1
|
Xiao S, Yu T, Yang F, Yuan H, Ni J. LMAN2 interacts with HEATR3 to expedite HER2-positive breast cancer advancement and inflammation and Akt/ERK/NF-κB signaling. Biochem Cell Biol 2025. [PMID: 39772898 DOI: 10.1139/bcb-2024-0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
The paper aimed to reveal the impacts and the possible mechanism of action of lectin mannose-binding 2 protein (LMAN2) in HER2-positive breast cancer (BC). The expression, prognostic potential of LMAN2, and the correlation between LMAN2 and HEAT repeat containing 3 (HEATR3) in BC were analyzed in TCGA database. Intact, Mentha, and BioGrid databases predicted LMAN2-HEATR3 interactions. Reverse transcription-quantitative PCR and Western blot examined LMAN2 expression. Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine staining, wound healing, and transwell assays, respectively, detected the aggressive cellular biological behaviors including proliferation, migration, and invasion. Western blot analyzed the expression of matrix metalloproteinases, HEATR3, and protein kinase B (Akt)/extracellular signal-regulated kinase (ERK)/nuclear factor-kappaB (NF-κB) signaling-related proteins. Co-immunoprecipitation assay was used to prove the relationship of LMAN2 with HEATR3. Enzyme-linked immunosorbent assay detected inflammatory cytokine levels. LMAN2 was overexpressed in HER2-positive BC tissues and cells and indicated unfavorable prognosis of BC patients. LMAN2 knockdown suppressed HER2-positive BC cell proliferation, migration, and invasion. LMAN2 interacted with and had a positive correlation with HEATR3. HEATR3 up-regulation reversed the repressive role of LMAN2 interference in the progression of HER2-positive BC, Akt/ERK/NF-κB signaling, and inflammatory response. Altogether, LMAN2 silencing might exert anti-tumor and anti-inflammatory properties and inactivate Akt/ERK/NF-κB signaling in HER2-positive BC via binding to HEATR3.
Collapse
Affiliation(s)
- Sujian Xiao
- Department of Breast Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Tong Yu
- Blood Transfusion Department, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Fulan Yang
- Department of Breast Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Huozhong Yuan
- Department of Breast Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Jun Ni
- Department of Breast Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| |
Collapse
|
2
|
Zhang Y, Tang X, Wang Y, Shi F, Gao X, Guo Y, Liu Q, Ma W. Recent advances targeting chemokines for breast cancer. Int Immunopharmacol 2024; 146:113865. [PMID: 39718056 DOI: 10.1016/j.intimp.2024.113865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024]
Abstract
Breast cancer (BC) is a complex and heterogeneous disease, and its onset and progression involve the interplay of multiple molecular mechanisms. Chemokines and their receptors are key regulators of cell migration and immune responses and contribute significantly to the pathophysiology of BC. This article reviews the classification, functions, and mechanisms of chemokines and their receptors in the proliferation, migration, invasion, and angiogenesis of BC cells. This study explores the regulatory roles of chemokines and their receptors in the immune microenvironment of BC, particularly the ways they influence the infiltration, polarization, and antitumor immune responses of immune cells. Finally, this article summarizes the current treatment strategies for breast cancer that utilize chemokines and their receptors and provides insights into future research directions and trends in this field.
Collapse
Affiliation(s)
- Yanan Zhang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Xiufeng Tang
- Department of Pharmacy and Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China.
| | - Ying Wang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Fengcui Shi
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Xing Gao
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Yingxin Guo
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Qian Liu
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Wenjian Ma
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
3
|
Ricci AD, Rizzo A, Schirizzi A, D’Alessandro R, Frega G, Brandi G, Shahini E, Cozzolongo R, Lotesoriere C, Giannelli G. Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications. Cancers (Basel) 2024; 16:3542. [PMID: 39456636 PMCID: PMC11505966 DOI: 10.3390/cancers16203542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/06/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Treatment options for intrahepatic cholangiocarcinoma (iCCA), a highly malignant tumor with poor prognosis, are limited. Recent developments in immunotherapy and immune checkpoint inhibitors (ICIs) have offered new hope for treating iCCA. However, several issues remain, including the identification of reliable biomarkers of response to ICIs and immune-based combinations. Tumor immune microenvironment (TIME) of these hepatobiliary tumors has been evaluated and is under assessment in this setting in order to boost the efficacy of ICIs and to convert these immunologically "cold" tumors to "hot" tumors. Herein, the review TIME of ICCA and its critical function in immunotherapy. Moreover, this paper also discusses potential avenues for future research, including novel targets for immunotherapy and emerging treatment plans aimed to increase the effectiveness of immunotherapy and survival rates for iCCA patients.
Collapse
Affiliation(s)
- Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Giorgio Frega
- Osteoncology, Soft Tissue and Bone Sarcomas, Innovative Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Raffaele Cozzolongo
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| |
Collapse
|
4
|
van Houtum EJ, Valk AH, Granado D, Lok J, van den Bogaard L, Remkes N, van Eck van der Sluijs J, Span PN, Cornelissen LA, Adema GJ. Siglec-7 and Siglec-9 expression in primary triple negative and oestrogen receptor positive breast cancer and in vitro signalling. Clin Transl Immunology 2024; 13:e1524. [PMID: 39246414 PMCID: PMC11378723 DOI: 10.1002/cti2.1524] [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: 05/27/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 09/10/2024] Open
Abstract
Objectives PD-1/PD-L1 immune checkpoint blockade can be an effective treatment for advanced breast cancer patients. However, patients with oestrogen receptor positive (ER+) tumors often display only low lymphocyte infiltration, while a large part of triple negative (TN) breast tumors does not generate an effective immunotherapy response. Therefore, new treatment strategies have to be developed. Here, we investigate Siglec-7 and Siglec-9 as novel ITIM-bearing inhibitory immune checkpoint receptors similar to PD-1, but expressed on a broader range of immune cells. Methods We assessed Siglec-7 and Siglec-9 (ligand) expression in TN and ER+ breast cancer tumors and their breast cancer cell line-induced signalling. Results We report that Siglec-7 and Siglec-9 are highly expressed in TN tumors, and to a low extent in ER+ tumors. Siglec-7 was observed on myeloid cells, T cells, and NK cells and Siglec-9 preferentially on myeloid cells. Expression of sialoglycans, including Siglec-7 and Siglec-9 ligands, was observed in both TN and ER+ breast cancer tissue sections. Expression levels of Siglec-7 and Siglec-9 ligands were higher on in vitro cultured TN cell lines than ER+ cell lines. Importantly, by applying chimeric Siglec-7 reporter cells, we showed the induction of Siglec-7 signalling by multiple TN cell lines, but only by one ER+ cell line. Moreover, Siglec-7 signalling is directly related to Siglec-7 ligand expression levels of breast cancer cell lines. Conclusion These data imply that immunotherapy targeting Siglec receptors may be particularly interesting for TN breast cancer patients not responding to current treatment strategies with tumors displaying high immune cell infiltration.
Collapse
Affiliation(s)
- Eline Jh van Houtum
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Anne Hc Valk
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Daniel Granado
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Jasper Lok
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Lune van den Bogaard
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Naomi Remkes
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Jesper van Eck van der Sluijs
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Paul N Span
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Lenneke Am Cornelissen
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| | - Gosse J Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
5
|
Liu H, Cai G, Yuan S, Zhou X, Gui R, Huang R. Platelet Membrane-Camouflaged Silver Metal-Organic Framework Biomimetic Nanoparticles for the Treatment of Triple-Negative Breast Cancer. Mol Pharm 2024; 21:3577-3590. [PMID: 38857525 DOI: 10.1021/acs.molpharmaceut.4c00307] [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] [Indexed: 06/12/2024]
Abstract
Triple-negative breast cancer (TNBC) is characterized by high malignancy and limited treatment options. Given the pressing need for more effective treatments for TNBC, this study aimed to develop platelet membrane (PM)-camouflaged silver metal-organic framework nanoparticles (PM@MOF-Ag NPs), a biomimetic nanodrug. PM@MOF-Ag NP construction involved the utilization of 2-methylimidazole and silver nitrate to prepare silver metal-organic framework (MOF-Ag) NPs. The PM@MOF-Ag NPs, due to their camouflage, possess excellent blood compatibility, immune escape ability, and a strong affinity for 4T1 tumor cells. This enhances their circulation time in vivo and promotes the aggregation of PM@MOF-Ag NPs at the 4T1 tumor site. Importantly, PM@MOF-Ag NPs demonstrated promising antitumor activity in vitro and in vivo. We further revealed that PM@MOF-Ag NPs induced tumor cell death by overproducing reactive oxygen species and promoting cell apoptosis. Moreover, PM@MOF-Ag NPs enhanced apoptosis by upregulating the ratios of Bax/Bcl-2 and cleaved caspase3/pro-caspase3. Notably, PM@MOF-Ag NPs exhibited no significant organ toxicity, whereas the administration of MOF-Ag NPs resulted in liver inflammation compared to the control group.
Collapse
Affiliation(s)
- Haiting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Guangqing Cai
- Department of Orthopedics, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Changsha, Hunan 410013, P. R. China
| | - Shuai Yuan
- Guangzhou Customs District Technology Center, Guangzhou 510700, China
| | - Xionghui Zhou
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Rong Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| |
Collapse
|
6
|
Cai X, Liu W, Zhang J, Li Z, Liu M, Hu S, Luo J, Peng K, Ye B, Wang Y, Yan R. Study of Iron Complex Photosensitizer with Hollow Double-Shell Nano Structure Used to Enhance Ferroptosis and Photodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309086. [PMID: 38321834 DOI: 10.1002/smll.202309086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/07/2023] [Indexed: 02/08/2024]
Abstract
Ferroptosis therapy, which uses ferroptosis inducers to produce lethal lipid peroxides and induce tumor cell death, is considered a promising cancer treatment strategy. However, challenges remain regarding how to increase the accumulation of reactive oxygen species (ROS) in the tumor microenvironment (TME) to enhance antitumor efficacy. In this study, a hyaluronic acid (HA) encapsulated hollow mesoporous manganese dioxide (H-MnO2) with double-shell nanostructure is designed to contain iron coordinated cyanine near-infrared dye IR783 (IR783-Fe) for synergistic ferroptosis photodynamic therapy against tumors. The nano photosensitizer IR783-Fe@MnO2-HA, in which HA actively targets the CD44 receptor, subsequently dissociates and releases Fe3+ and IR783 in acidic TME. First, Fe3+ consumes glutathione to produce Fe2+, which promotes the Fenton reaction in cells to produce hydroxyl free radicals (·OH) and induce ferroptosis of tumor cells. In addition, MnO2 catalyzes the production of O2 from H2O2 and enhances the production of singlet oxygen (1O2) by IR783 under laser irradiation, thus increasing the production and accumulation of ROS to provide photodynamic therapy. The highly biocompatible IR783-Fe@MnO2-HA nano-photosensitizers have exhibited tumor-targeting ability and efficient tumor inhibition in vivo due to the synergistic effect of photodynamic and ferroptosis antitumor therapies.
Collapse
Affiliation(s)
- Xinrui Cai
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Weixing Liu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiahao Zhang
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongrui Li
- Electron Microbeam Analysis Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mengkang Liu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Shuo Hu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Jun Luo
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Kai Peng
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Baofen Ye
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Yue Wang
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Ran Yan
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
7
|
Hu J, Wang J, Guo X, Fan Q, Li X, Li K, Wang Z, Liang S, Amin B, Zhang N, Chen C, Zhu B. MSLN induced EMT, cancer stem cell traits and chemotherapy resistance of pancreatic cancer cells. Heliyon 2024; 10:e29210. [PMID: 38628720 PMCID: PMC11019237 DOI: 10.1016/j.heliyon.2024.e29210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Chemoresistance is one of the main reasons for poor prognosis of pancreatic cancer. The effects of mesothelin (MSLN) on chemoresistance in pancreatic cancer are still unclear. We aim to investigate potential roles of MSLN in chemoresistance and its relationship with proliferation, epithelial-mesenchymal transition (EMT) and cancer stemness of pancreatic cancer cells. Human pancreatic cancer cell lines ASPC-1 and Mia PaCa-2 with high and low expression of MSLN, respectively, were selected. The ASPC-1 with MSLN knockout (KO) and Mia PaCa-2 of MSLN overexpression (OE) were generated. The effects of MSLN on cell phenotypes, expression of EMT-related markers, clone formation, tumor sphere formation, and pathologic role of MSLN in tumorigenesis were detected. Sensitivity of tumor cells to gemcitabine was evaluated. The results showed that adhesion, proliferation, migration and invasion were decreased significantly in ASPC-1 with MSLN KO, whereas increased significantly in Mia PaCa-2 with MSLN OE. The size and the number of clones and tumor spheres were decreased in ASPC-1 with MSLN KO, and increased in Mia PaCa-2 with MSLN OE. In xenograft model, tumor volume was decreased (tumor grew slower) in MSLN KO group compared to control group, while increased in MSLN OE group. Mia PaCa-2 with MSLN OE had a higher IC50 of gemcitabine, while ASPC-1 with MSLN KO had a lower IC50. We concluded that MSLN could induce chemoresistance by enhancing migration, invasion, EMT and cancer stem cell traits of pancreatic cancer cells. Targeting MSLN could represent a promising therapeutic strategy for reversing EMT and chemoresistance in pancreatic cancer cells.
Collapse
Affiliation(s)
- Jili Hu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Henan, 450052, China
- The First Affiliated Hospital of Zhengzhou University & Institute of Reproductive Health, Henan Academy of Innovations In Medical Science & NHC Key Laboratory of Birth Defects Prevention, China
| | - Jia Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Department of General Surgery, Third Hospital, Peking University, Beijing, 100871, China
| | - Xu Guo
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Qing Fan
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xinming Li
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Kai Li
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Zhuoyin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Shuntao Liang
- Center for Biomedical Innovation, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Buhe Amin
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Nengwei Zhang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Chaowen Chen
- Department of General Surgery, Third Hospital, Peking University, Beijing, 100871, China
| | - Bin Zhu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Department of General Surgery, Beijing Shijitan Hospital, Peking University Ninth School of Clinical Medicine, Beijing, China
| |
Collapse
|
8
|
Singh A, Mishra R, Mazumder A. Breast cancer and its therapeutic targets: A comprehensive review. Chem Biol Drug Des 2024; 103:e14384. [PMID: 37919259 DOI: 10.1111/cbdd.14384] [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: 05/20/2023] [Revised: 09/14/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Breast cancer is a common and deadly disease, so there is a constant need for research to find efficient targets and therapeutic approaches. Breast cancer can be classified on a molecular and histological base. Breast cancer can be divided into ER (estrogen receptor)-positive and ER-negative, HER2 (human epidermal growth factor receptor2)-positive and HER2-negative subtypes based on the presence of specific biomarkers. Targeting hormone receptors, such as the HER2, progesterone receptor (PR), and ER, is very significant and plays a vital role in the onset and progression of breast cancer. Endocrine treatments and HER2-targeted drugs are examples of targeted therapies now being used against these receptors. Emerging immune-based medicines with promising outcomes in the treatment of breast cancer include immune checkpoint inhibitors, cancer vaccines, and adoptive T-cell therapy. It is also explored how immune cells and the tumor microenvironment affect breast cancer development and treatment response. The major biochemical pathways, signaling cascades, and DNA repair mechanisms that are involved in the development and progression of breast cancer, include the PI3K/AKT/mTOR system, the MAPK pathway, and others. These pathways are intended to be inhibited by a variety of targeted drugs, which are then delivered with the goal of restoring normal cellular function. This review aims to shed light on types of breast cancer with the summarization of different therapeutic approaches which can target different pathways for tailored medicines and better patient outcomes.
Collapse
Affiliation(s)
- Ayushi Singh
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Rakhi Mishra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| |
Collapse
|
9
|
Maheshwari N, Sharma MC. Photoresponsive 'chemo-free' phytotherapy: formulation development for the treatment of triple-negative breast cancer. Nanomedicine (Lond) 2024; 19:5-24. [PMID: 38179960 DOI: 10.2217/nnm-2023-0231] [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] [Indexed: 01/06/2024] Open
Abstract
Aim: The present investigation aimed to develop a chemo-free, nanophytosomal system to treat triple-negative breast cancer (TNBC) via a phyto-photo dual treatment strategy. Method: Size, shape, surface analysis, photoprovoked release profile, photothermal stability, (3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide assay, apoptotic assay, DNA fragmentation, in vitro cellular uptake evaluation, mitochondrial membrane potential and caspase-3 assay, and photodynamic evaluation. Results: Biological experiments using MDA-MB-231 cells displayed dose-dependent synergistic anti-TNBC activity of PhytoS/Houttuynia cordata extract (HCE)/IR780 as compared with Phyto/HCE, PhytoS/IR780 and even more promising under laser treatment. Apoptotic assay and DNA fragmentation analysis also showed enhanced anti-TNBC effects. Investigation found that HCE acts via suppression of mitochondrial membrane potential and inducing caspase-3 activity in cells. Conclusion: Our findings suggested that photo-empowered phytotherapy can be employed effectively and safely against TNBC.
Collapse
Affiliation(s)
- Neha Maheshwari
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Indore, Madhya Pradesh, 452001, India
| | - Mukesh C Sharma
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Indore, Madhya Pradesh, 452001, India
| |
Collapse
|
10
|
Shuai Y, Ma Z, Ju J, Wei T, Gao S, Kang Y, Yang Z, Wang X, Yue J, Yuan P. Liquid-based biomarkers in breast cancer: looking beyond the blood. J Transl Med 2023; 21:809. [PMID: 37957623 PMCID: PMC10644618 DOI: 10.1186/s12967-023-04660-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
In recent decades, using circulating tumor cell (CTC), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), exosomes and etc. as liquid biomarkers has received enormous attention in various tumors, including breast cancer (BC). To date, efforts in the area of liquid biopsy predominantly focus on the analysis of blood-based markers. It is worth noting that the identifications of markers from non-blood sources provide unique advantages beyond the blood and these alternative sources may be of great significance in offering supplementary information in certain settings. Here, we outline the latest advances in the analysis of non-blood biomarkers, predominantly including urine, saliva, cerebrospinal fluid, pleural fluid, stool and etc. The unique advantages of such testings, their current limitations and the appropriate use of non-blood assays and blood assays in different settings are further discussed. Finally, we propose to highlight the challenges of these alternative assays from basic to clinical implementation and explore the areas where more investigations are warranted to elucidate its potential utility.
Collapse
Affiliation(s)
- You Shuai
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhonghua Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jie Ju
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tong Wei
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Songlin Gao
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yikun Kang
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zixuan Yang
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xue Wang
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jian Yue
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
11
|
Chen Q, Hu Q, Chen Y, Shen N, Zhang N, Li A, Li L, Li J. PRMT6 methylation of STAT3 regulates tumor metastasis in breast cancer. Cell Death Dis 2023; 14:655. [PMID: 37813837 PMCID: PMC10562413 DOI: 10.1038/s41419-023-06148-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
Overcoming distant metastasis stands as a paramount challenge in enhancing the outcomes of breast cancer treatments. Thus, delving deeper into comprehending the intricate mechanisms underlying breast cancer metastasis becomes imperative, offering potential avenues for pioneering therapeutic approaches. PRMT6, an arginine N-methyltransferase, possesses the ability to methylate both histone and non-histone proteins. It has been reported that methylation of non-histone proteins impacts their cellular localization, stability, and activation, consequently influencing tumor progression. However, the extent to which PRMT6-mediated non-histone protein methylation influences cancer cell metastasis, particularly in the context of breast cancer, remains elusive. In this study, we established that PRMT6 exerted a positive regulatory influence on breast cancer metastasis through both in vivo and in vitro experiments. Mechanistically, we innovatively revealed that PRMT6 asymmetrically di-methylated STAT3 at arginine 729 (STAT3 R729me2a). This modification proved indispensable for STAT3's membrane localization, its interaction with JAK2, STAT3 Y705 phosphorylation, and PRMT6-driven cancer cell metastasis. From a clinical perspective, we unearthed the promising potential of STAT3 R729me2a as a robust prognostic marker for predicting the overall survival time of breast cancer patients. In terms of therapeutic intervention, we demonstrated the significant capability of the PRMT6 inhibitor, EPZ020411, to curtail breast cancer metastasis both in vivo and in vitro. In sum, our study unveils the pivotal biological role of PRMT6-mediated STAT3 R729me2a in breast cancer metastasis and underscores the prospective utility of PRMT6 inhibitors as effective therapeutic strategies against STAT3-driven metastatic breast cancer.
Collapse
Affiliation(s)
- Qianzhi Chen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Chen
- Department of Hematology, Wuhan No. 1 Hospital, 430022, Wuhan, China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Shen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ning Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anshu Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Lei Li
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Junjun Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Xu H, Li L, Dong B, Lu J, Zhou K, Yin X, Sun H. TRAF6 promotes chemoresistance to paclitaxel of triple negative breast cancer via regulating PKM2-mediated glycolysis. Cancer Med 2023; 12:19807-19820. [PMID: 37746908 PMCID: PMC10587986 DOI: 10.1002/cam4.6552] [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: 04/07/2023] [Revised: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
Ample evidence reveals that glycolysis is crucial to tumor progression; however, the underlying mechanism of its drug resistance is still worth being further explored. TRAF6, an E3 ubiquitin ligase, is well recognized to overexpress in various types of cancer, which predicts a poor prognosis. In our study, we discovered that TRAF6 was expressed more significantly in the case of triple-negative breast cancer (TNBC) than in other of breast cancers, promoting chemoresistance to paclitaxel; that inhibited TRAF6 expression in the chemoresistant TNBC (TNBC-CR) cells enhanced the sensitivity by decreasing glucose uptake and lactate production; that TRAF6 regulated glycolysis and facilitated chemoresistance via binding directly to PKM2; and that overexpressing PKM2 in the TNBC-CR cells with TRAF6 knocked down regained significantly TRAF6-dependent drug resistance and glycolysis. Additionally, we verified that TRAF6 could facilitate PKM2-mediated glycolysis and chemoresistance in animal models and clinical tumor tissues. Thus, we identified the novel function of TRAF6 to promote glycolysis and drug resistance in TNBC with the regulation of PKM2, which could provide a potential molecular target for TNBC treatment.
Collapse
Affiliation(s)
- Han Xu
- Department of General SurgeryJing'an District Center Hospital of ShanghaiShanghaiChina
| | - Longzhi Li
- Department of General SurgeryJing'an District Center Hospital of ShanghaiShanghaiChina
| | - Bing Dong
- Department of General SurgeryJing'an District Center Hospital of ShanghaiShanghaiChina
| | - Ji Lu
- Department of General SurgeryJing'an District Center Hospital of ShanghaiShanghaiChina
| | - Kun Zhou
- Department of General SurgeryJing'an District Center Hospital of ShanghaiShanghaiChina
| | - Xiaoxing Yin
- Department of General SurgeryJing'an District Center Hospital of ShanghaiShanghaiChina
| | - Huizhen Sun
- Department of Obstetrics and GynecologyXinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
13
|
Zhao HQ, Jiang J. Chemokines and receptors in the development and progression of malignant tumors. Cytokine 2023; 170:156335. [PMID: 37591136 DOI: 10.1016/j.cyto.2023.156335] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/19/2023]
Abstract
Cancer cells, endothelial cells, inflammatory cells and various cytokines form a part of the tumor microenvironment (TME). Chemokines constitute the largest family of cytokines, and are mainly secreted by tumor cells and inflammatory cells in the TME. They play an important role in tumor development and progression by promoting tumor growth and metastasis, angiogenesis, and targeting the chemoattraction of inflammatory cells. Currently, some chemokine receptor antagonists are being used in clinical trials as targeted anti-tumor drugs. In this article, we review the roles of chemokines in the development and progression of malignant tumors based on recently published papers, taking into consideration of the new anti-tumor therapeutic strategies targeting chemokines and receptors.
Collapse
Affiliation(s)
- Han-Qing Zhao
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, PR China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, PR China.
| |
Collapse
|
14
|
Huang Y, Zeng A, Song L. Facts and prospects of peptide in targeted therapy and immune regulation against triple-negative breast cancer. Front Immunol 2023; 14:1255820. [PMID: 37691919 PMCID: PMC10485606 DOI: 10.3389/fimmu.2023.1255820] [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: 07/09/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. Due to the lack of specific therapeutic targets, treatment options are limited, and the recurrence and metastasis rate is high, the overall survival of patients is poor. However, with the discovery of some new targets and the corresponding immune regulation after targeting these targets, TNBC has a new hope in treatment. The peptide has a simple structure, strong binding affinity, and high stability, and has great potential in targeted therapy and immune regulation against TNBC. This review will discuss how single peptides and peptide combinations target triple-negative breast cancer to exert immunomodulatory effects. Among them, single peptides target specific receptors on TNBC cells, act as decoys to target key ligands in the regulatory pathway, and target TME-related cells. The combinations of peptides work in the form of cancer vaccines, engineered exosomes, microRNAs and other immune-related molecular pathways, immune checkpoint inhibitors, chimeric antigen receptor T cells, and drug-peptide conjugates. This article is mainly dedicated to exploring new treatment methods for TNBC to improve the curative effect and prolong the survival time of patients.
Collapse
Affiliation(s)
- Yongxiu Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Institute of Translational Pharmacology and Clinical Application, Sichuan Academy of Chinese Medical Science, Chengdu, Sichuan, China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Qayoom H, Sofi S, Mir MA. Targeting tumor microenvironment using tumor-infiltrating lymphocytes as therapeutics against tumorigenesis. Immunol Res 2023; 71:588-599. [PMID: 37004645 DOI: 10.1007/s12026-023-09376-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/25/2023] [Indexed: 04/04/2023]
Abstract
The immune system plays a vital role in suppressing tumor cell progression. The tumor microenvironment augmented with significant levels of tumor-infiltrating lymphocytes has been widely investigated and it is suggested that tumor-infiltrating lymphocytes have shown a significant role in the prognosis of cancer patients. Compared to ordinary non-infiltrating lymphocytes, tumor-infiltrating lymphocytes (TILs) are a significant population of lymphocytes that infiltrate tumor tissue and have a higher level of specific immunological reactivity against tumor cells. They serve as an effective immunological defense against various malignancies. TILs are a diverse group of immune cells that are divided into immune subsets based on the pathological and physiological impact they have on the immune system. TILs mainly consist of B-cells, T-cells, or natural killer cells with diverse phenotypic and functional properties. TILs are known to be superior to other immune cells in that they can recognize a wide range of heterogeneous tumor antigens by producing many clones of T cell receptors (TCRs), outperforming treatments like TCR-T cell and CAR-T therapy. With the introduction of genetic engineering technologies, tumor-infiltrating lymphocytes (TILs) have become a ground-breaking therapeutic option for malignancies, but because of the hindrances opposed by the immune microenvironment and the mutation of antigens, the development of TILs as therapeutic has been hindered. By giving some insight into the many variables, such as the various barriers inhibiting its usage as a potential therapeutic agent, we have examined various aspects of TILs in this work.
Collapse
Affiliation(s)
- Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India
| | - Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India.
| |
Collapse
|
16
|
Adinew GM, Messeha S, Taka E, Mochona B, Redda KK, Soliman KFA. Thymoquinone Inhibition of Chemokines in TNF-α-Induced Inflammatory and Metastatic Effects in Triple-Negative Breast Cancer Cells. Int J Mol Sci 2023; 24:9878. [PMID: 37373025 PMCID: PMC10298461 DOI: 10.3390/ijms24129878] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The lack of identifiable molecular targets or biomarkers hinders the development of treatment options in triple-negative breast cancer (TNBC). However, natural products offer a promising alternative by targeting inflammatory chemokines in the tumor microenvironment (TME). Chemokines are crucial in promoting breast cancer growth and metastasis and correlate to the altered inflammatory process. In the present study, we evaluated the anti-inflammatory and antimetastatic effects of the natural product thymoquinone (TQ) on TNF-α-stimulated TNBC cells (MDA-MB-231 and MDA-MB-468) to study the cytotoxic, antiproliferative, anticolony, antimigratory, and antichemokine effects using enzyme-linked immunosorbent assays, quantitative real-time reverse transcription-polymerase chain reactions, and Western blots were used in sequence to validate the microarray results further. Four downregulated inflammatory cytokines were identified, CCL2 and CCL20 in MDA-MB-468 cells and CCL3 and CCL4 in MDA-MB-231 cells. Furthermore, when TNF-α-stimulated MDA-MB-231 cells were compared with MDA-MB-468 cells, the two cells were sensitive to TQ's antichemokine and antimetastatic effect in preventing cell migration. It was concluded from this investigation that genetically different cell lines may respond to TQ differently, as TQ targets CCL3 and CCL4 in MDA-MB-231 cells and CCL2 and CCL20 in MDA-MB-468 cells. Therefore, the results indicate that TQ may be recommended as a component of the therapeutic strategy for TNBC treatment. These outcomes stem from the compound's capacity to suppress the chemokine. Even though these findings support the usage of TQ as part of a therapy strategy for TNBC associated with the identified chemokine dysregulations, additional in vivo studies are needed to confirm these in vitro results.
Collapse
Affiliation(s)
- Getinet M. Adinew
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (G.M.A.); (S.M.); (E.T.); (K.K.R.)
| | - Samia Messeha
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (G.M.A.); (S.M.); (E.T.); (K.K.R.)
| | - Equar Taka
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (G.M.A.); (S.M.); (E.T.); (K.K.R.)
| | - Bereket Mochona
- Department of Chemistry, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Kinfe K. Redda
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (G.M.A.); (S.M.); (E.T.); (K.K.R.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (G.M.A.); (S.M.); (E.T.); (K.K.R.)
| |
Collapse
|
17
|
Siddiqui JA, Nasser MW. Editorial: Role of chemokines in tumor heterogeneity. Semin Cancer Biol 2023; 92:128-129. [PMID: 37028577 DOI: 10.1016/j.semcancer.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
18
|
Mir MA, Bashir M, Ishfaq. Role of the CXCL8–CXCR1/2 Axis in Cancer and Inflammatory Diseases. CYTOKINE AND CHEMOKINE NETWORKS IN CANCER 2023:291-329. [DOI: 10.1007/978-981-99-4657-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Yousuf U, Sofi S, Makhdoomi A, Mir MA. Identification and analysis of dysregulated fatty acid metabolism genes in breast cancer subtypes. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:256. [PMID: 36224382 DOI: 10.1007/s12032-022-01861-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022]
Abstract
Breast cancer is one of the most aggressive and lethal types of transformation among women. An anomaly of normal fatty acid metabolism is acknowledged as a critical trigger for malignant transformations including breast cancer, but the prospect of targeting fatty acid metabolism for the treatment of malignancy has remained unrecognized so far. It has been observed that specific fatty acid metabolism genes are involved in the commencement and development of breast cancer. These specific genes have also been observed to be related to different isotypes/molecular subtypes of breast cancer. The main purpose of this study was to scrutinize the prognostic significance, functional role, and expression pattern of fatty acid metabolism genes. In-Silico tools like TCGA BrCA, Gepia2, Ualcan Analysis, UCSC Xena, Kaplan-Meier plotter, Bc-gene EXminer, String, gene ontology, and KEGG databases, were used to assess the expression pattern of the fatty acid metabolism genes in breast cancer patients and also among the different molecular sub-types of breast cancer. Differential gene expression analysis revealed dysregulation of FABP4, FABP5, PLIN1, PLIN2, PLIN4, PLIN5, LPIN1, MGLL, PNPLA2, PNPLA7, ACSL1, and ACOX2 showing a fold change > ± 1.5. Also, most of these genes show downregulation in Ualcan analysis of different isotypes/molecular subtypes of breast cancer. The study reveals that the screened genes i.e., FABP4, FABP5, PLIN1, PLIN2, PLIN4, PLIN5, LPIN1, MGLL, PNPLA2, PNPLA7, ACSL1, and ACOX2 can be used as biomarkers that reveal poor prognosis and may serve as therapeutic targets for the treatment of breast cancer.
Collapse
Affiliation(s)
- Umar Yousuf
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Aanisa Makhdoomi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India.
| |
Collapse
|
20
|
Mehraj U, Wani NA, Hamid A, Alkhanani M, Almilaibary A, Mir MA. Adapalene inhibits the growth of triple-negative breast cancer cells by S-phase arrest and potentiates the antitumor efficacy of GDC-0941. Front Pharmacol 2022; 13:958443. [PMID: 36003501 PMCID: PMC9393306 DOI: 10.3389/fphar.2022.958443] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022] Open
Abstract
Although advances in diagnostics and therapeutics have prolonged the survival of triple-negative breast cancer (TNBC) patients, metastasis, therapeutic resistance, and lack of targeted therapies remain the foremost hurdle in the effective management of TNBC. Thus, evaluation of new therapeutic agents and their efficacy in combination therapy is urgently needed. The third-generation retinoid adapalene (ADA) has potent antitumor activity, and using ADA in combination with existing therapeutic regimens may improve the effectiveness and minimize the toxicities and drug resistance. The current study aimed to assess the anticancer efficacy of adapalene as a combination regimen with the PI3K inhibitor (GDC-0941) in TNBC in vitro models. The Chou–Talalay’s method evaluated the pharmacodynamic interactions (synergism, antagonism, or additivity) of binary drug combinations. Flow cytometry, Western blotting, and in silico studies were used to analyze the mechanism of GDC–ADA synergistic interactions in TNBC cells. The combination of GDC and ADA demonstrated a synergistic effect in inhibiting proliferation, migration, and colony formation of tumor cells. Accumulation of reactive oxygen species upon co-treatment with GDC and ADA promoted apoptosis and enhanced sensitivity to GDC in TNBC cells. The findings indicate that ADA is a promising therapeutic agent in treating advanced BC tumors and enhance sensitivity to GDC in inhibiting tumor growth in TNBC models while reducing therapeutic resistance.
Collapse
Affiliation(s)
- Umar Mehraj
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, India
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Mustfa Alkhanani
- Biology Department, College of Science, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Abdullah Almilaibary
- Department of Family and Community Medicine, Albaha University, Albaha, Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, India
- *Correspondence: Manzoor Ahmad Mir,
| |
Collapse
|
21
|
Dewidar SA, Hamdy O, Eltantawy A, El-Mesery M, El Gayar AM, Soliman MM. Effect of concomitant use of pitavastatin with neoadjuvant chemotherapy protocols in breast cancer patients: A randomized controlled clinical trial. Saudi Pharm J 2022; 30:1486-1496. [PMID: 36387337 PMCID: PMC9649354 DOI: 10.1016/j.jsps.2022.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/19/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction Preclinical studies have demonstrated the possible anticancer effects of statins, but the synergistic effect of concomitant statin use with standard chemotherapy protocols in patients with breast cancer has not yet been investigated. Aim The current study aimed to evaluate the efficacy of concomitant pitavastatin use with neoadjuvant chemotherapy protocols in patients with breast cancer. Methods This study was a randomized controlled clinical trial. A total of 70 adult female patients with pathologically-proven invasive breast cancer were randomized to receive or not receive pitavastatin (2 mg) oral tablets once daily concomitantly with standard neoadjuvant chemotherapy protocols for 6 months. The primary outcomes of this study were changes in tumor size and changes to the Ki67 index. In addition, secondary outcomes were changes in cyclin D1 and cleaved caspase-3 serum levels. This study was registered at ClinicalTrials.gov (Identifier: NCT04705909). Results Patients in the pitavastatin group showed significantly higher median (IQR) reductions in tumor size [−19.8 (−41.5, 9.5)] compared to those in the control group [−5.0 (−15.5, 0.0), p = 0.0009]. The change in Ki67 from baseline to the end of therapy was similar between the two groups (p = 0.12). By the end of therapy, the cyclin D1 levels in the pitavastatin group were significantly decreased [median (IQR) change of − 10.0 (−20.2, −2.9) from baseline], whereas the control group showed an increase in cyclin D1 levels [14.8 (4.1, 56.4)]. The median (IQR) caspase−3 was elevated in the pitavastatin group 1.6 (0.2, 2.2), and decreased in the control group (−0.2 (−1.1, 0.0), p = 0.0002). Subgroup analysis of the pitavastatin group revealed that patients with positive human epidermal growth receptor 2 (HER2) had higher median (IQR) reductions in Ki67 [−35.0 (−70.0, −12.5)] than those with negative HER2 [2.5 (−15.0, 10.0), p = 0.04]. All patients who achieved a complete pathological response (n = 9) exhibited an HER2-neu positive receptor at baseline. Conclusion Concomitant use of pitavastatin with standard neoadjuvant chemotherapy protocols may improve neoadjuvant chemotherapy responses in patients with breast cancer.
Collapse
Affiliation(s)
- Samar A. Dewidar
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Omar Hamdy
- Surgical Oncology Department, Oncology Center, Mansoura University, Mansoura University, Mansoura, Egypt
| | - Ahmed Eltantawy
- Medical Oncology Unit, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Mesery
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amal M. El Gayar
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Moetaza M. Soliman
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Corresponding author at: Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
22
|
Sofi S, Mehraj U, Qayoom H, Aisha S, Almilaibary A, Alkhanani M, Mir MA. Targeting cyclin-dependent kinase 1 (CDK1) in cancer: molecular docking and dynamic simulations of potential CDK1 inhibitors. Med Oncol 2022; 39:133. [PMID: 35723742 PMCID: PMC9207877 DOI: 10.1007/s12032-022-01748-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
Cell cycle dysregulation is a characteristic hallmark of malignancies, which results in uncontrolled cell proliferation and eventual tumor formation. Cyclin-dependent kinase 1 (CDK1) is a member of the family of cell cycle regulatory proteins involved in cell cycle maintenance. Given that overexpression of CDK1 has been associated with cancer, CDK1 inhibitors may restore equilibrium to the skewed cell cycle system and operate as an effective therapeutic drug. This study aimed to identify and classify inhibitors having a higher affinity for CDK1 and also evaluate the expression pattern and prognostic relevance of CDK1 in a wide range of cancers. We investigated therapeutic molecules structurally similar to dinaciclib for their ability to inhibit CDK1 selectively. To assess the therapeutic potential of screened Dinaciclib analogs, we used drug likeliness analysis, molecular docking, and simulation analysis. CDK1 was found to be highly upregulated across several malignancies and is associated with poor overall and relapse-free survival. Molecular docking and dynamics evaluation identified two novel dinaciclib analogs as potent CDK1 inhibitors with high binding affinity and stability compared to dinaciclib. The results indicate that increased CDK1 expression is associated with decreased OS and RFS. Additionally, dinaciclib analogs are prospective replacements for dinaciclib since they exhibit increased binding affinity, consistent with MDS findings, and have acceptable ADMET qualities. The discovery of new compounds may pave the road for their future application in cancer prevention through basic, preclinical, and clinical research.
Collapse
Affiliation(s)
- Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Umar Mehraj
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Shariqa Aisha
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India
| | - Abdullah Almilaibary
- Department of Family and Community Medicine, Albaha University, Albaha, 65511, Kingdom of Saudi Arabia
| | - Mustfa Alkhanani
- Emergency Service Department, College of Applied Science, AlMaarefa University, Riyadh, 13713, Kingdom of Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, J&K, 190006, India.
| |
Collapse
|
23
|
Mehraj U, Alshehri B, Khan AA, Bhat AA, Bagga P, Wani NA, Mir MA. Expression Pattern and Prognostic Significance of Chemokines in Breast cancer: An Integrated Bioinformatics Analysis. Clin Breast Cancer 2022; 22:567-578. [DOI: 10.1016/j.clbc.2022.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 03/24/2022] [Accepted: 04/20/2022] [Indexed: 12/24/2022]
|