, with Breast Imaging Reporting and Data System (BI-RADS) category 4 lesions. Data were dichotomized and trichotomized to measure precision in risk stratification and probability of malignancy (POM) estimation. iBRISK score was also evaluated as a continuous predictor of malignancy, and cost savings analysis was performed. Results The iBRISK model's accuracy was 89.5%, area under the receiver operating characteristic curve (AUC) was 0.93 (95% CI: 0.92, 0.95), sensitivity was 100%, and specificity was 81%. A total of 4209 women (median age, 56 years [IQR, 45-65 years]) were included in the multicenter dataset. Only two of 1228 patients (0.16%) in the "low" POM group had malignant lesions, while in the "high" POM group, the malignancy rate was 85.9%. iBRISK score as a continuous predictor of malignancy yielded an AUC of 0.97 (95% CI: 0.97, 0.98). Estimated potential cost savings were more than $420 million. Conclusion iBRISK demonstrated high sensitivity in the malignancy prediction of BI-RADS 4 lesions. iBRISK may safely obviate biopsies in up to 50% of patients in low or moderate POM groups and reduce biopsy-associated costs.Keywords: Mammography, Breast, Oncology, Biopsy/Needle Aspiration, Radiomics, Precision Mammography, AI-augmented Biopsy Decision Support Tool, Breast Cancer Risk Calculator, BI-RADS 4 Mammography Risk Stratification, Overbiopsy Reduction, Probability of Malignancy (POM) Assessment, Biopsy-based Positive Predictive Value (PPV3) Supplemental material is available for this article. Published under a CC BY 4.0 license.See also the commentary by McDonald and Conant in this issue.
Collapse
Affiliation(s)
- Chika F. Ezeana
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Tiancheng He
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Tejal A. Patel
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Virginia Kaklamani
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Maryam Elmi
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Erika Brigmon
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Pamela M. Otto
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Kenneth A. Kist
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Heather Speck
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Lin Wang
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Joe Ensor
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Ya-Chen T. Shih
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Bumyang Kim
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - I-Wen Pan
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Adam L. Cohen
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Kristen Kelley
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - David Spak
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | - Wei T. Yang
- From the Department of Systems Medicine and Bioengineering, Houston
Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, Tex (C.F.E.,
T.H., L.W., S.T.C.W.); Houston Methodist Neal Cancer Center, Houston Methodist
Hospital, Houston, Tex (J.E., J.C.C.); Departments of General Oncology (T.A.P.),
Health Services Research (Y.C.T.S., B.K., I.W.P.), and Radiology (D.S., W.T.Y.),
University of Texas MD Anderson Cancer Center, Houston, Tex; University of Texas
Health Science Center, San Antonio, Tex (V.K., M.E., E.B., P.M.O., K.A.K.);
University of the Incarnate Word School of Osteopathic Medicine, San Antonio,
Tex (H.S.); Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
(A.L.C., K.K.); and Department of Radiology, Houston Methodist Hospital, Weill
Cornell Medicine, 6670 Bertner Ave, Houston, TX 77030 (S.T.C.W.)
| | | | | |
Collapse
|
15
|
Hossain F, Ucar DA, Monticone G, Ran Y, Majumder S, Larter K, Luu H, Wyczechowska D, Heidari S, Xu K, Shanthalingam S, Matossian M, Xi Y, Burow M, Collins-Burow B, Del Valle L, Hicks C, Zabaleta J, Golde T, Osborne B, Miele L. Sulindac sulfide as a non-immune suppressive γ-secretase modulator to target triple-negative breast cancer. Front Immunol 2023; 14:1244159. [PMID: 37901240 PMCID: PMC10612326 DOI: 10.3389/fimmu.2023.1244159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) comprises a heterogeneous group of clinically aggressive tumors with high risk of recurrence and metastasis. Current pharmacological treatment options remain largely limited to chemotherapy. Despite promising results, the efficacy of immunotherapy and chemo-immunotherapy in TNBC remains limited. There is strong evidence supporting the involvement of Notch signaling in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, including g-secretase inhibitors (GSIs), are quite effective in preclinical models of TNBC. However, the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects, since Notch plays key roles in T-cell activation, including CD8 T-cells in tumors. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and ideally, do not affect tumor immunity. We identified sulindac sulfide (SS), the active metabolite of FDA-approved NSAID sulindac, as a potential candidate to replace GSIs. Methods We investigated the pharmacological and immunotherapeutic properties of SS in TNBC models in vitro, ex-vivo and in vivo. Results We confirmed that SS, a known γ-secretase modulator (GSM), inhibits Notch1 cleavage in TNBC cells. SS significantly inhibited mammosphere growth in all human and murine TNBC models tested. In a transplantable mouse TNBC tumor model (C0321), SS had remarkable single-agent anti-tumor activity and eliminated Notch1 protein expression in tumors. Importantly, SS did not inhibit Notch cleavage in T- cells, and the anti-tumor effects of SS were significantly enhanced when combined with a-PD1 immunotherapy in our TNBC organoids and in vivo. Discussion Our data support further investigation of SS for the treatment of TNBC, in conjunction with chemo- or -chemo-immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be a cost-effective, rapidly deployable therapeutic option for a patient population in need of more effective therapies.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Deniz A. Ucar
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Giulia Monticone
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Yong Ran
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Kristina Larter
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Hanh Luu
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Dorota Wyczechowska
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Soroor Heidari
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Keli Xu
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | | | - Yaguang Xi
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Matthew Burow
- School of Medicine, Tulane University, New Orleans, LA, United States
| | | | - Luis Del Valle
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
- Department of Pathology, Louisiana State University Health Sciences Center - New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Chindo Hicks
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Todd Golde
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Barbara Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| |
Collapse
|
16
|
Okada N, Ueki C, Shimazaki M, Tsujimoto G, Kohno S, Muranaka H, Yoshikawa K, Takahashi C. NFYA promotes malignant behavior of triple-negative breast cancer in mice through the regulation of lipid metabolism. Commun Biol 2023; 6:596. [PMID: 37268670 DOI: 10.1038/s42003-023-04987-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 05/26/2023] [Indexed: 06/04/2023] Open
Abstract
Two splicing variants exist in NFYA that exhibit high expression in many human tumour types. The balance in their expression correlates with prognosis in breast cancer, but functional differences remain unclear. Here, we demonstrate that NFYAv1, a long-form variant, upregulates the transcription of essential lipogenic enzymes ACACA and FASN to enhance the malignant behavior of triple-negative breast cancer (TNBC). Loss of the NFYAv1-lipogenesis axis strongly suppresses malignant behavior in vitro and in vivo, indicating that the NFYAv1-lipogenesis axis is essential for TNBC malignant behavior and that the axis might be a potential therapeutic target for TNBC. Furthermore, mice deficient in lipogenic enzymes, such as Acly, Acaca, and Fasn, exhibit embryonic lethality; however, Nfyav1-deficient mice exhibited no apparent developmental abnormalities. Our results indicate that the NFYAv1-lipogenesis axis has tumour-promoting effects and that NFYAv1 may be a safe therapeutic target for TNBC.
Collapse
Affiliation(s)
- Nobuhiro Okada
- Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
| | - Chihiro Ueki
- Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Masahiro Shimazaki
- Laboratory for Malignancy Control Research, Medical Innovation Center, Kyoto University, Kyoto, 606-8501, Japan
| | - Goki Tsujimoto
- Graduate School of Interdisciplinary Science & Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Susumu Kohno
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Hayato Muranaka
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Kiyotsugu Yoshikawa
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyoto, 610-0395, Japan
| | - Chiaki Takahashi
- Division of Oncology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| |
Collapse
|
17
|
Abuhadra N, Sun R, Bassett RL, Huo L, Chang JT, Teshome M, Clayborn AR, White JB, Ravenberg EE, Adrada BE, Candelaria RP, Yang W, Ding Q, Symmans WF, Arun B, Damodaran S, Koenig KB, Layman RM, Lim B, Litton JK, Thompson A, Ueno NT, Piwnica-Worms H, Hortobagyi GN, Valero V, Tripathy D, Rauch GM, Moulder S, Yam C. Targeting chemotherapy resistance in mesenchymal triple-negative breast cancer: a phase II trial of neoadjuvant angiogenic and mTOR inhibition with chemotherapy. Invest New Drugs 2023:10.1007/s10637-023-01357-4. [PMID: 37043123 DOI: 10.1007/s10637-023-01357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/27/2023] [Indexed: 04/13/2023]
Affiliation(s)
- Nour Abuhadra
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Ryan Sun
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roland L Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mediget Teshome
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alyson R Clayborn
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jason B White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Elizabeth E Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Beatriz E Adrada
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rosalind P Candelaria
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Yang
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kimberly B Koenig
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Rachel M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Alastair Thompson
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gaiane M Rauch
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stacy Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
Feng J, Wang L, Zhang K, Ni S, Li B, Liu J, Wang D. Identification and panoramic analysis of drug response-related genes in triple negative breast cancer using as an example NVP-BEZ235. Sci Rep 2023; 13:5984. [PMID: 37045929 PMCID: PMC10097725 DOI: 10.1038/s41598-023-32757-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
Taking NVP-BEZ235 (BEZ235) as an example to screen drug response-related genes (DRRGs) and explore their potential value in triple-negative breast cancer (TNBC). Through high-throughput technique, multidimensional transcriptome expression data (mRNA, miRNA and lncRNA) of BEZ235-treated and -untreated MDA-MB-468 cell lines were obtained. Combined with transcriptome data of the MDA-MB-468 cells and TCGA-TNBC tissues, differential gene expression analysis and WGCNA were performed to identify DRRGs associated with tumor trait by simulating the drug response microenvironment (DRM) of BEZ235-treated patients. Based on DRRGs, we constructed a ceRNA network and verified the expression levels of three key molecules by RT-qPCR, which not only demonstrated the successful construction of a BEZ235-treated cell line model but also explained the antitumor mechanism of BEZ235. Four molecular subtypes related to the DRM with survival difference were proposed using cluster analysis, namely glycolysis subtype, proliferation depression subtype, immune-suppressed subtype, and immune-activated subtype. A novel prognostic signature consisting of four DRRGs was established by Lasso-Cox analysis, which exhibited outstanding performance in predicting overall survival compared with several excellent reported signatures. The high- and low-risk groups were characterized by enrichment of metabolism-related pathways and immune-related pathways, respectively. Of note, the low-risk group had a better response to immune checkpoint blockade. Besides, pRRophetic analysis found that patients in the low-risk group were more sensitive to methotrexate and cisplation, whereas more resistant to BEZ235, docetaxel and paclitaxel. In conclusion, the DRRGs exemplified by BEZ235 are potential biomarkers for TNBC molecular typing, prognosis prediction and targeted therapy. The novel DRRGs-guided strategy for predicting the subtype, survival and therapy efficacy, might be also applied to more cancers and drugs other than TNBC and BEZ235.
Collapse
Affiliation(s)
- Jia Feng
- Department of Clinical Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Luchang Wang
- Department of Clinical Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Kaijiong Zhang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Sujiao Ni
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Baolin Li
- Department of Clinical Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Dongsheng Wang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
19
|
Hoogenboezem EN, Patel SS, Cavnar AB, Lo JH, Babb LM, Francini N, Patil P, Colazo JM, Michell DL, Sanchez VM, McCune JT, Ma J, DeJulius CR, Lee LH, Rosch JC, Allen RM, Stokes LD, Hill JL, Vickers KC, Cook RS, Duvall CL. Structural Optimization of siRNA Conjugates for Albumin Binding Achieves Effective MCL1-Targeted Cancer Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528574. [PMID: 36824780 PMCID: PMC9948981 DOI: 10.1101/2023.02.14.528574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
The high potential for therapeutic application of siRNAs to silence traditionally undruggable oncogenic drivers remains largely untapped due to the challenges of tumor cell delivery. Here, siRNAs were optimized for in situ binding to albumin through C18 lipid modifications to improve pharmacokinetics and tumor delivery. Systematic variation of siRNA conjugates revealed a lead structure with divalent C18 lipids each linked through three repeats of hexaethylene glycol connected by phosphorothioate bonds. Importantly, we discovered that locating the branch site of the divalent lipid structure proximally (adjacent to the RNA) rather than at a more distal site (after the linker segment) promotes association with albumin, while minimizing self-assembly and lipoprotein association. Comparison to higher albumin affinity (diacid) lipid variants and siRNA directly conjugated to albumin underscored the importance of conjugate hydrophobicity and reversibility of albumin binding for siRNA delivery and bioactivity in tumors. The lead conjugate increased tumor siRNA accumulation 12-fold in orthotopic mouse models of triple negative breast cancer over the parent siRNA. When applied for silencing of the anti-apoptotic oncogene MCL-1, this structure achieved approximately 80% MCL1 silencing in orthotopic breast tumors. Furthermore, application of the lead conjugate structure to target MCL1 yielded better survival outcomes in three independent, orthotopic, triple negative breast cancer models than an MCL1 small molecule inhibitor. These studies provide new structure-function insights on optimally leveraging siRNA-lipid conjugate structures that associate in situ with plasma albumin for molecular-targeted cancer therapy.
Collapse
Affiliation(s)
| | - Shrusti S. Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Ashley B. Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Justin H. Lo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Lauren M. Babb
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Prarthana Patil
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Violeta M. Sanchez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joshua T. McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Jinqi Ma
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | | | | | - Jonah C. Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Ryan M. Allen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Larry D. Stokes
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Jordan L. Hill
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Kasey C. Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|
20
|
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: 7] [Impact Index Per Article: 3.5] [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
|
21
|
Liu X, Bai M, Li H, Ye P, Duan X, Wu C, Huang Z, Lu S, Zhang J, Zhao Z, Guo F, You R, Qin W, Wang W, Han A, Shen L, Wang Y, Zhao Z, Luo H, Wu J. Single-cell RNA-sequencing uncovers compound kushen injection synergistically improves the efficacy of chemotherapy by modulating the tumor environment of breast cancer. Front Immunol 2022; 13:965342. [DOI: 10.3389/fimmu.2022.965342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDue to lack of enough specific targets and the immunosuppressive tumor microenvironment (TME) of triple-negative breast cancer (TNBC), TNBC patients often cannot benefit from a single treatment option. This study aims to explore the regulatory effects of Compound kushen injection (CKI) plus chemotherapy on the TME of TNBC from a single cell level.MethodsA mouse TNBC model in BALB/c mice was established to evaluate the antitumor efficacy and toxicity of CKI combined with chemotherapy. Flow cytometry was used to observe the influence of CKI on the lymphocyte populations in the tumor bearing mice. Both bulk RNA sequencing (RNA-seq) and single-cell RNA-seq (scRNA-seq) were applied to portray the modulation of CKI combined with chemotherapy on the TME of TNBC mice.ResultsCKI significantly enhanced the anticancer activity of chemotherapy in vivo with no obvious side effects. Flow cytometry results revealed a significantly higher activation of CD8+ T lymphocytes in the spleens and tumors of the mice with combination therapy. Bulk RNA-seq indicated that CKI could promote the cytotoxic immune cell infiltrating into tumor tissues. Meanwhile, scRNA-seq further revealed that CKI combined with chemotherapy could enhance the percentage of tumor-infiltrating CD8+ T cells, inhibit tumor-promoting signaling pathways, and promote T cell activation and positive regulation of immune response. In addition, CKI showed obvious anticancer activity against MDA-MB-231 breast tumor cells in vitro.ConclusionsThe combination of CKI and chemotherapy might provide a higher efficiency and lower toxicity strategy than a single chemotherapy drug for TNBC. CKI potentiates the anti-TNBC effects of chemotherapy by activating anti-tumor immune response in mice.
Collapse
|
22
|
Monti E, Mancini A, Marras E, Gariboldi MB. Targeting Mitochondrial ROS Production to Reverse the Epithelial-Mesenchymal Transition in Breast Cancer Cells. Curr Issues Mol Biol 2022; 44:5277-5293. [PMID: 36354671 PMCID: PMC9689492 DOI: 10.3390/cimb44110359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 03/03/2024] Open
Abstract
Experimental evidence implicates reactive oxygen species (ROS) generation in the hypoxic stabilization of hypoxia-inducible factor (HIF)-1α and in the subsequent expression of promoters of tumor invasiveness and metastatic spread. However, the role played by mitochondrial ROS in hypoxia-induced Epithelial-Mesenchymal Transition (EMT) activation is still unclear. This study was aimed at testing the hypothesis that the inhibition of hypoxia-induced mitochondrial ROS production, mainly at the mitochondrial Complex III UQCRB site, could result in the reversion of EMT, in addition to decreased HIF-1α stabilization. The role of hypoxia-induced ROS increase in HIF-1α stabilization and the ability of antioxidants, some of which directly targeting mitochondrial Complex III, to block ROS production and HIF-1α stabilization and prevent changes in EMT markers were assessed by evaluating ROS, HIF-1α and EMT markers on breast cancer cells, following 48 h treatment with the antioxidants. The specific role of UQCRB in hypoxia-induced EMT was also evaluated by silencing its expression through RNA interference and by assessing the effects of its downregulation on ROS production, HIF-1α levels, and EMT markers. Our results confirm the pivotal role of UQCRB in hypoxic signaling inducing EMT. Thus, UQCRB might be a new therapeutic target for the development of drugs able to reverse EMT by blocking mitochondrial ROS production.
Collapse
Affiliation(s)
- Elena Monti
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Alessandro Mancini
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- BioUp Sagl, 6900 Lugano, Switzerland
| | - Emanuela Marras
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Marzia Bruna Gariboldi
- Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| |
Collapse
|
23
|
Racial Disparity in Quadruple Negative Breast Cancer: Aggressive Biology and Potential Therapeutic Targeting and Prevention. Cancers (Basel) 2022; 14:cancers14184484. [PMID: 36139643 PMCID: PMC9497140 DOI: 10.3390/cancers14184484] [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: 06/30/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Quadruple negative breast cancer (QNBC), a subgroup of triple negative BC, has emerged as a highly aggressive BC subtype that disproportionately afflicts and impacts Black/African-American (AA) women. In this article, we review molecular distinctions in Black/AA and White/European-American (EA) QNBC biology as well as address potential non-genetic risk factors that could be underlying this racially disparate burden. We aim to provide deeper insight and provide a framework for novel discovery of actionable therapeutic targets and identify lifestyle changes to improve outcomes for Black/AA QNBC patients. Abstract Black/African-American (AA) women, relative to their White/European-American (EA) counterparts, experience disproportionately high breast cancer mortality. Central to this survival disparity, Black/AA women have an unequal burden of aggressive breast cancer subtypes, such as triple-negative breast cancer (ER/PR-, HER2-wild type; TNBC). While TNBC has been well characterized, recent studies have identified a highly aggressive androgen receptor (AR)-negative subtype of TNBC, quadruple-negative breast cancer (ER/PR-, HER2-wildtype, AR-; QNBC). Similar to TNBC, QNBC disproportionately impacts Black/AA women and likely plays an important role in the breast cancer survival disparities experienced by Black/AA women. Here, we discuss the racial disparities of QNBC and molecular signaling pathways that may contribute to the aggressive biology of QNBC in Black/AA women. Our immediate goal is to spotlight potential prevention and therapeutic targets for Black/AA QNBC; ultimately our goal is to provide greater insight into reducing the breast cancer survival burden experienced by Black/AA women.
Collapse
|
24
|
Fleisher B, Werkman C, Jacobs B, Varkey J, Taha K, Ait-Oudhia S. KIFC1: A Reliable Prognostic Biomarker in Rb-positive Triple-negative Breast Cancer Patients Treated With Doxorubicin in Combination With Abemaciclib. CANCER DIAGNOSIS & PROGNOSIS 2022; 2:525-532. [PMID: 36060015 PMCID: PMC9425577 DOI: 10.21873/cdp.10137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND/AIM Triple-negative breast cancer (TNBC) prevalence and risk of relapse are greatest in African American (AA) patients. Doxorubicin (DOX) and abemaciclib (ABE) synergism in Rb-positive TNBC cells (MDA-MB-231), and antagonism in Rb-negative TNBC cells (MDA-MB-468) have been previously shown. Here, we assessed Kinesin-like protein 1 (KIFC1) as an ethnic-specific prognostic biomarker of the DOX+ABE combination for the Rb-status in TNBC. MATERIALS AND METHODS Literature search for TNBC prognostic biomarkers in the AA population was conducted. MDA-MB-231 and MDA-MB-468 cells were exposed over 72 h to four treatment arms: 1) control (medium without drug), 2) DOX at 50% inhibitory concentration in MDA-MB-231 (0.565 μM) and MDA-MB-468 (0.121 μM), 3) ABE alone (2 μM), and 4) DOX+ABE combination at their corresponding concentrations in each cell-line. KIFC1 protein expression and temporal changes were quantified in MDA-MB-231 cells using western blot. RESULTS KIFC1, Kaiso, and Annexin A2 are literature-identified AA-specific TNBC prognostic biomarkers. KIFC1 was found to be uncorrelated to other proposed biomarkers, suggesting it may predict risk independently of other TNBC biomarkers. In both cell lines, DOX alone did not significantly change KIFC1 expression relative to control. Conversely, ABE reduced KIFC1 expression in MDA-MB-231 but not in MDA-MB-468 cells. The combination DOX+ABE resulted in a greatest reduction in KIFC1 in MDA-MB-231 cells with a more rapid time-to-full inhibition of KIFC1 compared to ABE alone. CONCLUSION Change in KIFC1 expression is primarily driven by ABE in Rb-positive TNBC cells. DOX increases ABE speed to achieve a full inhibition of KIFC1 in Rb-positive, yet, without influencing its expression in Rb-negative TNBC cells.
Collapse
Affiliation(s)
- Brett Fleisher
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, College of Pharmacy, Orlando, FL, U.S.A
| | - Carolin Werkman
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, College of Pharmacy, Orlando, FL, U.S.A
| | - Brehanna Jacobs
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, College of Pharmacy, Orlando, FL, U.S.A
| | - Justin Varkey
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, College of Pharmacy, Orlando, FL, U.S.A
| | - Kareem Taha
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida, College of Pharmacy, Orlando, FL, U.S.A
| | - Sihem Ait-Oudhia
- Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co., Inc, Kenilworth, NJ, U.S.A
| |
Collapse
|
25
|
Wang J, Li Q, Luo Y, Han Y, Ma F, Cai R, Li Q, Fan Y, Wang J, Zhang P, Xu B. Development and external validation of a clinical nomogram for individually predicting survival of metaplastic breast cancer. Clin Breast Cancer 2022; 22:e798-e806. [DOI: 10.1016/j.clbc.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/03/2022]
|
26
|
Jiang D, Qiu T, Peng J, Li S, Tala, Ren W, Yang C, Wen Y, Chen CH, Sun J, Wu Y, Liu R, Zhou J, Wu K, Liu W, Mao X, Zhou Z, Chen C. YB-1 is a positive regulator of KLF5 transcription factor in basal-like breast cancer. Cell Death Differ 2022; 29:1283-1295. [PMID: 35022570 PMCID: PMC9177637 DOI: 10.1038/s41418-021-00920-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/25/2022] Open
Abstract
Y-box binding protein 1 (YB-1) is a well-known oncogene highly expressed in various cancers, including basal-like breast cancer (BLBC). Beyond its role as a transcription factor, YB-1 is newly defined as an epigenetic regulator involving RNA 5-methylcytosine. However, its specific targets and pro-cancer functions are poorly defined. Here, based on clinical database, we demonstrate a positive correlation between Kruppel-like factor 5 (KLF5) and YB-1 expression in breast cancer patients, but a negative correlation with that of Dachshund homolog 1 (DACH1). Mechanistically, YB-1 enhances KLF5 expression not only through transcriptional activation that can be inhibited by DACH1, but also by stabilizing KLF5 mRNA in a RNA 5-methylcytosine modification-dependent manner. Additionally, ribosomal S6 kinase 2 (RSK2) mediated YB-1 phosphorylation at Ser102 promotes YB-1/KLF5 transcriptional complex formation, which co-regulates the expression of BLBC specific genes, Keratin 16 (KRT16) and lymphocyte antigen 6 family member D (Ly6D), to promote cancer cell proliferation. The RSK inhibitor, LJH685, suppressed BLBC cell tumourigenesis in vivo by disturbing YB-1-KLF5 axis. Our data suggest that YB-1 positively regulates KLF5 at multiple levels to promote BLBC progression. The novel RSK2-YB-1-KLF5-KRT16/Ly6D axis provides candidate diagnostic markers and therapeutic targets for BLBC.
Collapse
Affiliation(s)
- Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China
| | - Ting Qiu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China
| | - Junjiang Peng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Siyuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tala
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Wenlong Ren
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- College of Life Sciences, China University of Science and Technology, Hefei, Anhui, China
| | - Chuanyu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yi Wen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Chuan-Huizi Chen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Jian Sun
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China
| | - Yingying Wu
- The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Rong Liu
- The First Affiliated Hospital, Peking University, Beijing, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Wen Liu
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Zhongmei Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- Kunming College of Lifesciences, University of Chinese Academy Sciences, Kunming, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
27
|
Yu B, Su J, Shi Q, Liu Q, Ma J, Ru G, Zhang L, Zhang J, Hu X, Tang J. KMT5A-methylated SNIP1 promotes triple-negative breast cancer metastasis by activating YAP signaling. Nat Commun 2022; 13:2192. [PMID: 35449131 PMCID: PMC9023492 DOI: 10.1038/s41467-022-29899-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 04/01/2022] [Indexed: 12/15/2022] Open
Abstract
Smad nuclear-interacting protein 1 (SNIP1) is a transcription repressor related to the TGF-β signaling pathway and associates with c-MYC, a key regulator of cell proliferation and tumor development. Currently, the mechanism by which SNIP1 regulates tumorigenesis and cancer metastasis is unknown. Here, we identify that SNIP1 is a non-histone substrate of lysine methyltransferase KMT5A, which undergoes KMT5A-mediated mono-methylation to promote breast cancer cell growth, invasion and lung metastasis. Mechanistically, we show KMT5A-mediated K301 methylation of SNIP1 represents a sensing signal to release histone acetyltransferase KAT2A and promotes the interaction of c-MYC and KAT2A, and the recruitment of c-MYC/KAT2A complex to promoter of c-MYC targets. This event ultimately inhibits the Hippo kinase cascade to enhance triple-negative breast cancer (TNBC) metastasis by transcriptionally activating MARK4. Co-inhibition of KMT5A catalytic activity and YAP in TNBC xenograft-bearing animals attenuates breast cancer metastasis and increases survival. Collectively, this study presents an KMT5A methylation-dependent regulatory mechanism governing oncogenic function of SNIP1. SNIP1 methylation initiates its oncogenic functions. Here, the authors show that SNIP1 is methylated by KMT5A and this leads to downstream signalling that activates the YAP pathway, resulting in tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Bo Yu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Jun Su
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025, P. R. China
| | - Qiqi Shi
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jun Ma
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Guoqing Ru
- Department of Pathology, Zhejiang Provincial People's Hospital, People' s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, P. R. China
| | - Lei Zhang
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China. .,Department of Phase I Clinical Trial Center, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| | - Jianming Tang
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, P. R. China.
| |
Collapse
|
28
|
Identification of a novel mechanism for reversal of doxorubicin-induced chemotherapy resistance by TXNIP in triple-negative breast cancer via promoting reactive oxygen-mediated DNA damage. Cell Death Dis 2022; 13:338. [PMID: 35414060 PMCID: PMC9005717 DOI: 10.1038/s41419-022-04783-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022]
Abstract
Given that triple-negative breast cancer (TNBC) lacks specific receptors (estrogen and progesterone receptors and human epidermal growth factor receptor 2) and cannot be treated with endocrine therapy, chemotherapy has remained the mainstay of treatment. Drug resistance is reportedly the main obstacle to the clinical use of doxorubicin (DOX) in this patient population. Accordingly, screening molecules related to chemoresistance and studying their specific mechanisms has clinical significance for improving the efficacy of chemotherapy in TNBC patients. Thioredoxin-interacting protein (TXNIP) is a metabolism-related protein that plays a tumor suppressor role in various malignant tumors; however, the specific role of TXNIP in tumor chemoresistance has not been reported. In the present study, we explored the potential molecular mechanism of TXNIP in the chemoresistance of TNBC for the first time. The results showed that TXNIP inhibited the proliferation of TNBC drug-resistant cells and promoted apoptosis in vitro and in vivo. Furthermore, TXNIP promoted the synthesis of reactive oxygen species (ROS) and the accumulation of DNA damage caused by DOX and increased γ-H2AX levels in a time and dose-dependent manner. Moreover, ROS scavenger pretreatment could block DNA damage induced by TXNIP and restore the resistance of TNBC resistant cells to DOX to a certain extent. In addition, we found that the small molecule c-Myc inhibitor 10058-F4 promoted TXNIP expression, increased ROS synthesis in cells, and could enhance the cytotoxicity of chemotherapy drugs in vitro and in vivo when combined with DOX. These results indicated that c-Myc inhibitor 10058-F4 could induce TXNIP upregulation in TNBC drug-resistant cells, and the upregulated TXNIP increased the accumulation of ROS-dependent DNA damage, thereby decreasing chemotherapy resistance of TNBC. Our findings reveal a new mechanism of mediating drug resistance and provide a new drug combination strategy to overcome DOX resistance in TNBC.
Collapse
|
29
|
An Efficient Algorithm for the Detection of Outliers in Mislabeled Omics Data. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2021:9436582. [PMID: 34976114 PMCID: PMC8716222 DOI: 10.1155/2021/9436582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/30/2021] [Indexed: 11/18/2022]
Abstract
High dimensionality and noise have made it difficult to detect related biomarkers in omics data. Through previous study, penalized maximum trimmed likelihood estimation is effective in identifying mislabeled samples in high-dimensional data with mislabeled error. However, the algorithm commonly used in these studies is the concentration step (C-step), and the C-step algorithm that is applied to robust penalized regression does not ensure that the criterion function is gradually optimized iteratively, because the regularized parameters change during the iteration. This makes the C-step algorithm runs very slowly, especially when dealing with high-dimensional omics data. The AR-Cstep (C-step combined with an acceptance-rejection scheme) algorithm is proposed. In simulation experiments, the AR-Cstep algorithm converged faster (the average computation time was only 2% of that of the C-step algorithm) and was more accurate in terms of variable selection and outlier identification than the C-step algorithm. The two algorithms were further compared on triple negative breast cancer (TNBC) RNA-seq data. AR-Cstep can solve the problem of the C-step not converging and ensures that the iterative process is in the direction that improves criterion function. As an improvement of the C-step algorithm, the AR-Cstep algorithm can be extended to other robust models with regularized parameters.
Collapse
|
30
|
Etiopathogenesis of Breast Cancer and Prevention. Breast Cancer 2022. [DOI: 10.1007/978-981-16-4546-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
Tian JH, Liu SH, Yu CY, Wu LG, Wang LB. The Role of Non-Coding RNAs in Breast Cancer Drug Resistance. Front Oncol 2021; 11:702082. [PMID: 34589423 PMCID: PMC8473733 DOI: 10.3389/fonc.2021.702082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is one of the commonly occurring malignancies in females worldwide. Despite significant advances in therapeutics, the mortality and morbidity of BC still lead to low survival and poor prognosis due to the drug resistance. There are certain chemotherapeutic, endocrine, and target medicines often used for BC patients, including anthracyclines, taxanes, docetaxel, cisplatin, and fluorouracil. The drug resistance mechanisms of these medicines are complicated and have not been fully elucidated. It was reported that non-coding RNAs (ncRNAs), such as micro RNAs (miRNA), long-chain non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) performed key roles in regulating tumor development and mediating therapy resistance. However, the mechanism of these ncRNAs in BC chemotherapeutic, endocrine, and targeted drug resistance was different. This review aims to reveal the mechanism and potential functions of ncRNAs in BC drug resistance and to highlight the ncRNAs as a novel target for achieving improved treatment outcomes for BC patients.
Collapse
Affiliation(s)
- Jin-Hai Tian
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| | - Shi-Hai Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuan-Yang Yu
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| | - Li-Gang Wu
- Department of Oncology, General Hospital of Ningxia Medical University, Yingchuan, China
| | - Li-Bin Wang
- The Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, China, Yinchuan, China.,The Clinical Medicine College of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
32
|
Recurrence biomarkers of triple negative breast cancer treated with neoadjuvant chemotherapy and anti-EGFR antibodies. NPJ Breast Cancer 2021; 7:124. [PMID: 34535679 PMCID: PMC8448841 DOI: 10.1038/s41523-021-00334-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
To find metastatic recurrence biomarkers of triple-negative breast cancer (TNBC) treated by neoadjuvant chemotherapy and anti-EGFR antibodies (NAT), we evaluated tumor genomic, transcriptomic, and immune features, using MSK-IMPACT assay, gene arrays, Nanostring technology, and TIL assessment on H&E. Six patients experienced a rapid fatal recurrence (RR) and other 6 had later non-fatal recurrences (LR). Before NAT, RR had low expression of 6 MHC class I and 13 MHC class II genes but were enriched in upregulated genes involved in the cell cycle-related pathways. Their TIL number before NAT in RR was very low (<5%) and did not increase after treatment. In post-NAT residual tumors, RR cases showed high expression of SOX2 and CXCR4. Our results indicate that high expression of cell cycle genes, combined with cold immunological phenotype, may predict strong TNBC resistance to NAT and rapid progression after it. This biomarker combination is worth validation in larger studies.
Collapse
|
33
|
Fallica A, Barbaraci C, Amata E, Pasquinucci L, Turnaturi R, Dichiara M, Intagliata S, Gariboldi MB, Marras E, Orlandi VT, Ferroni C, Martini C, Rescifina A, Gentile D, Varchi G, Marrazzo A. Nitric Oxide Photo-Donor Hybrids of Ciprofloxacin and Norfloxacin: A Shift in Activity from Antimicrobial to Anticancer Agents. J Med Chem 2021; 64:11597-11613. [PMID: 34319100 PMCID: PMC8389907 DOI: 10.1021/acs.jmedchem.1c00917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Indexed: 12/11/2022]
Abstract
The potential anticancer effect of fluoroquinolone antibiotics has been recently unveiled and related to their ability to interfere with DNA topoisomerase II. We herein envisioned the design and synthesis of novel Ciprofloxacin and Norfloxacin nitric oxide (NO) photo-donor hybrids to explore the potential synergistic antitumor effect exerted by the fluoroquinolone scaffold and NO eventually produced upon light irradiation. Anticancer activity, evaluated on a panel of tumor cell lines, showed encouraging results with IC50 values in the low micromolar range. Some compounds displayed intense antiproliferative activity on triple-negative and doxorubicin-resistant breast cancer cell lines, paving the way for their potential use to treat aggressive, refractory and multidrug-resistant breast cancer. No significant additive effect was observed on PC3 and DU145 cells following NO release. Conversely, antimicrobial photodynamic experiments on both Gram-negative and Gram-positive microorganisms displayed a significant killing rate in Staphylococcus aureus, accounting for their potential effectiveness as selective antimicrobial photosensitizers.
Collapse
Affiliation(s)
- Antonino
Nicolò Fallica
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Carla Barbaraci
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Lorella Pasquinucci
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Rita Turnaturi
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Sebastiano Intagliata
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Marzia Bruna Gariboldi
- Department
of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy
| | - Emanuela Marras
- Department
of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy
| | - Viviana Teresa Orlandi
- Department
of Biotechnology and Life Sciences (DBSV), University of Insubria, Via JH Dunant 3, 21100 Varese, Italy
| | - Claudia Ferroni
- Institute
for the Organic Synthesis and Photoreactivity − ISOF, Via Piero Gobetti, 101, 40129 Bologna, Italy
| | - Cecilia Martini
- Institute
for the Organic Synthesis and Photoreactivity − ISOF, Via Piero Gobetti, 101, 40129 Bologna, Italy
| | - Antonio Rescifina
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Davide Gentile
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| | - Greta Varchi
- Institute
for the Organic Synthesis and Photoreactivity − ISOF, Via Piero Gobetti, 101, 40129 Bologna, Italy
| | - Agostino Marrazzo
- Department
of Drug and Health Sciences (DSFS), University
of Catania, Viale A. Doria, 6, 95125 Catania, Italy
| |
Collapse
|
34
|
Hu Y, Gao J, Wang M, Li M. Potential Prospect of CDK4/6 Inhibitors in Triple-Negative Breast Cancer. Cancer Manag Res 2021; 13:5223-5237. [PMID: 34234565 PMCID: PMC8257068 DOI: 10.2147/cmar.s310649] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/03/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive, difficult-to-treat subtype of cancer with a poor prognosis; there is an urgent need for effective, targeted molecular therapies. The cyclin D/cyclin-dependent kinase (CDK)4/6–retinoblastoma protein (Rb) pathway plays a critical role in regulating cell cycle checkpoints, a process which is often disrupted in cancer cells. Selective CDK4/6 inhibitors can prevent retinoblastoma protein phosphorylation by invoking cell cycle arrest in the first growth phase (G1), and may therefore represent an effective treatment option. In this article, we review the molecular mechanisms and therapeutic efficacy of CDK4/6 inhibitors in combination with other targeted therapies for the treatment of triple-negative breast cancer. Three selective CDK4/6 inhibitors have so far received the approval of the Food and Drug Administration (FDA) for patients with estrogen receptor (ER)+/human epidermal growth factor receptor 2 (HER2) breast cancer. Trilaciclib, a small molecule short-acting inhibitor of CDK4/6, has also been approved recently for people with small cell lung cancer, and is also expected to be clinically effective against breast cancer. Although the efficacy of CDK4/6 inhibitors in patients with triple-negative breast cancer remains uncertain, their use in conjunction with other targeted therapies may improve outcomes and is therefore currently being explored. Identifying biomarkers for response or resistance to CDK4/6 inhibitor treatment may optimize the personalization of treatment strategies for this disease. Ongoing and future clinical trials and biomarker studies will shed further light on these topics, and help to realize the full potential of CDK4/6 inhibitor treatment in triple-negative breast cancer.
Collapse
Affiliation(s)
- Ye Hu
- Department of Oncology & Department of Breast Surgery, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Jiyue Gao
- Department of Oncology & Department of Breast Surgery, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Meiling Wang
- Department of Oncology & Department of Breast Surgery, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Man Li
- Department of Oncology & Department of Breast Surgery, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| |
Collapse
|
35
|
van Barele M, Heemskerk-Gerritsen BAM, Louwers YV, Vastbinder MB, Martens JWM, Hooning MJ, Jager A. Estrogens and Progestogens in Triple Negative Breast Cancer: Do They Harm? Cancers (Basel) 2021; 13:2506. [PMID: 34063736 PMCID: PMC8196589 DOI: 10.3390/cancers13112506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancers (TNBC) occur more frequently in younger women and do not express estrogen receptor (ER) nor progesterone receptor (PR), and are therefore often considered hormone-insensitive. Treatment of premenopausal TNBC patients almost always includes chemotherapy, which may lead to premature ovarian insufficiency (POI) and can severely impact quality of life. Hormone replacement therapy (HRT) is contraindicated for patients with a history of hormone-sensitive breast cancer, but the data on safety for TNBC patients is inconclusive, with a few randomized trials showing increased risk-ratios with wide confidence intervals for recurrence after HRT. Here, we review the literature on alternative pathways from the classical ER/PR. We find that for both estrogens and progestogens, potential alternatives exist for exerting their effects on TNBC, ranging from receptor conversion, to alternative receptors capable of binding estrogens, as well as paracrine pathways, such as RANK/RANKL, which can cause progestogens to indirectly stimulate growth and metastasis of TNBC. Finally, HRT may also influence other hormones, such as androgens, and their effects on TNBCs expressing androgen receptors (AR). Concluding, the assumption that TNBC is completely hormone-insensitive is incorrect. However, the direction of the effects of the alternative pathways is not always clear, and will need to be investigated further.
Collapse
Affiliation(s)
- Mark van Barele
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Bernadette A. M. Heemskerk-Gerritsen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Yvonne V. Louwers
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Mijntje B. Vastbinder
- Department of Internal Medicine, Ijsselland Hospital, Prins Constantijnweg 2, 2906 ZC Capelle aan den IJssel, The Netherlands;
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| |
Collapse
|
36
|
Ji X, Guo D, Ma J, Yin M, Yu Y, Liu C, Zhou Y, Sun J, Li Q, Chen N, Fan C, Song H. Epigenetic Remodeling Hydrogel Patches for Multidrug-Resistant Triple-Negative Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100949. [PMID: 33792093 DOI: 10.1002/adma.202100949] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Indexed: 05/14/2023]
Abstract
The induced expansion of tumor-initiating cells (T-ICs) upon repeated exposure of tumors to chemotherapeutic drugs forms a major cause for chemoresistance and cancer metastasis. Here, a tumor-microenvironment-responsive hydrogel patch is designed to modulate the plasticity of T-ICs in triple-negative breast cancer (TNBC), which is insensitive to hormone- and HER2-targeting. The on-site formation of the hydrogel network patches tumors in a chemoresistant TNBC murine model and senses intratumoral reactive oxygen species for linker cleavage and payload release. Patch-mediated inhibition of the histone demethylase lysine-specific demethylase 1 (LSD1) epigenetically regulates the switch of T-ICs from self-renewal to differentiation, rehabilitating their chemosensitivity. Moreover, the hydrogel patch enhances tumor immunogenicity and increases T-cell infiltration via epigenetic activation of innate immunity. A single-dose of the hydrogel patch harboring LSD1 inhibitor and chemotherapy agent efficiently suppresses tumor growth, postsurgical relapse, and metastasis. The superior efficacy against multidrug resistance further reveals the broad applicability of epigenetic remodeling hydrogel patches.
Collapse
Affiliation(s)
- Xiaoyuan Ji
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Daoxia Guo
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Min Yin
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Yun Yu
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chang Liu
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanfeng Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinli Sun
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Nan Chen
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
37
|
Chou CW, Huang YM, Chang YJ, Huang CY, Hung CS. Identified the novel resistant biomarkers for taxane-based therapy for triple-negative breast cancer. Int J Med Sci 2021; 18:2521-2531. [PMID: 34104083 PMCID: PMC8176163 DOI: 10.7150/ijms.59177] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022] Open
Abstract
Developing treatment strategies for triple-negative breast cancer (TNBC) has become an important clinical challenge. Currently, taxane-based chemotherapy is one of the standard treatments for TNBC. However, determining the key factor of taxane-resistance is urgently in need for clinical treatment for breast cancer. We used GEO data to generate paclitaxel resistance in two basal-like TNBC cell lines (SUM149 and MDA-MB-468). Seventy-one common upregulated differentially expressed genes (DEGs) and 11 downregulated DEGs were found to be related to paclitaxel resistance. By constructing protein-protein interactions, 28 hub proteins with a degree cutoff criterion of ≥1 were found. Nine hub genes (COL4A6, COL4A5, IL6, PDGFA, LPAR1, FYB, IL20, IL18R1 and INHBA) are involved in important signaling pathways. We found that upregulated PDGFA and downregulated COL4A6 were significantly associated with an insensitive response to neoadjuvant paclitaxel-based therapy. A Kaplan-Meier plot was created to check the prognostic values of 11 hub DEGs in terms of recurrence-free survival. High expressions of PDGFA and LAMB3 were correlated with poor recurrence-free survival, while low levels of FYB, IL18R1, and RASGRP1 indicated poorer relapse-free survival. Our results suggest that PDGFA, COL4A6, LPAR1, FYB, COL4A5, and RASGRP1 might be candidate target genes for taxane-based therapy in basal-like TNBC.
Collapse
Affiliation(s)
- Ching-Wen Chou
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Min Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Section of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chien-Yu Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
- Division of Colonrectal Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University
| | - Chin-Sheng Hung
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
| |
Collapse
|
38
|
Hoang VT, Matossian MD, La J, Hoang K, Ucar DA, Elliott S, Burks HE, Wright TD, Patel S, Bhatt A, Phamduy T, Chrisey D, Buechlein A, Rusch DB, Nephew KP, Anbalagan M, Rowan B, Cavanaugh JE, Flaherty PT, Miele L, Collins-Burow BM, Burow ME. Dual inhibition of MEK1/2 and MEK5 suppresses the EMT/migration axis in triple-negative breast cancer through FRA-1 regulation. J Cell Biochem 2021; 122:835-850. [PMID: 33876843 DOI: 10.1002/jcb.29916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023]
Abstract
Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. Constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway has been linked to chemoresistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT) when cells adopt a motile and invasive phenotype through loss of epithelial markers (CDH1), and acquisition of mesenchymal markers (VIM, CDH2). Although MAPK/ERK1/2 kinase inhibitors (MEKi) are useful antitumor agents in a clinical setting, including the Food and Drug Administration (FDA)-approved MEK1,2 dual inhibitors cobimetinib and trametinib, there are limitations to their clinical utility, primarily adaptation of the BRAF pathway and ocular toxicities. The MEK5 (HGNC: MAP2K5) pathway has important roles in metastatic progression of various cancer types, including those of the prostate, colon, bone and breast, and elevated levels of ERK5 expression in breast carcinomas are linked to a worse prognoses in TNBC patients. The purpose of this study is to explore MEK5 regulation of the EMT axis and to evaluate a novel pan-MEK inhibitor on clinically aggressive TNBC cells. Our results show a distinction between the MEK1/2 and MEK5 cascades in maintenance of the mesenchymal phenotype, suggesting that the MEK5 pathway may be necessary and sufficient in EMT regulation while MEK1/2 signaling further sustains the mesenchymal state of TNBC cells. Furthermore, additive effects on MET induction are evident through the inhibition of both MEK1/2 and MEK5. Taken together, these data demonstrate the need for a better understanding of the individual roles of MEK1/2 and MEK5 signaling in breast cancer and provide a rationale for the combined targeting of these pathways to circumvent compensatory signaling and subsequent therapeutic resistance.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Margarite D Matossian
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jacqueline La
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kristine Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Deniz A Ucar
- Department of Genetics and Stanley S. Scott Cancer Center, LSUHSC, New Orleans, Louisiana, USA
| | - Steven Elliott
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Hope E Burks
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Thomas D Wright
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Saloni Patel
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Akshita Bhatt
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Theresa Phamduy
- Department of Physics, Tulane University, New Orleans, Louisiana, USA
| | - Douglas Chrisey
- Department of Physics, Tulane University, New Orleans, Louisiana, USA
| | - Aaron Buechlein
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, Indiana, USA
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, USA
| | - Kenneth P Nephew
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, Indiana, USA
| | - Murali Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Brian Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jane E Cavanaugh
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Patrick T Flaherty
- Department of Medicinal Chemistry, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Lucio Miele
- Department of Genetics and Stanley S. Scott Cancer Center, LSUHSC, New Orleans, Louisiana, USA
| | - Bridgette M Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Tulane Cancer Center, New Orleans, Louisiana, USA
| | - Matthew E Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
39
|
Repositioning metformin and propranolol for colorectal and triple negative breast cancers treatment. Sci Rep 2021; 11:8091. [PMID: 33854147 PMCID: PMC8047046 DOI: 10.1038/s41598-021-87525-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 03/22/2021] [Indexed: 12/27/2022] Open
Abstract
Drug repositioning refers to new uses for existing drugs outside the scope of the original medical indications. This approach fastens the process of drug development allowing finding effective drugs with reduced side effects and lower costs. Colorectal cancer (CRC) is often diagnosed at advanced stages, when the probability of chemotherapy resistance is higher. Triple negative breast cancer (TNBC) is the most aggressive type of breast cancer, highly metastatic and difficult to treat. For both tumor types, available treatments are generally associated to severe side effects. In our work, we explored the effect of combining metformin and propranolol, two repositioned drugs, in both tumor types. We demonstrate that treatment affects viability, epithelial-mesenchymal transition and migratory potential of CRC cells as we described before for TNBC. We show that combined treatment affects different steps leading to metastasis in TNBC. Moreover, combined treatment is also effective preventing the development of 5-FU resistant CRC. Our data suggest that combination of metformin and propranolol could be useful as a putative adjuvant treatment for both TNBC and CRC and an alternative for chemo-resistant CRC, providing a low-cost alternative therapy without associated toxicity.
Collapse
|
40
|
Kalra M, Tong Y, Jones DR, Walsh T, Danso MA, Ma CX, Silverman P, King MC, Badve SS, Perkins SM, Miller KD. Cisplatin +/- rucaparib after preoperative chemotherapy in patients with triple-negative or BRCA mutated breast cancer. NPJ Breast Cancer 2021; 7:29. [PMID: 33753748 PMCID: PMC7985189 DOI: 10.1038/s41523-021-00240-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
Patients with triple-negative breast cancer (TNBC) who have residual disease after neoadjuvant therapy have a high risk of recurrence. We tested the impact of DNA-damaging chemotherapy alone or with PARP inhibition in this high-risk population. Patients with TNBC or deleterious BRCA mutation (TNBC/BRCAmut) who had >2 cm of invasive disease in the breast or persistent lymph node (LN) involvement after neoadjuvant therapy were assigned 1:1 to cisplatin alone or with rucaparib. Germline mutations were identified with BROCA analysis. The primary endpoint was 2-year disease-free survival (DFS) with 80% power to detect an HR 0.5. From Feb 2010 to May 2013, 128 patients were enrolled. Median tumor size at surgery was 1.9 cm (0-11.5 cm) with 1 (0-38) involved LN; median Residual Cancer Burden (RCB) score was 2.6. Six patients had known deleterious BRCA1 or BRCA2 mutations at study entry, but BROCA identified deleterious mutations in 22% of patients with available samples. Toxicity was similar in both arms. Despite frequent dose reductions (21% of patients) and delays (43.8% of patients), 73% of patients completed planned cisplatin. Rucaparib exposure was limited with median concentration 275 (82-4694) ng/mL post-infusion on day 3. The addition of rucaparib to cisplatin did not increase 2-year DFS (54.2% cisplatin vs. 64.1% cisplatin + rucaparib; P = 0.29). In the high-risk post preoperative TNBC/BRCAmut setting, the addition of low-dose rucaparib did not improve 2-year DFS or increase the toxicity of cisplatin. Genetic testing was underutilized in this high-risk population.
Collapse
Affiliation(s)
- Maitri Kalra
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Yan Tong
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David R Jones
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Tom Walsh
- University of Washington, Seattle, WA, USA
| | | | - Cynthia X Ma
- Siteman Cancer Center, Washington University, St. Louis, MO, USA
| | - Paula Silverman
- University Hospitals Ireland Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | | | - Sunil S Badve
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Susan M Perkins
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathy D Miller
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA.
| |
Collapse
|
41
|
Fleisher B, Lezeau J, Werkman C, Jacobs B, Ait-Oudhia S. In vitro to Clinical Translation of Combinatorial Effects of Doxorubicin and Abemaciclib in Rb-Positive Triple Negative Breast Cancer: A Systems-Based Pharmacokinetic/Pharmacodynamic Modeling Approach. BREAST CANCER-TARGETS AND THERAPY 2021; 13:87-105. [PMID: 33628047 PMCID: PMC7899308 DOI: 10.2147/bctt.s292161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/19/2021] [Indexed: 11/23/2022]
Abstract
Background Doxorubicin (DOX) and its pegylated liposomal formulation (L_DOX) are the standard of care for triple-negative breast cancer (TNBC). However, resistance to DOX often occurs, motivating the search for alternative treatment approaches. The retinoblastoma protein (Rb) is a potential pharmacological target for TNBC treatment since its expression has been associated with resistance to DOX-based therapy. Methods DOX (0.01–20 μM) combination with abemaciclib (ABE, 1–6 μM) was evaluated over 72 hours on Rb-positive (MDA-MB-231) and Rb-negative (MDA-MB-468) TNBC cells. Combination indices (CI) for DOX+ABE were calculated using Compusyn software. The TNBC cell viability time-course and fold-change from the control of phosphorylated-Rb (pRb) protein expression were measured with CCK8-kit and enzyme-linked immunosorbent assay. A cell-based pharmacodynamic (PD) model was developed, where pRb protein dynamics drove cell viability response. Clinical pharmacokinetic (PK) models for DOX, L_DOX, and ABE were developed using data extracted from the literature. After scaling cancer cell growth to clinical TNBC tumor growth, the time-to-tumor progression (TTP) was predicted for human dosing regimens of DOX, ABE, and DOX+ABE. Results DOX and ABE combinations were synergistic (CI<1) in MDA-MB-231 and antagonistic (CI>1) in MDA-MB-468. The maximum inhibitory effects (Imax) for both drugs were set to one. The drug concentrations producing 50% of Imax for DOX and ABE were 0.565 and 2.31 μM (MDA-MB-231) and 0.121 and 1.61 μM (MDA-MB-468). The first-orders rate constants of abemaciclib absorption (ka) and doxorubicin release from L_DOX (kRel) were estimated at 0.31 and 0.013 h−1. Their linear clearances were 21.7 (ABE) and 32.1 L/h (DOX). The estimated TTP for intravenous DOX (75 mg/m2 every 21 days), intravenous L_DOX (50 mg/m2 every 28 days), and oral ABE (200 mg twice a day) were 125, 31.2, and 8.6 days shorter than drug-free control. The TTP for DOX+ABE and L_DOX+ABE were 312 days and 47.5 days shorter than control, both larger than single-agent DOX, suggesting improved activity with the DOX+ABE combination. Conclusion The developed translational systems-based PK/PD model provides an in vitro-to-clinic modeling platform for DOX+ABE in TNBC. Although model-based simulations suggest improved outcomes with combination over monotherapy, tumor relapse was not prevented with the combination. Hence, DOX+ABE may not be an effective treatment combination for TNBC.
Collapse
Affiliation(s)
- Brett Fleisher
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Jovin Lezeau
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Carolin Werkman
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Brehanna Jacobs
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Sihem Ait-Oudhia
- Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co, Inc, Kenilworth, New Jersey, USA
| |
Collapse
|
42
|
Trailblazing perspectives on targeting breast cancer stem cells. Pharmacol Ther 2021; 223:107800. [PMID: 33421449 DOI: 10.1016/j.pharmthera.2021.107800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer (BCa) is one of the most prevalent malignant tumors affecting women's health worldwide. The recurrence and metastasis of BCa have made it a long-standing challenge to achieve remission-persistent or disease-undetectable clinical outcomes. Cancer stem cells (CSCs) possess the ability to self-renew and generate heterogeneous tumor bulk. The existence of CSCs has been found to be vital in the initiation, metastasis, therapy resistance, and recurrence of tumors across cancer types. Because CSCs grow slowly in their dormant state, they are insensitive to conventional chemotherapies; however, when CSCs emerge from their dormant state and become clinically evident, they usually acquire genetic traits that make them resistant to existing therapies. Moreover, CSCs also show evidence of acquired drug resistance in synchrony with tumor relapses. The concept of CSCs provides a new treatment strategy for BCa. In this review, we highlight the recent advances in research on breast CSCs and their association with epithelial-mesenchymal transition (EMT), circulating tumor cells (CTCs), plasticity of tumor cells, tumor microenvironment (TME), T-cell modulatory protein PD-L1, and non-coding RNAs. On the basis that CSCs are associated with multiple dysregulated biological processes, we envisage that increased understanding of disease sub-classification, selected combination of conventional treatment, molecular aberration directed therapy, immunotherapy, and CSC targeting/sensitizing strategy might improve the treatment outcome of patients with advanced BCa. We also discuss novel perspectives on new drugs and therapeutics purposing the potent and selective expunging of CSCs.
Collapse
|
43
|
Badwe R, Parmar V, Nair N. Predicting pathological complete response post neoadjuvant chemotherapy and personalizing therapy in breast cancer. CANCER RESEARCH, STATISTICS, AND TREATMENT 2021. [DOI: 10.4103/crst.crst_310_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
44
|
Mukherjee S, Adhikary S, Gadad SS, Mondal P, Sen S, Choudhari R, Singh V, Adhikari S, Mandal P, Chaudhuri S, Sengupta A, Lakshmanaswamy R, Chakrabarti P, Roy S, Das C. Suppression of poised oncogenes by ZMYND8 promotes chemo-sensitization. Cell Death Dis 2020; 11:1073. [PMID: 33323928 PMCID: PMC7738522 DOI: 10.1038/s41419-020-03129-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
Abstract
The major challenge in chemotherapy lies in the gain of therapeutic resistance properties of cancer cells. The relatively small fraction of chemo-resistant cancer cells outgrows and are responsible for tumor relapse, with acquired invasiveness and stemness. We demonstrate that zinc-finger MYND type-8 (ZMYND8), a putative chromatin reader, suppresses stemness, drug resistance, and tumor-promoting genes, which are hallmarks of cancer. Reinstating ZMYND8 suppresses chemotherapeutic drug doxorubicin-induced tumorigenic potential (at a sublethal dose) and drug resistance, thereby resetting the transcriptional program of cells to the epithelial state. The ability of ZMYND8 to chemo-sensitize doxorubicin-treated metastatic breast cancer cells by downregulating tumor-associated genes was further confirmed by transcriptome analysis. Interestingly, we observed that ZMYND8 overexpression in doxorubicin-treated cells stimulated those involved in a good prognosis in breast cancer. Consistently, sensitizing the cancer cells with ZMYND8 followed by doxorubicin treatment led to tumor regression in vivo and revert back the phenotypes associated with drug resistance and stemness. Intriguingly, ZMYND8 modulates the bivalent or poised oncogenes through its association with KDM5C and EZH2, thereby chemo-sensitizing the cells to chemotherapy for better disease-free survival. Collectively, our findings indicate that poised chromatin is instrumental for the acquisition of chemo-resistance by cancer cells and propose ZMYND8 as a suitable epigenetic tool that can re-sensitize the chemo-refractory breast carcinoma.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinogenesis/drug effects
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Movement/genetics
- Down-Regulation/drug effects
- Down-Regulation/genetics
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Epigenesis, Genetic/drug effects
- Epithelial-Mesenchymal Transition/drug effects
- Epithelial-Mesenchymal Transition/genetics
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Genome, Human
- Histone Demethylases/metabolism
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Oncogenes
- Phenotype
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Treatment Outcome
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/pathology
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Shravanti Mukherjee
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, 700032, India
| | - Shrikanth S Gadad
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, Department of Obstetrics and Gynaecology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Payel Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Sabyasachi Sen
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Ramesh Choudhari
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
- Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapura, Karnataka, 586103, India
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
- Homi Bhaba National Institute, Mumbai, India
| | - Pratiti Mandal
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, 700032, India
| | - Soumi Chaudhuri
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Amrita Sengupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX, 79905, USA
| | - Partha Chakrabarti
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, 700032, India
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata, 700032, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India.
- Homi Bhaba National Institute, Mumbai, India.
| |
Collapse
|
45
|
Zhang X, Powell K, Li L. Breast Cancer Stem Cells: Biomarkers, Identification and Isolation Methods, Regulating Mechanisms, Cellular Origin, and Beyond. Cancers (Basel) 2020; 12:E3765. [PMID: 33327542 PMCID: PMC7765014 DOI: 10.3390/cancers12123765] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Despite recent advances in diagnosis and treatment, breast cancer (BC) is still a major cause of cancer-related mortality in women. Breast cancer stem cells (BCSCs) are a small but significant subpopulation of heterogeneous breast cancer cells demonstrating strong self-renewal and proliferation properties. Accumulating evidence has proved that BCSCs are the driving force behind BC tumor initiation, progression, metastasis, drug resistance, and recurrence. As a heterogeneous disease, BC contains a full spectrum of different BC subtypes, and different subtypes of BC further exhibit distinct subtypes and proportions of BCSCs, which correspond to different treatment responses and disease-specific outcomes. This review summarized the current knowledge of BCSC biomarkers and their clinical relevance, the methods for the identification and isolation of BCSCs, and the mechanisms regulating BCSCs. We also discussed the cellular origin of BCSCs and the current advances in single-cell lineage tracing and transcriptomics and their potential in identifying the origin and lineage development of BCSCs.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH 43210, USA;
| | | | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH 43210, USA;
| |
Collapse
|
46
|
Xu X, Zhang M, Xu F, Jiang S. Wnt signaling in breast cancer: biological mechanisms, challenges and opportunities. Mol Cancer 2020; 19:165. [PMID: 33234169 PMCID: PMC7686704 DOI: 10.1186/s12943-020-01276-5] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling is a highly conserved signaling pathway that plays a critical role in controlling embryonic and organ development, as well as cancer progression. Genome-wide sequencing and gene expression profile analyses have demonstrated that Wnt signaling is involved mainly in the processes of breast cancer proliferation and metastasis. The most recent studies have indicated that Wnt signaling is also crucial in breast cancer immune microenvironment regulation, stemness maintenance, therapeutic resistance, phenotype shaping, etc. Wnt/β-Catenin, Wnt-planar cell polarity (PCP), and Wnt-Ca2+ signaling are three well-established Wnt signaling pathways that share overlapping components and play different roles in breast cancer progression. In this review, we summarize the main findings concerning the relationship between Wnt signaling and breast cancer and provide an overview of existing mechanisms, challenges, and potential opportunities for advancing the therapy and diagnosis of breast cancer.
Collapse
Affiliation(s)
- Xiufang Xu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Miaofeng Zhang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Faying Xu
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| | - Shaojie Jiang
- School of Medical Imaging, Hangzhou Medical College, Hangzhou, 310053 Zhejiang China
| |
Collapse
|
47
|
Bei Y, Cheng N, Chen T, Shu Y, Yang Y, Yang N, Zhou X, Liu B, Wei J, Liu Q, Zheng W, Zhang W, Su H, Zhu W, Ji J, Shen P. CDK5 Inhibition Abrogates TNBC Stem-Cell Property and Enhances Anti-PD-1 Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001417. [PMID: 33240752 PMCID: PMC7675186 DOI: 10.1002/advs.202001417] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/12/2020] [Indexed: 06/11/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, in which the higher frequency of cancer stem cells (CSCs) correlates with the poor clinical outcome. An aberrant activation of CDK5 is found to associate with TNBC progression closely. CDK5 mediates PPARγ phosphorylation at its Ser 273, which induces CD44 isoform switching from CD44s to CD44v, resulting in an increase of stemness of TNBC cells. Blocking CDK5/pho-PPARγ significantly reduces CD44v+ BCSCs population in tumor tissues, thus abrogating metastatic progression in TNBC mouse model. Strikingly, diminishing stemness transformation reverses immunosuppressive microenvironment and enhances anti-PD-1 therapeutic efficacy on TNBC. Mechanistically, CDK5 switches the E3 ubiquitin ligase activity of PPARγ and directly protects ESRP1 from a ubiquitin-dependent proteolysis. This finding firstly indicates that CDK5 blockade can be a potent strategy to diminish stemness transformation and increase the response to PD-1 blockade in TNBC therapy.
Collapse
Affiliation(s)
- Yuncheng Bei
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Nan Cheng
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Ting Chen
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
- Laura and Isaac Perlmutter Cancer CenterNew York University Langone Medical CenterNew YorkNYUSA
| | - Yuxin Shu
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Ye Yang
- State Key Laboratory Cultivation Base for TCM Quality and EfficacyNanjing University of Chinese MedicineNanjing210023P. R. China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Xinyu Zhou
- State Key Laboratory of Protein and Plant Gene ResearchCollege of Life SciencesPeking UniversityBeijing100871P. R. China
| | - Baorui Liu
- The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008P. R. China
| | - Jia Wei
- The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008P. R. China
| | - Qin Liu
- The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210008P. R. China
| | - Wei Zheng
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Wenlong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Huifang Su
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
| | - Wei‐Guo Zhu
- Guangdong Key Laboratory of Genome Instability and Human DiseaseShenzhen University Carson Cancer CenterDepartment of Biochemistry and Molecular BiologyShenzhen University School of MedicineShenzhen518060P. R. China
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene ResearchCollege of Life SciencesPeking UniversityBeijing100871P. R. China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer CenterNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjing210046P. R. China
- Guangdong Key Laboratory of Genome Instability and Human DiseaseShenzhen University Carson Cancer CenterDepartment of Biochemistry and Molecular BiologyShenzhen University School of MedicineShenzhen518060P. R. China
| |
Collapse
|
48
|
Muthiah I, Rajendran K, Dhanaraj P. In silico molecular docking and physicochemical property studies on effective phytochemicals targeting GPR116 for breast cancer treatment. Mol Cell Biochem 2020; 476:883-896. [PMID: 33106912 DOI: 10.1007/s11010-020-03953-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/15/2020] [Indexed: 11/28/2022]
Abstract
G protein-coupled receptor 116 (GPR116), an orphan adhesion receptor, found an important role in cell adhesion and migration in eukaryotes. Abnormal expression of GPCR identified in various cancers turns focus of research community towards GPCR to identify the targeting drug against GPCR. Though GPR116 role was studied in progression of metastasis in triple-negative breast cancer (TNBC), unfortunately, still no drugs targeting GPR116 were identified. TNBC is a hormone-negative aggressive breast cancer found even in young women. Since TNBC has no target receptor for therapy, it would be desirable to target GPR116. Currently, chemotherapy is the only promising option for TNBC; however, these drugs cause chemoresistance. Hence this current study concentrated on finding drugable natural phytochemical ligands targeting GPR116 using in silico approach. Best docked ligand with target and active binding site amino acids were identified in molecular docking study. Pharmacokinetic properties (ADME) were assessed by Qikprop. Result showed that pharmacokinetics properties of natural phytochemicals were as good as existing chemotherapeutic cancer drugs. This study indicates that phytochemicals could be a promising target for GPR116. This in silico analysis facilitates further research to design the drug targeting GPR116 for treatment of TNBC.
Collapse
Affiliation(s)
- Indiraleka Muthiah
- Department of Biotechnology, Mepco Schlenk Engineering College, Sivakasi, Tamilnadu, India
| | - Karthikeyan Rajendran
- Department of Biotechnology, Mepco Schlenk Engineering College, Sivakasi, Tamilnadu, India.
| | - Premnath Dhanaraj
- Department of Biotechnology, School of Agriculture and Biosciences, Karunya Institute of Technology and Science (Deemed To Be University), Coimbatore, Tamilnadu, 641114, India
| |
Collapse
|
49
|
Sun M, Liu X, Xia L, Chen Y, Kuang L, Gu X, Li T. A nine-lncRNA signature predicts distant relapse-free survival of HER2-negative breast cancer patients receiving taxane and anthracycline-based neoadjuvant chemotherapy. Biochem Pharmacol 2020; 189:114285. [PMID: 33069665 DOI: 10.1016/j.bcp.2020.114285] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
Multi-gene prognostic signatures of long non-coding RNAs (lncRNAs) provide new insights into mechanisms of HER2-negative breast cancer development and progression, and predict distant relapse-free survival (DRFS) of patients receiving taxane and anthracycline-based neoadjuvant chemotherapy. The aim of this study was to develop such a multi-lncRNAs signature. Optimal multiple candidate signature lncRNAs associated with DRFS were firstly identified by a univariate Cox proportional hazard regression survival analysis and a robust likelihood-based survival analysis of the GEO dataset GSE25055. A nine-lncRNA prognostic risk score model Risk Score = 0.0289 × EXPLOC100507388 - 0.0814 × EXPLINC00094 - 0.2422 × EXPSMG7-AS1 - 0.2433 × EXPPP14571 + 0.4690 × EXPASAP1-IT1 - 0.2483 × EXPLOC103344931 - 0.2464 × EXPFAM182A + 0.3349 × EXPHCG26 - 0.0216 × EXPLINC00963 was built according to the coefficients of multivariate survival analysis of the association between the candidate lncRNAs and survival. EXPlncRNA was the standardized log2-transformed expression level of the gene. According to this model, higher scores predicted lower survival probability. The area under Receiver operating characteristic (ROC) curve (AUC) was 0.777 to 0.823 from 1- to 7- year survival rate. The model and its individual lncRNAs differentiated survival probability between the higher scores (expression) and the lower scores (expression). The nine-lncRNA signature had the robust prognostic power compared with ER, PR, tumor size (T), lymph node invasion (N), TNM stage, pathologic response, chemosensitivity prediction and PAM50 signature. These results were consistent with those based on the GEO dataset GSE25065. The predictive nomograms integrating both the nine-lncRNA signature classifier and clinical-pathological risk factors were robust in predicting 1-, 3- and 5- year survival probabilities. These results supported that the nine-lncRNA signature was a robust and effective model in predicting DRFS of patients with HER2-negative breast cancer following taxane and anthracycline-based neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Min Sun
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Department of Anesthesiology, Institute of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xiaoxiao Liu
- Department of Oncology, Xinchang Hospital Affiliated to Wenzhou Medical University, 117 Gushan Middle Road, Xinchang County 312500, Zhejiang Province, China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yuying Chen
- Department of Anesthesiology, Institute of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Li Kuang
- Department of Oncology, Dongfeng General Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xinsheng Gu
- College of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, China.
| | - Tian Li
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; School of Basic Medicine, The Fourth Military Medical University, Xi'an 710000, China.
| |
Collapse
|
50
|
Sharma A, Mishra T, Thacker G, Mishra M, Narender T, Trivedi AK. Chebulinic acid inhibits MDA‐MB‐231 breast cancer metastasis and promotes cell death through down regulation of SOD1 and induction of autophagy. Cell Biol Int 2020; 44:2553-2569. [DOI: 10.1002/cbin.11463] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/17/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Akshay Sharma
- Division of Cancer Biology CSIR‐Central Drug Research Institute Lucknow Uttar Pradesh India
| | - Tripti Mishra
- Medicinal and Process Chemistry Division CSIR‐Central Drug Research Institute (CSIR‐CDRI) Lucknow Uttar Pradesh India
| | - Gatha Thacker
- Division of Cancer Biology CSIR‐Central Drug Research Institute Lucknow Uttar Pradesh India
| | - Mukul Mishra
- Division of Cancer Biology CSIR‐Central Drug Research Institute Lucknow Uttar Pradesh India
| | - Tadigoppula Narender
- Medicinal and Process Chemistry Division CSIR‐Central Drug Research Institute (CSIR‐CDRI) Lucknow Uttar Pradesh India
| | - Arun Kumar Trivedi
- Division of Cancer Biology CSIR‐Central Drug Research Institute Lucknow Uttar Pradesh India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad Uttar Pradesh India
| |
Collapse
|