101
|
Christensen IB, Abrahamsen M, Ribas L, Buch‐Larsen K, Marina D, Andersson M, Larsen S, Schwarz P, Dela F, Gillberg L. Peripheral blood mononuclear cells exhibit increased mitochondrial respiration after adjuvant chemo- and radiotherapy for early breast cancer. Cancer Med 2023; 12:16985-16996. [PMID: 37439084 PMCID: PMC10501284 DOI: 10.1002/cam4.6333] [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: 04/06/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Adjuvant chemo- and radiotherapy cause cellular damage to tumorous and healthy dividing cells. Chemotherapy has been shown to cause mitochondrial respiratory dysfunction in non-tumorous tissues, but the effects on human peripheral blood mononuclear cells (PBMCs) remain unknown. AIM We aimed to investigate mitochondrial respiration of PBMCs before and after adjuvant chemo- and radiotherapy in postmenopausal patients with early breast cancer (EBC) and relate these to metabolic parameters of the patients. METHODS Twenty-three postmenopausal women diagnosed with EBC were examined before and shortly after chemotherapy with (n = 18) or without (n = 5) radiotherapy. Respiration (O2 flux per million PBMCs) was assessed by high-resolution respirometry of intact and permeabilized PBMCs. Clinical metabolic characteristics and mitochondrial DNA (mtDNA) content of PBMCs (mtDN relative to nuclear DNA) were furthermore assessed. RESULTS Respiration of intact and permeabilized PBMCs from EBC patients significantly increased with adjuvant chemo- and radiotherapy (p = 6 × 10-5 and p = 1 × 10-7 , respectively). The oxygen flux attributed to specific mitochondrial complexes and respiratory states increased by 17-43% compared to before therapy initiation. Similarly, PBMC mtDNA content increased by 40% (p = 0.002). Leukocytes (p = 0.0001), hemoglobin (p = 0.0003), and HDL cholesterol (p = 0.003) concentrations decreased whereas triglyceride (p = 0.01) and LDL (p = 0.02) concentrations increased after treatment suggesting a worsened metabolic state. None of the metabolic parameters or the mtDNA content of PBMCs correlated significantly with PBMC respiration. CONCLUSION This study shows that mitochondrial respiration and mtDNA content in circulating PBMCs increase after adjuvant chemo- and radiotherapy in postmenopausal patients with EBC. Besides the increased mtDNA content, a shift in PBMC subpopulation proportions towards cells relying on oxidative phosphorylation, who may be less sensitive to chemotherapy, might influence the increased mitochondrial respiration observed iafter chemotherapy.
Collapse
Affiliation(s)
| | | | - Lucas Ribas
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| | | | - Djordje Marina
- Department of EndocrinologyRigshospitaletCopenhagenDenmark
| | | | - Steen Larsen
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
- Clinical Research CentreMedical University of BialystokBialystokPoland
| | - Peter Schwarz
- Department of EndocrinologyRigshospitaletCopenhagenDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Flemming Dela
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of GeriatricsBispebjerg University HospitalCopenhagenDenmark
| | - Linn Gillberg
- Xlab, Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
102
|
Zheng C, Zhang D, Kong Y, Niu M, Zhao H, Song Q, Feng Q, Li X, Wang L. Dynamic regulation of drug biodistribution by turning tumors into decoys for biomimetic nanoplatform to enhance the chemotherapeutic efficacy of breast cancer with bone metastasis. EXPLORATION (BEIJING, CHINA) 2023; 3:20220124. [PMID: 37933240 PMCID: PMC10624374 DOI: 10.1002/exp.20220124] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/10/2023] [Indexed: 11/08/2023]
Abstract
Breast cancer with bone metastasis accounts for serious cancer-associated pain which significantly reduces the quality of life of affected patients and promotes cancer progression. However, effective treatment using nanomedicine remains a formidable challenge owing to poor drug delivery efficiency to multiple cancer lesions and inappropriate management of cancer-associated pain. In this study, using engineered macrophage membrane (EMM) and drugs loaded nanoparticle, we constructed a biomimetic nanoplatform (EMM@DJHAD) for the concurrent therapy of bone metastatic breast cancer and associated pain. Tumor tropism inherited from EMM provided the targeting ability for both primary and metastatic lesions. Subsequently, the synergistic combination of decitabine and JTC801 boosted the lytic and inflammatory responses accompanied by a tumoricidal effect, which transformed the tumor into an ideal decoy for EMM, resulting in prolonged troop migration toward tumors. EMM@DJHAD exerted significant effects on tumor suppression and a pronounced analgesic effect by inhibiting µ-opioid receptors in bone metastasis mouse models. Moreover, the nanoplatform significantly reduced the severe toxicity induced by chemotherapy agents. Overall, this biomimetic nanoplatform with good biocompatibility may be used for the effective treatment of breast cancer with bone metastasis.
Collapse
Affiliation(s)
- Cuixia Zheng
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
- Translational Medical Center of Huaihe HospitalHenan UniversityKaifengP. R. China
| | - Dandan Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
| | - Yueyue Kong
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
| | - Mengya Niu
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
| | - Hongjuan Zhao
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
| | - Qingling Song
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
| | - Qianhua Feng
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
- Henan Key Laboratory Targeting Therapy and Diagnosis for Critical DiseasesZhengzhouP. R. China
| | - Xingru Li
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
| | - Lei Wang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhouP. R. China
- Henan Key Laboratory Targeting Therapy and Diagnosis for Critical DiseasesZhengzhouP. R. China
- GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
- Henan International Joint Laboratory of Ovarian Malignant TumorZhengzhouP. R. China
| |
Collapse
|
103
|
Swaminathan H, Saravanamurali K, Yadav SA. Extensive review on breast cancer its etiology, progression, prognostic markers, and treatment. Med Oncol 2023; 40:238. [PMID: 37442848 DOI: 10.1007/s12032-023-02111-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
As the most frequent and vulnerable malignancy among women, breast cancer universally manifests a formidable healthcare challenge. From a biological and molecular perspective, it is a heterogenous disease and is stratified based on the etiological factors driving breast carcinogenesis. Notably, genetic predispositions and epigenetic impacts often constitute the heterogeneity of this disease. Typically, breast cancer is classified intrinsically into histological subtypes in clinical landscapes. These stratifications empower physicians to tailor precise treatments among the spectrum of breast cancer therapeutics. In this pursuit, numerous prognostic algorithms are extensively characterized, drastically changing how breast cancer is portrayed. Therefore, it is a basic requisite to comprehend the multidisciplinary rationales of breast cancer to assist the evolution of novel therapeutic strategies. This review aims at highlighting the molecular and genetic grounds of cancer additionally with therapeutic and phytotherapeutic context. Substantially, it also renders researchers with an insight into the breast cancer cell lines as a model paradigm for breast cancer research interventions.
Collapse
Affiliation(s)
- Harshini Swaminathan
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - K Saravanamurali
- Virus Research and Diagnostics Laboratory, Department of Microbiology, Coimbatore Medical College, Coimbatore, India
| | - Sangilimuthu Alagar Yadav
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| |
Collapse
|
104
|
Mehata AK, Singh V, Singh N, Mandal A, Dash D, Koch B, Muthu MS. Chitosan- g-estrone Nanoparticles of Palbociclib Vanished Hypoxic Breast Tumor after Targeted Delivery: Development and Ultrasound/Photoacoustic Imaging. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37433149 DOI: 10.1021/acsami.3c03184] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Breast cancer is the leading cause of death among women globally. Approximately 80% of all breast cancers diagnosed are overexpressed with estrogen receptors (ERs). In this study, we have developed an estrone (Egen)-grafted chitosan-based polymeric nanocarrier for the targeted delivery of palbociclib (PLB) to breast cancer. The nanoparticles (NPs) were prepared by solvent evaporation using the ionic gelation method and characterized for particle size, zeta potential, polydispersity, surface morphology, surface chemistry, drug entrapment efficiency, cytotoxicity assay, cellular uptake, and apoptosis study. The developed PLB-CS NPs and PLB-CS-g-Egen NPs had a particle size of 116.3 ± 1.53 nm and 141.6 ± 1.97 nm, respectively. The zeta potential of PLB-CS NPs and PLB-CS-g-Egen NPs was found to be 18.70 ± 0.416 mV and 12.45 ± 0.574 mV, respectively. The morphological analysis demonstrated that all NPs were spherical in shape and had a smooth surface. An in vitro cytotoxicity assay was performed in estrogen receptor (ER)-expressing MCF7 cells and T47D cells, which suggested that targeted NPs were 57.34- and 30.32-fold more cytotoxic compared to the pure PLB, respectively. Additionally, cell cycle analysis confirmed that cell cycle progression from the G1 into S phase was blocked more efficiently by targeted NPs compared to nontargeted NPs and PLB in MCF7 cells. In vivo pharmacokinetic studies demonstrated that entrapment of the PLB in the NPs improved the half-life and bioavailability by ∼2-3-fold. Further, ultrasound and photoacoustic imaging of DMBA induced breast cancer in the Sprague-Dawley (SD) rat showed that targeted NPs completely vanished breast tumor, reduced hypoxic tumor volume, and suppressed tumor angiogenesis more efficiently compared to the nontargeted NPs and free PLB. Further, in vitro hemocompatibility and histopathology studies suggested that NPs were biocompatible and safe for clinical use.
Collapse
Affiliation(s)
- Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Virendra Singh
- Cancer Biology Laboratory, Department of Zoology Institute of Science, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Nitesh Singh
- Department of Biochemistry, Institute of Medical Sciences, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Abhijit Mandal
- Department of Radiotherapy and Radiation Medicine, Institute of Medical Sciences, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Biplob Koch
- Cancer Biology Laboratory, Department of Zoology Institute of Science, (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (BHU), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
105
|
Ramezani R, Mohammadian M, Hosseini ES, Zare M. The effect of bovine milk lactoferrin-loaded exosomes (exoLF) on human MDA-MB-231 breast cancer cell line. BMC Complement Med Ther 2023; 23:228. [PMID: 37422619 DOI: 10.1186/s12906-023-04045-1] [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: 11/22/2022] [Accepted: 06/18/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Cancer is still the most challenging disease and is responsible for many deaths worldwide. Considerable research now focuses on targeted therapy in cancer using natural components to improve anti-tumor efficacy and reduce unfavorable effects. Lactoferrin is an iron-binding glycoprotein found in body fluids. Increasing evidence suggests that lactoferrin is a safe agent capable of inducing anti-cancer effects. Therefore, we conducted a study to evaluate the effects of the exosomal form of bovine milk lactoferrin on a human MDA-MB-231 breast cancer cell line. METHODS The exosomes were isolated from cancer cells by ultracentrifugation and incorporated with bovine milk lactoferrin through the incubation method. The average size of the purified exosome was determined using SEM imaging and DLS analysis. The maximum percentage of lactoferrin-loaded exosomes (exoLF) was achieved by incubating 1 mg/ml of lactoferrin with 30 µg/ml of MDA-MB-231 cells-derived exosomes. Following treatment of MDA-MB-231 cancer cells and normal cells with 1 mg/ml exoLF MTT assay applied to evaluate the cytotoxicity, PI/ annexin V analysis was carried out to illustrate the apoptotic phenotype, and the real-time PCR was performed to assess the pro-apoptotic protein, Bid, and anti-apoptotic protein, Bcl-2. RESULTS The average size of the purified exosome was about 100 nm. The maximum lactoferrin loading efficiency of exoLF was 29.72%. MTT assay showed that although the 1 mg/ml exoLF treatment of MDA-MB-231 cancer cells induced 50% cell growth inhibition, normal mesenchymal stem cells remained viable. PI/ annexin V analysis revealed that 34% of cancer cells had late apoptotic phenotype after treatment. The real-time PCR showed an elevated expression of pro-apoptotic protein Bid and diminished anti-apoptotic protein Bcl-2 following exoLF treatment. CONCLUSION These results suggested that exoLF could induce selective cytotoxicity against cancer cells compared to normal cells. Incorporating lactoferrin into the exosome seems an effective agent for cancer therapy. However, further studies are required to evaluate anti-tumor efficacy and the underlying mechanism of exoLF in various cancer cell lines and animal models.
Collapse
Affiliation(s)
- Reihaneh Ramezani
- Department of Family Therapy, Women Research Center, Alzahra University, Tehran, Iran.
| | - Mozhdeh Mohammadian
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elaheh Sadat Hosseini
- Department of Genetics, Faculty of Life Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Mehrak Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
106
|
Fatima GN, Fatma H, Saraf SK. Vaccines in Breast Cancer: Challenges and Breakthroughs. Diagnostics (Basel) 2023; 13:2175. [PMID: 37443570 DOI: 10.3390/diagnostics13132175] [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: 04/17/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer is a problem for women's health globally. Early detection techniques come in a variety of forms ranging from local to systemic and from non-invasive to invasive. The treatment of cancer has always been challenging despite the availability of a wide range of therapeutics. This is either due to the variable behaviour and heterogeneity of the proliferating cells and/or the individual's response towards the treatment applied. However, advancements in cancer biology and scientific technology have changed the course of the cancer treatment approach. This current review briefly encompasses the diagnostics, the latest and most recent breakthrough strategies and challenges, and the limitations in fighting breast cancer, emphasising the development of breast cancer vaccines. It also includes the filed/granted patents referring to the same aspects.
Collapse
Affiliation(s)
- Gul Naz Fatima
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, Uttar Pradesh, India
| | - Hera Fatma
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, Uttar Pradesh, India
| | - Shailendra K Saraf
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, Uttar Pradesh, India
| |
Collapse
|
107
|
Burguin A, Roy J, Ouellette G, Maltais R, Bherer J, Diorio C, Poirier D, Durocher F. Aminosteroid RM-581 Decreases Cell Proliferation of All Breast Cancer Molecular Subtypes, Alone and in Combination with Breast Cancer Treatments. J Clin Med 2023; 12:4241. [PMID: 37445276 DOI: 10.3390/jcm12134241] [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: 05/15/2023] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer (BC) is a heterogenous disease classified into four molecular subtypes (Luminal A, Luminal B, HER2 and triple-negative (TNBC)) depending on the expression of the estrogen receptor (ER), the progesterone receptor (PR) and the human epidermal receptor 2 (HER2). The development of effective treatments for BC, especially TNBC, remains a challenge. Aminosteroid derivative RM-581 has previously shown an antiproliferative effect in multiple cancers in vitro and in vivo. In this study, we evaluated its effect in BC cell lines representative of BC molecular subtypes, including metastatic TNBC. We found that RM-581 has an antiproliferative effect on all BC molecular subtypes, especially on Luminal A and TNBC, in 2D and 3D cultures. The combination of RM-581 and trastuzumab or trastuzumab-emtansine enhanced the anticancer effect of each drug for HER2-positive BC cell lines, and the combination of RM-581 and taxanes (docetaxel or paclitaxel) improved the antiproliferative effect of RM-581 in TNBC and metastatic TNBC cell lines. We also confirmed that RM-581 is an endoplasmic reticulum (EnR)-stress aggravator by inducing an increase in EnR-stress-induced apoptosis markers such as BIP/GRP78 and CHOP and disrupting lipid homeostasis. This study demonstrates that RM-581 could be effective for the treatment of BC, especially TNBC.
Collapse
Affiliation(s)
- Anna Burguin
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| | - Jenny Roy
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center, Québec, QC G1V 4G2, Canada
| | - Geneviève Ouellette
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| | - René Maltais
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center, Québec, QC G1V 4G2, Canada
| | - Juliette Bherer
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| | - Caroline Diorio
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
| | - Donald Poirier
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center, Québec, QC G1V 4G2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC GIV 0A6, Canada
- Cancer Research Centre, CHU de Québec-Research Centre, Québec, QC G1R 3S3, Canada
| |
Collapse
|
108
|
Zhuang H, Yu B, Tao D, Xu X, Xu Y, Wang J, Jiao Y, Wang L. The role of m6A methylation in therapy resistance in cancer. Mol Cancer 2023; 22:91. [PMID: 37264402 PMCID: PMC10233906 DOI: 10.1186/s12943-023-01782-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
Abstract
Cancer therapy resistance is the main cause of cancer treatment failure. The mechanism of therapy resistance is a hot topic in epigenetics. As one of the most common RNA modifications, N6-methyladenosine (m6A) is involved in various processes of RNA metabolism, such as stability, splicing, transcription, translation, and degradation. A large number of studies have shown that m6A RNA methylation regulates the proliferation and invasion of cancer cells, but the role of m6A in cancer therapy resistance is unclear. In this review, we summarized the research progress related to the role of m6A in regulating therapy resistance in cancers.
Collapse
Affiliation(s)
- Hengzhao Zhuang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 21500, China
| | - Bo Yu
- Department of Radiotherapy, The Affiliated Jiangyin People's Hospital of Nantong University, Jiangyin, 214400, China
| | - Dan Tao
- Department of Radiation Oncology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 21500, China
| | - Xiaoyan Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 21500, China
| | - Yijun Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 21500, China
| | - Jian Wang
- Department of Radiotherapy, The Affiliated Jiangyin People's Hospital of Nantong University, Jiangyin, 214400, China.
| | - Yang Jiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, 215000, China.
| | - Lili Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, 21500, China.
| |
Collapse
|
109
|
Ramos E, Egea J, López-Muñoz F, Gil-Martín E, Romero A. Therapeutic Potential of Melatonin Counteracting Chemotherapy-Induced Toxicity in Breast Cancer Patients: A Systematic Review. Pharmaceutics 2023; 15:1616. [PMID: 37376065 DOI: 10.3390/pharmaceutics15061616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/05/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
The purpose of this systematic review is to provide an overview of the existing knowledge on the therapeutic potential of melatonin to counteract the undesirable effects of chemotherapy in breast cancer patients. To this aim, we summarized and critically reviewed preclinical- and clinical-related evidence according to the PRISMA guidelines. Additionally, we developed an extrapolation of melatonin doses in animal studies to the human equivalent doses (HEDs) for randomized clinical trials (RCTs) with breast cancer patients. For the revision, 341 primary records were screened, which were reduced to 8 selected RCTs that met the inclusion criteria. We assembled the evidence drawn from these studies by analyzing the remaining gaps and treatment efficacy and suggested future translational research and clinical trials. Overall, the selected RCTs allow us to conclude that melatonin combined with standard chemotherapy lines would derive, at least, a better quality of life for breast cancer patients. Moreover, regular doses of 20 mg/day seemed to increase partial response and 1-year survival rates. Accordingly, this systematic review leads us to draw attention to the need for more RCTs to provide a comprehensive view of the promising actions of melatonin in breast cancer and, given the safety profile of this molecule, adequate translational doses should be established in further RCTs.
Collapse
Affiliation(s)
- Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Institute Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Francisco López-Muñoz
- Faculty of Health, Camilo José Cela University of Madrid (UCJC), 28692 Madrid, Spain
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute, 28041 Madrid, Spain
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, 36310 Vigo, Spain
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
110
|
Tuğrul B, Balcan E, Öztel Z, Çöllü F, Gürcü B. Prion protein-dependent regulation of p53-MDM2 crosstalk during endoplasmic reticulum stress and doxorubicin treatments might be essential for cell fate in human breast cancer cell line, MCF-7. Exp Cell Res 2023:113656. [PMID: 37245583 DOI: 10.1016/j.yexcr.2023.113656] [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: 03/01/2023] [Revised: 05/09/2023] [Accepted: 05/21/2023] [Indexed: 05/30/2023]
Abstract
In this study, we investigated the effect of doxorubicin and tunicamycin treatment alone or in combination on MDM-, Cul9-and prion protein (PrP)-mediated subcellular regulation of p53 in the context of apoptosis and autophagy. MTT analysis was performed to determine the cytotoxic effect of the agents. Apoptosis was monitorized by ELISA, flow cytometry and JC-1 assay. Monodansylcadaverine assay was performed for autophagy. Western blotting and immunofluorescence were performed to determine p53, MDM2, CUL9 and PrP levels. Doxorubicin increased p53, MDM2 and CUL9 levels in a dose-dependent manner. Expression of p53 and MDM2 was higher at the 0.25 μM concentration of tunicamycin compared to the control, but it decreased at 0.5 μM and 1 μM concentrations. CUL9 expression was significantly decreased only after treatment of tunicamycin at 0.25 μM. According to its glycosylation status, the upper band of PrP increased only in combination treatment. In combination treatment, p53 expression was higher than control, whereas MDM2 and CUL9 expressions were decreased. Combination treatments may make MCF-7 cells more susceptible to apoptosis rather than autophagy. In conclusion, PrP may be important in determining the fate of cell death through crosstalk between proteins such as p53 and MDM2 under endoplasmic reticulum (ER) stress conditions. Further studies are needed to obtain in-depth information on these potential molecular networks.
Collapse
Affiliation(s)
- Berrin Tuğrul
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Molecular Biology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Erdal Balcan
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Molecular Biology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Zübeyde Öztel
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Molecular Biology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Fatih Çöllü
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Zoology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Beyhan Gürcü
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Zoology Section, 45140, Yunusemre, Manisa, Turkey.
| |
Collapse
|
111
|
Zahari S, Syafruddin SE, Mohtar MA. Impact of the Cancer Cell Secretome in Driving Breast Cancer Progression. Cancers (Basel) 2023; 15:2653. [PMID: 37174117 PMCID: PMC10177134 DOI: 10.3390/cancers15092653] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Breast cancer is a complex and heterogeneous disease resulting from the accumulation of genetic and epigenetic alterations in breast epithelial cells. Despite remarkable progress in diagnosis and treatment, breast cancer continues to be the most prevalent cancer affecting women worldwide. Recent research has uncovered a compelling link between breast cancer onset and the extracellular environment enveloping tumor cells. The complex network of proteins secreted by cancer cells and other cellular components within the tumor microenvironment has emerged as a critical player in driving the disease's metastatic properties. Specifically, the proteins released by the tumor cells termed the secretome, can significantly influence the progression and metastasis of breast cancer. The breast cancer cell secretome promotes tumorigenesis through its ability to modulate growth-associated signaling pathways, reshaping the tumor microenvironment, supporting pre-metastatic niche formation, and facilitating immunosurveillance evasion. Additionally, the secretome has been shown to play a crucial role in drug resistance development, making it an attractive target for cancer therapy. Understanding the intricate role of the cancer cell secretome in breast cancer progression will provide new insights into the underlying mechanisms of this disease and aid in the development of more innovative therapeutic interventions. Hence, this review provides a nuanced analysis of the impact of the cancer cell secretome on breast cancer progression, elucidates the complex reciprocal interaction with the components of the tumor microenvironment and highlights emerging therapeutic opportunities for targeting the constituents of the secretome.
Collapse
Affiliation(s)
| | | | - M. Aiman Mohtar
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (S.Z.); (S.E.S.)
| |
Collapse
|
112
|
Li J, Yuan Y, Bian L, Lin Q, Yang H, Ma L, Xin L, Li F, Zhang S, Wang T, Liu Y, Jiang Z. A comparison between clinical decision support system and clinicians in breast cancer. Heliyon 2023; 9:e16059. [PMID: 37215843 PMCID: PMC10192819 DOI: 10.1016/j.heliyon.2023.e16059] [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: 03/30/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Objective We are building a clinical decision support system (CSCO AI) for breast cancer patients to improve the efficiency of clinical decision-making. We aimed to assess cancer treatment regimens given by CSCO AI and different levels of clinicians. Methods 400 breast cancer patients were screened from the CSCO database. Clinicians with similar levels were randomly assigned one of the volumes (200 cases). CSCO AI was asked to assess all cases. Three reviewers were independently asked to evaluate the regimens from clinicians and CSCO AI. Regimens were masked before evaluation. The primary outcome was the proportion of high-level conformity (HLC). Results The overall concordance between clinicians and CSCO AI was 73.9% (3621/4900). It was 78.8% (2757/3500) in the early-stage, higher than that in the metastatic stage (61.7% [864/1400], p < 0.001). The concordance was 90.7% (635/700) and 56.4% (395/700) in adjuvant radiotherapy and second-line therapy respectively. HLC in CSCO AI was 95.8% (95%CI:94.0%-97.6%), significantly higher than that in clinicians (90.8%, 95%CI:89.8%-91.8%). Considering professions, the HLC of surgeons was 85.9%, lower than that of CSCO AI (OR = 0.25,95%CI: 0.16-0.41). The most significant difference in HLC was in first-line therapy (OR = 0.06, 95%CI:0.01-0.41). When clinicians were divided according to their levels, there was no statistical significance between CSCO AI and higher level clinicians. Conclusions Decision from CSCO AI for breast cancer was superior than most clinicians did except in second-line therapy. The improvements in process outcomes suggest that CSCO AI can be widely used in clinical practice.
Collapse
Affiliation(s)
- Jianbin Li
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing
| | - Yang Yuan
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Li Bian
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Qiang Lin
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Hua Yang
- Department of Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Li Ma
- Department of General Surgery, Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ling Xin
- Department of Breast Surgery, Peking University First Hospital, Beijing, China
| | - Feng Li
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Shaohua Zhang
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Tao Wang
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yinhua Liu
- Department of Breast Surgery, Peking University First Hospital, Beijing, China
| | - Zefei Jiang
- Department of Oncology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
113
|
Qiu Y, Wang X, Sun Y, Du YE, Yin G, Luo H, Wen S, Lang L, Liu M, Tang X. TCF12 regulates exosome release from epirubicin-treated CAFs to promote ER+ breast cancer cell chemoresistance. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166727. [PMID: 37137433 DOI: 10.1016/j.bbadis.2023.166727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are the predominant stromal cells in the microenvironment and play important roles in tumor progression, including chemoresistance. However, the response of CAFs to chemotherapeutics and their effects on chemotherapeutic outcomes are largely unknown. In this study, we showed that epirubicin (EPI) treatment triggered ROS which initiated autophagy in CAFs, TCF12 inhibited autophagy flux and further promoted exosome secretion. Inhibition of EPI-induced reactive oxygen species (ROS) production with N-acetyl-L-cysteine (NAC) or suppression of autophagic initiation with short interfering RNA (siRNA) against ATG5 blunted exosome release from CAFs. Furthermore, exosome secreted from EPI-treated CAFs not only prevented ROS accumulation in CAFs but also upregulated the CXCR4 and c-Myc protein levels in recipient ER+ breast cancer cells, thus promoting EPI resistance of tumor cells. Together, the current study provides novel insights into the role of stressed CAFs in promoting tumor chemoresistance and reveal a new function of TCF12 in regulating autophagy impairment and exosome release.
Collapse
Affiliation(s)
- Yuxiang Qiu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xing Wang
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Yan-E Du
- Department of Laboratory Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siyang Wen
- Department of Laboratory Medicine, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Lei Lang
- Department of Clinical Laboratory, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing 400014, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Xi Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
114
|
Wang Y, Wang S, Wang Q, Tang W, Lin L, Zhang T, Hu M, Wang X. Identification of a luminescent platinum(II) complex with BODIPY derivative as novel photodynamic therapy agent for triple negative breast cancer cells. J Inorg Biochem 2023; 242:112160. [PMID: 36791603 DOI: 10.1016/j.jinorgbio.2023.112160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Triple-negative breast cancer (TNBC) is one of the most malignant breast tumors for its poor prognosis and high tumor recurrence. It is urgent to develop new strategy or effective agents to overcome resistance and improve therapeutic effectiveness. Boron-dipyrromethene (BODIPY) based photosensitizers possess exciting photophysical features suitable for theranostic applications, namely, photodynamic therapy (PDT). We have designed a luminescent monofunctional platinum(II) complex with BODIPY derivative, namely I2BC-Pt, as novel high PDT agent against triple negative breast cancer (TNBC). The di-iodinated BODIPY complex I2BC-Pt showed excellent PDT effect against TNBC cells in green light (520 nm) giving IC50 values of 0.11-0.13 μM in MDA-MB-231 and MDA-MB-468 cells. I2BC-Pt predominately aggregated in the mitochondria of MDA-MB-231 cells and emitted green fluorescence. Besides, the anticancer mechanism studies demonstrated that I2BC-Pt could help DNA repair through attenuating RAD51, FoxM1 and BRCA1/2, and induce p53-mediated apoptosis of TNBC cells. Taken together, I2BC-Pt could be potentially developed as a BODIPY-based photosensitizers for TNBC therapy.
Collapse
Affiliation(s)
- Yujing Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Shuping Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, PR China
| | - Qingqing Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Wanyu Tang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Tao Zhang
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Xiaobo Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, PR China.
| |
Collapse
|
115
|
Jiang T, Zhu J, Jiang S, Chen Z, Xu P, Gong R, Zhong C, Cheng Y, Sun X, Yi W, Yang J, Zhou W, Cheng Y. Targeting lncRNA DDIT4-AS1 Sensitizes Triple Negative Breast Cancer to Chemotherapy via Suppressing of Autophagy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2207257. [PMID: 37096846 DOI: 10.1002/advs.202207257] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/06/2023] [Indexed: 05/03/2023]
Abstract
In this study, it is found that the lncRNA, DNA damage inducible transcript 4 antisense RNA1 (DDIT4-AS1), is highly expressed in triple-negative breast cancer (TNBC) cell lines and tissues due to H3K27 acetylation in the promoter region, and promotes the proliferation, migration, and invasion of TNBC cells via activating autophagy. Mechanistically, it is shown that DDIT4-AS1 induces autophagy by stabilizing DDIT4 mRNA via recruiting the RNA binding protein AUF1 and promoting the interaction between DDIT4 mRNA and AUF1, thereby inhibiting mTOR signaling pathway. Furthermore, silencing of DDIT4-AS1 enhances the sensitivity of TNBC cells to chemotherapeutic agents such as paclitaxel both in vitro and in vivo. Using a self-activatable siRNA/drug core-shell nanoparticle system, which effectively deliver both DDIT4-AS1 siRNA and paclitaxel to the tumor-bearing mice, a significantly enhanced antitumor activity is achieved. Importantly, the codelivery nanoparticles exert a stronger antitumor effect on breast cancer patient-derived organoids. These findings indicate that lncRNA DDIT4-AS1-mediated activation of autophagy promotes progression and chemoresistance of TNBC, and targeting of DDIT4-AS1 may be exploited as a new therapeutic approach to enhancing the efficacy of chemotherapy against TNBC.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaojiao Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Shilong Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zonglin Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ping Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Changxin Zhong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yueying Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xinyuan Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Jinming Yang
- Department of Cancer Biology and Toxicology, Department of Pharmacology, College of Medicine and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China
| |
Collapse
|
116
|
Liu Y, Hong G, Mao L, Su Z, Liu T, Liu H. A Novel Paclitaxel Derivative for Triple-Negative Breast Cancer Chemotherapy. Molecules 2023; 28:molecules28093662. [PMID: 37175072 PMCID: PMC10180349 DOI: 10.3390/molecules28093662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
Paclitaxel-triethylenetetramine hexaacetic acid conjugate (PTX-TTHA), a novel semi-synthetic taxane, is designed to improve the water solubility and cosolvent toxicity of paclitaxel in several aminopolycarboxylic acid groups. In this study, the in vitro and in vivo antitumor effects and mechanisms of PTX-TTHA against triple-negative breast cancer (TNBC) and its intravenous toxicity were evaluated. Results showed the water solubility of PTX-TTHA was greater than 5 mg/mL, which was about 7140-fold higher than that of paclitaxel (<0.7 µg/mL). PTX-TTHA (10-105 nmol/L) could significantly inhibit breast cancer proliferation and induce apoptosis by stabilizing microtubules and arresting the cell cycle in the G2/M phase in vitro, with its therapeutic effect and mechanism similar to paclitaxel. However, when the MDA-MB-231 cell-derived xenograft (CDX) tumor model received PTX-TTHA (13.73 mg/kg) treatment once every 3 days for 21 days, the tumor inhibition rate was up to 77.32%. Furthermore, PTX-TTHA could inhibit tumor proliferation by downregulating Ki-67, and induce apoptosis by increasing pro-apoptotic proteins (Bax, cleaved caspase-3) and TdT-mediated dUTP nick end labeling (TUNEL) positive apoptotic cells, and reducing anti-apoptotic protein (Bcl-2). Moreover, PTX-TTHA demonstrated no sign of acute toxicity on vital organs, hematological, and biochemical parameters at the limit dose (138.6 mg/kg, i.v.). Our study indicated that PTX-TTHA showed better water solubility than paclitaxel, as well as comparable in vitro and in vivo antitumor activity in TNBC models. In addition, the antitumor mechanism of PTX-TTHA was related to microtubule regulation and apoptosis signaling pathway activation.
Collapse
Affiliation(s)
- Yuetong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ge Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Lina Mao
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Zhe Su
- Tianjin Institute for Drug Control, Tianjin 300070, China
| | - Tianjun Liu
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
117
|
Lv W, He X, Wang Y, Zhao C, Dong M, Wu Y, Zhang Q. A novel immune score model predicting the prognosis and immunotherapy response of breast cancer. Sci Rep 2023; 13:6403. [PMID: 37076508 PMCID: PMC10115816 DOI: 10.1038/s41598-023-31153-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/07/2023] [Indexed: 04/21/2023] Open
Abstract
Breast cancer (BC) is one of the most common malignancies. However, the existing pathological grading system cannot accurately and effectively predict the survival rate and immune checkpoint treatment response of BC patients. In this study, based on The Cancer Genome Atlas (TCGA) database, a total of 7 immune-related genes (IRGs) were screened out to construct a prognostic model. Subsequently, the clinical prognosis, pathological characteristics, cancer-immunity cycle, tumour immune dysfunction and exclusion (TIDE) score, and immune checkpoint inhibitor (ICI) response were compared between the high- and low-risk groups. In addition, we determined the potential regulatory effect of NPR3 on BC cell proliferation, migration, and apoptosis. The model consisting of 7 IRGs was an independent prognostic factor. Patients with lower risk scores exhibited longer survival times. Moreover, the expression of NPR3 was increased but the expression of PD-1, PD-L1, and CTLA-4 was decreased in the high-risk group compared to the low-risk group. In addition, compared with si-NC, si-NPR3 suppressed proliferation and migration but promoted apoptosis in both MDA-MB-231 and MCF-7 cells. This study presents a model for predicting survival outcomes and provides a strategy to guide effective personalized immunotherapy in BC patients.
Collapse
Affiliation(s)
- Wenchang Lv
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Xiao He
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yichen Wang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Chongru Zhao
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Menglu Dong
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
118
|
Mao J, Lu Q, Li P, Shi S, Li J, Li Y, Chen S, Xie X. CCDC3 Gene Regulates the Proliferation of Breast Cancer Cells. Bull Exp Biol Med 2023; 174:653-658. [PMID: 37052857 DOI: 10.1007/s10517-023-05763-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Indexed: 04/14/2023]
Abstract
We studied the effect of CCDC3 on the viability of human breast cancer cell line MDA-MB-231. The levels of CCDC3 mRNA and the corresponding protein in MDA-MB-231, MCF-7, T-47D, and HCC1937 cell lines were measured by reverse transcription quantitative real-time PCR and Western blotting. Since MDA-MB-231 cells had higher expression of mRNA CCDC3 and CCDC3 protein, we used this cell line for transfection with small interfering RNA by lentivirus. Cell Counting Kit-8 and clone formation assay were used to detect the effects of CCDC3 knockdown on cell viability; flow cytometry was used to detect the effects of CCDC3 knockdown on cell apoptosis and cell cycle. In MDA-MB-231 cell line, the CCDC3 protein level was significantly down-regulated after CCDC3 knockdown in comparison with the control group (p<0.05). The cell viability and the number of clones in the CCDC3 knockdown group were significantly reduced (p<0.05), while the apoptosis rate significantly increased (p<0.05). Thus, after CCDC3 knockdown, cell viability is weakened in MDA-MB-231 cells, and cell apoptosis rate is increased. Therefore, CCDC3 gene is promising as a new candidate target for BC treatment.
Collapse
Affiliation(s)
- J Mao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Q Lu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - P Li
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - S Shi
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - J Li
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Y Li
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - S Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - X Xie
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
119
|
El-Far M, Essam A, El-Senduny FF, El-Azim AO, Yahia S, El-Sherbiny IM. Novel highly effective combination of naturally-derived quercetin and ascorbyl palmitate and their nanoformulations as an advancement therapy of cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
120
|
LPCAT1 is transcriptionally regulated by FOXA1 to promote breast cancer progression and paclitaxel resistance. Oncol Lett 2023; 25:134. [PMID: 36909375 PMCID: PMC9996177 DOI: 10.3892/ol.2023.13720] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/09/2023] [Indexed: 02/17/2023] Open
Abstract
Bioinformatics analysis indicates that lysophosphatidylcholine acyltransferase 1 (LPCAT1) and forkhead box A1 (FOXA1) are highly expressed in breast cancer tissues and their expression levels are correlated. Therefore, the aim of the present study was to investigate their involvement in the malignant progression and drug resistance of breast cancer. The clinical significance of LPCAT1 was analyzed using The Cancer Genome Atlas data. The enrichment of LPCAT1 in breast cancer cells was determined and the effects of LPCAT1 knockdown on cell proliferation, colony formation, migration, invasion and paclitaxel (PTX) resistance were evaluated. The association between LPCAT1 and FOXA1 was verified using luciferase reporter and chromatin immunoprecipitation assays. Thereafter, the ability of FOXA1 overexpression to regulate LPCAT1 regulation was evaluated. The results revealed that a high LPCAT1 level was associated with poor overall survival in patients with breast cancer. Furthermore, LPCAT1 was found to be highly expressed in breast cancer cells, and its knockdown resulted in suppressed proliferation, colony formation, migration and invasion, and weakened PTX resistance. Furthermore, FOXA1 overexpression attenuated the effects of LPCAT1 knockdown on cells, indicating that FOXA1 transcriptionally regulates LPCAT1. In summary, the present study reveals that LPCAT1 is transcriptionally regulated by FOXA1, which influences breast cancer cell proliferation, metastatic potential and PTX resistance.
Collapse
|
121
|
Afzal M, Alarifi A, Karami AM, Ayub R, Abduh NAY, Saeed WS, Muddassir M. Antiproliferative Mechanisms of a Polyphenolic Combination of Kaempferol and Fisetin in Triple-Negative Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24076393. [PMID: 37047366 PMCID: PMC10094218 DOI: 10.3390/ijms24076393] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/16/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Herein, we investigate the combinatorial therapeutic effects of naturally occurring flavonoids kaempferol (K) and fisetin (F) on triple-negative breast cancer (TNBC: MDA-MB-231 cell line). Dose-dependent MTT assay results show that K and F exhibited cytotoxicity in MDA-MB-231 cells at 62 and 75 μM (IC50), respectively, after 24 h. However, combined K + F led to 40% and more than 50% TNBC cell death observed at 10 and 20 μM, respectively, which revealed the synergistic association of both. The combination of K and F was determined to be more effective in inhibiting cell viability than either of the agents alone. The morphological changes associated with significant apoptotic cell death were observed under a fluorescent microscope, strongly supporting the synergistic association between K and F. We also proposed that combining the effects of both polyphenols, as opposed to their individual effects, would increase their in vitro efficacy. Furthermore, we assessed the cell death pathway by the combinational treatment via reactive oxygen species-induced DNA damage and the mitochondrially mediated apoptotic pathway. This study reveals the prominent synergistic role of phytochemicals, which helps in elevating the therapeutic efficacy of dietary nutrients and that anticancer effects may be a result of nutrients that act in concert.
Collapse
Affiliation(s)
- Mohd. Afzal
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence:
| | - Abdullah Alarifi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Rashid Ayub
- Department of Science Technology Unit, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naaser A. Y. Abduh
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Waseem Sharaf Saeed
- Restorative Dental Sciences Department, College of Dentistry, King Saud University, Riyadh 11545, Saudi Arabia
| | - Mohd. Muddassir
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
122
|
Sánchez-León ML, Jiménez-Cortegana C, Silva Romeiro S, Garnacho C, de la Cruz-Merino L, García-Domínguez DJ, Hontecillas-Prieto L, Sánchez-Margalet V. Defining the Emergence of New Immunotherapy Approaches in Breast Cancer: Role of Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:5208. [PMID: 36982282 PMCID: PMC10048951 DOI: 10.3390/ijms24065208] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Breast cancer (BC) continues to be the most diagnosed tumor in women and a very heterogeneous disease both inter- and intratumoral, mainly given by the variety of molecular profiles with different biological and clinical characteristics. Despite the advancements in early detection and therapeutic strategies, the survival rate is low in patients who develop metastatic disease. Therefore, it is mandatory to explore new approaches to achieve better responses. In this regard, immunotherapy arose as a promising alternative to conventional treatments due to its ability to modulate the immune system, which may play a dual role in this disease since the relationship between the immune system and BC cells depends on several factors: the tumor histology and size, as well as the involvement of lymph nodes, immune cells, and molecules that are part of the tumor microenvironment. Particularly, myeloid-derived suppressor cell (MDSC) expansion is one of the major immunosuppressive mechanisms used by breast tumors since it has been associated with worse clinical stage, metastatic burden, and poor efficacy of immunotherapies. This review focuses on the new immunotherapies in BC in the last five years. Additionally, the role of MDSC as a therapeutic target in breast cancer will be described.
Collapse
Affiliation(s)
- María Luisa Sánchez-León
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carlos Jiménez-Cortegana
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Silvia Silva Romeiro
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Víctor Sánchez-Margalet
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| |
Collapse
|
123
|
Jin Y, Qiu L, Bao W, Lu M, Cao F, Ni H, Zhao B. High expression of IGHG1 promotes breast cancer malignant development by activating the AKT pathway. Cell Cycle 2023; 22:718-731. [PMID: 36404682 PMCID: PMC9980652 DOI: 10.1080/15384101.2022.2147141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/17/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022] Open
Abstract
This study researched the exact function of IgG1 heavy chain (IGHG1) on breast cancer (BC) progression. IGHG1 level within BC and paired normal tissues was acquired in Gene Expression Profiling Interactive Analysis dataset. Meanwhile, this work harvested tumor and paired healthy tissues in 42 BC cases. siRNA targeting IGHG1 was transfected into BC cells. SC79 was used to treat the transfected BC cells. CCK-8 assay, clone formation experiment, BrdU assay, Transwell experiment and flow cytometry were carried out to measure the viability, colony formation, proliferation, invasion, and apoptosis of BC cells. Paclitaxel and cisplatin sensitivity of BC cells was evaluated by MTT assay. Real-time quantitative reverse transcription-polymerase chain reaction and Western-blot were performed for measuring mRNA and protein expression. The overexpressed IGHG1 indicated dismal BC survival. IGHG1 silencing attenuated the viability, invasion, proliferation, epithelial-mesenchymal transition, but enhanced the apoptosis of BC cells. IGHG1 silencing enhanced the paclitaxel and cisplatin sensitivity of BC cells. IGHG1 silencing suppressed the activity of the MEK, AKT, and ERK pathways. AKT agonist partially reversed the inhibition of IGHG1 silencing on BC cell malignant phenotype and resistance to paclitaxel and cisplatin. IGHG1 promotes the malignant development of BC by activating the AKT pathway. It may be an effective target for BC treatment.
Collapse
Affiliation(s)
- Yongmei Jin
- Department of Nursing, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lin Qiu
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenqing Bao
- School of Medicine, Tongji University, Shanghai, China
| | - Minhao Lu
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Cao
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanchen Ni
- Department of Nursing, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bin Zhao
- Department of General Surgery, The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
124
|
Singh N, Reddy KP, Das P, Kishor BK, Datta P. Complex formulation strategies to overcome the delivery hurdles of laptinib in metastatic breast cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
125
|
Role of hypoxia-inducible factor-1α and survivin in enhancing radiosensitivity of breast cancer. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2023. [DOI: 10.1016/j.jrras.2023.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
126
|
Fan R, Tao X, Zhai X, Zhu Y, Li Y, Chen Y, Dong D, Yang S, Lv L. Application of aptamer-drug delivery system in the therapy of breast cancer. Biomed Pharmacother 2023; 161:114444. [PMID: 36857912 DOI: 10.1016/j.biopha.2023.114444] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Despite significant treatment advances, breast cancer remains the leading cause of cancer death in women. From the current treatment situation, in addition to developing chemoresistant tumours, distant organ metastasis, and recurrences, patients with breast cancer often have a poor prognosis. Aptamers as "chemical antibodies" may be a way to resolve this dilemma. Aptamers are single-stranded, non-coding oligonucleotides (DNA or RNA), resulting their many advantages, including stability for long-term storage, simplicity of synthesis and function, and low immunogenicity, a high degree of specificity and antidote. Aptamers have gained popularity as a method for diagnosing and treating specific tumors in recent years. This article introduces the application of ten different aptamer delivery systems in the treatment and diagnosis of breast cancer, and systematically reviews their latest research progress in breast cancer treatment and diagnosis. It provides a new direction for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Rui Fan
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunming Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanwei Chen
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
127
|
Liang Z, Liu L, Zhou Y, Liu W, Lu Y. Research Progress on Bioactive Metal Complexes against ER-Positive Advanced Breast Cancer. J Med Chem 2023; 66:2235-2256. [PMID: 36780448 DOI: 10.1021/acs.jmedchem.2c01458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Breast cancer is the most prevalent cancer in women and represents a serious disease that is harmful to life and health. In 1977, with the approval of tamoxifen, endocrine therapy has become the main clinical treatment for ER-positive (ER+) breast cancer. Although patients initially respond well to endocrine therapies, drug resistance often emerges and side effects can be challenging. To overcome drug resistance, the exploration for new drugs is a priority. Metal complexes have demonstrated significant antitumor activities, and platinum complexes are widely used in the clinic against various cancers, including breast cancer. In this Perspective, the first section describes the classification and mechanism of endocrine therapy drugs for ER+ breast cancer, and the second section summarizes research since 2000 into metal complexes with activity toward ER+ breast cancer. Finally, we discuss the opportunities, challenges, and future directions for metal complexes in the treatment of ER+ breast cancer.
Collapse
Affiliation(s)
- Zhenlin Liang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Lijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Yanyu Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China.,State key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210023, P. R. China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| |
Collapse
|
128
|
Qian Z, Chen L, Liu J, Jiang Y, Zhang Y. The emerging role of PPAR-alpha in breast cancer. Biomed Pharmacother 2023; 161:114420. [PMID: 36812713 DOI: 10.1016/j.biopha.2023.114420] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Breast cancer has been confirmed to have lipid disorders in the tumour microenvironment. Peroxisome proliferator-activated receptor alpha (PPARα) is a ligand-activated transcriptional factor that belongs to the family of nuclear receptors. PPARα regulates the expression of genes involved in fatty acid homeostasis and is a major regulator of lipid metabolism. Because of its effects on lipid metabolism, an increasing number of studies have investigated the relationship of PPARα with breast cancer. PPARα has been shown to impact the cell cycle and apoptosis in normal cells and tumoral cells through regulating genes of the lipogenic pathway, fatty acid oxidation, fatty acid activation, and uptake of exogenous fatty acids. Besides, PPARα is involved in the regulation of the tumour microenvironment (anti-inflammation and inhibition of angiogenesis) by modulating different signal pathways such as NF-κB and PI3K/AKT/mTOR. Some synthetic PPARα ligands are used in adjuvant therapy for breast cancer. PPARα agonists are reported to reduce the side effects of chemotherapy and endocrine therapy. In addition, PPARα agonists enhance the curative effects of targeted therapy and radiation therapy. Interestingly, with the emerging role of immunotherapy, attention has been focused on the tumour microenvironment. The dual functions of PPARα agonists in immunotherapy need further research. This review aims to consolidate the operations of PPARα in lipid-related and other ways, as well as discuss the current and potential applications of PPARα agonists in tackling breast cancer.
Collapse
Affiliation(s)
- Zhiwen Qian
- Department of Oncology, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Lingyan Chen
- Department of Oncology, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Jiayu Liu
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| | - Ying Jiang
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China; Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China.
| |
Collapse
|
129
|
Wang R, Yu W, Zhu T, Lin F, Hua C, Ru L, Guo P, Wan X, Xue G, Guo Z, Han S, Lv K, Zhang G, Ge H, Guo W, Xu L, Deng W. MED27 plays a tumor-promoting role in breast cancer progression by targeting KLF4. Cancer Sci 2023. [PMID: 36786527 DOI: 10.1111/cas.15757] [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: 08/30/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
The mediator complex usually cooperates with transcription factors to be involved in RNA polymerase II-mediated gene transcription. As one component of this complex, MED27 has been reported in our previous studies to promote thyroid cancer and melanoma progression. However, the precise function of MED27 in breast cancer development remains poorly understood. Here, we found that MED27 was more highly expressed in breast cancer samples than in normal tissues, especially in triple-negative breast cancer, and its expression level was elevated with the increase in pathological stage. MED27 knockdown in triple-negative breast cancer cells inhibited cancer cell metastasis and stemness maintenance, which was accompanied by downregulation of the expression of EMT- and stem traits-associated proteins, and vice versa in non-triple-negative breast cancer. Furthermore, MED27 knockdown sensitized breast cancer cells to epirubicin treatment by inducing cellular apoptosis and reducing tumorsphere-forming ability. Based on RNA-seq, we identified KLF4 as the possible downstream target of MED27. KLF4 overexpression reversed the MED27 silencing-mediated arrest of cellular metastasis and stemness maintenance capacity in breast cancer in vitro and in vivo. Mechanistically, MED27 transcriptionally regulated KLF4 by binding to its promoter region at positions -156 to +177. Collectively, our study not only demonstrated the tumor-promoting role of MED27 in breast cancer progression by transcriptionally targeting KLF4, but also suggested the possibility of developing the MED27/KLF4 signaling axis as a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Ruozhu Wang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianhua Zhu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fei Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Chunyu Hua
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liyuan Ru
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ping Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xinyu Wan
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guoqing Xue
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ziyue Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilong Han
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kuan Lv
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guohui Zhang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hanxiao Ge
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lingzhi Xu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| |
Collapse
|
130
|
Kim M, Lee J, Kim J, Choi B, Ki S. Effect of neoadjuvant chemotherapy on effect-site concentration of propofol for sedation in patients with breast cancer. Anesth Pain Med (Seoul) 2023; 18:29-36. [PMID: 36746899 PMCID: PMC9902627 DOI: 10.17085/apm.22201] [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: 07/04/2022] [Accepted: 12/05/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Some studies have demonstrated that chemotherapy drugs enhance sensitivity to anesthetics owing to its systemic toxicity, while others have demonstrated that chemotherapy drugs have no effect. This study aimed to determine whether neoadjuvant chemotherapy influences the effect-site concentration (Ce) of propofol for sedation in patients withbreast cancer. METHODS This study included patients aged 19-75 years who were scheduled to undergobreast cancer surgery under general anesthesia. Patients who received neoadjuvant chemotherapy were assigned to group C, whereas those who never received chemotherapy wereassigned to group N. Propofol was administered through an effect-site target-controlled infusion, and the Modified Observer's Assessment of Alertness/Sedation scale (MOAA/S) scoreand Bispectral Index (BIS) were recorded. When the plasma concentration and Ce wereequal to the target Ce, and if the MOAA/S score did not change, the target Ce was increasedby 0.2 μg/ml; otherwise, the Ce was maintained for 2 min and then increased. This processwas repeated until the MOAA/S score became 0. RESULTS No significant differences were observed in Ce values at each sedation level between both groups. Ce values for loss of consciousness (LOC) of groups C and N were 2.76± 0.29 and 2.67 ± 0.27 μg/ml (P = 0.285), respectively. However, the BIS value at LOC ofgroup C (63.87 ± 7.04) was lower than that (68.44 ± 6.01) of group N (P = 0.018). CONCLUSIONS Neoadjuvant chemotherapy for breast cancer has no effect on the Ce ofpropofol for sedation.
Collapse
Affiliation(s)
- Myounghun Kim
- Department of Anesthesiology and Pain Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Jeonghan Lee
- Department of Anesthesiology and Pain Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Jinhyeok Kim
- Department of Anesthesiology and Pain Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Beomseok Choi
- Department of Anesthesiology and Pain Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Seunghee Ki
- Department of Anesthesiology and Pain Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Korea,Corresponding author: Seunghee Ki, M.D. Department of Anesthesiology and Pain Medicine, Busan Paik Hospital, Inje University College of Medicine, 75 Bokjiro, Busanjin-gu, Busan 47392, Korea Tel: 82-51-890-6520 Fax: 82-51-898-4216 E-mail:
| |
Collapse
|
131
|
Meirelles LEDF, de Souza MVF, Carobeli LR, Morelli F, Mari NL, Damke E, Shinobu Mesquita CS, Teixeira JJV, Consolaro MEL, da Silva VRS. Combination of Conventional Drugs with Biocompounds Derived from Cinnamic Acid: A Promising Option for Breast Cancer Therapy. Biomedicines 2023; 11:275. [PMID: 36830811 PMCID: PMC9952910 DOI: 10.3390/biomedicines11020275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Despite the options available for breast cancer (BC) therapy, several adverse effects and resistance limit the success of the treatment. Furthermore, the use of a single drug is associated with a high failure rate. We investigated through a systematic review the in vitro effects of the combination between conventional drugs and bioactive compounds derived from cinnamic acid in BC treatment. The information was acquired from the following databases: PubMed, Web of Science, Embase, Scopus, Lilacs and Cochrane library. We focused on "Cinnamates", "Drug Combinations" and "Breast neoplasms" for publications dating between January 2012 and December 2022, based on the PRISMA statement. The references of the articles were carefully reviewed. Finally, nine eligible studies were included. The majority of these studies were performed using MCF-7, MDA-MB-231, MDA-MB-468 and BT-20 cell lines and the combination between cisplatin, paclitaxel, doxorubicin, tamoxifen, dactolisib and veliparib, with caffeic acid phenethyl ester, eugenol, 3-caffeoylquinic acid, salvianolic acid A, ferulic acid, caffeic acid, rosmarinic acid and ursolic acid. The combination improved overall conventional drug effects, with increased cytotoxicity, antimigratory effect and reversing resistance. Combining conventional drugs with bioactive compounds derived from cinnamic acid could emerge as a privileged scaffold for establishing new treatment options for different BC types.
Collapse
|
132
|
Wu M, Zhao T, Zhang Q, Zhang T, Wang L, Sun G. Prognostic analysis of breast cancer in Xinjiang based on Cox proportional hazards model and two-step cluster method. Front Oncol 2023; 12:1044945. [PMID: 36733362 PMCID: PMC9887128 DOI: 10.3389/fonc.2022.1044945] [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: 09/15/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023] Open
Abstract
Objective To examine the factors that affect the prognosis and survival of breast cancer patients who were diagnosed at the Affiliated Cancer Hospital of Xinjiang Medical University between 2015 and 2021, forecast the overall survival (OS), and assess the clinicopathological traits and risk level of prognosis of patients in various subgroups. Method First, nomogram model was constructed using the Cox proportional hazards models to identify the independent prognostic factors of breast cancer patients. In order to assess the discrimination, calibration, and clinical utility of the model, additional tools such as the receiver operating characteristic (ROC) curve, calibration curve, and clinical decision curve analysis (DCA) were used. Finally, using two-step cluster analysis (TCA), the patients were grouped in accordance with the independent prognostic factors. Kaplan-Meier survival analysis was employed to compare prognostic risk among various subgroups. Result T-stage, N-stage, M-stage, molecular subtyping, type of operation, and involvement in postoperative chemotherapy were identified as the independent prognostic factors. The nomogram was subsequently constructed and confirmed. The area under the ROC curve used to predict 1-, 3-, 5- and 7-year OS were 0.848, 0.820, 0.813, and 0.791 in the training group and 0.970, 0.898, 0.863, and 0.798 in the validation group, respectively. The calibration curves of both groups were relatively near to the 45° reference line. And the DCA curve further demonstrated that the nomogram has a higher clinical utility. Furthermore, using the TCA, the patients were divided into two subgroups. Additionally, the two groups' survival curves were substantially different. In particular, in the group with the worse prognosis (the majority of patients did not undergo surgical therapy or postoperative chemotherapy treatment), the T-, N-, and M-stage were more prevalent in the advanced, and the total points were likewise distributed in the high score side. Conclusion For the survival and prognosis of breast cancer patients in Xinjiang, the nomogram constructed in this paper has a good prediction value, and the clustering results further demonstrated that the selected factors were important. This conclusion can give a scientific basis for tailored treatment and is conducive to the formulation of focused treatment regimens for patients in practical practice.
Collapse
Affiliation(s)
- Mengjuan Wu
- Country College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Ting Zhao
- Department of Medical Record Management, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Qian Zhang
- Information Management and Big Date Center, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tao Zhang
- Country College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Lei Wang
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi, China
| | - Gang Sun
- Xinjiang Cancer Center/Key Laboratory of Oncology of Xinjiang Uyghur Autonomous Region, Urumqi, Xinjiang, China
- Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
133
|
Jugulytė N, Žukienė G, Bartkevičienė D. Emerging Use of Vaginal Laser to Treat Genitourinary Syndrome of Menopause for Breast Cancer Survivors: A Review. Medicina (B Aires) 2023; 59:medicina59010132. [PMID: 36676756 PMCID: PMC9860929 DOI: 10.3390/medicina59010132] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/03/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Breast cancer treatment, such as chemotherapy and endocrine therapy, can cause earlier and more sudden menopausal symptoms. Genitourinary syndrome of menopause (GSM) is one of the most bothersome side effects of breast cancer treatment, resulting in sexual dysfunction and impaired quality of life. GSM includes genital, urinary, and sexual symptoms. However, alleviating symptoms of GSM for breast cancer survivors may be challenging due to ineffectiveness, contraindications, and low adherence to treatment. The most recent data show the feasibility and safety of vaginal laser to treat GSM for breast cancer survivors. This narrative review provides the aspects of GSM in breast cancer patients, putting the focus on the efficacy and safety of vaginal laser therapy.
Collapse
Affiliation(s)
- Nida Jugulytė
- Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
- Correspondence:
| | - Guoda Žukienė
- Clinic of Obstetrics and Gynaecology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| | - Daiva Bartkevičienė
- Clinic of Obstetrics and Gynaecology, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
| |
Collapse
|
134
|
Kola P, Nagesh PKB, Roy PK, Deepak K, Reis RL, Kundu SC, Mandal M. Innovative nanotheranostics: Smart nanoparticles based approach to overcome breast cancer stem cells mediated chemo- and radioresistances. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1876. [PMID: 36600447 DOI: 10.1002/wnan.1876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
The alarming increase in the number of breast cancer patients worldwide and the increasing death rate indicate that the traditional and current medicines are insufficient to fight against it. The onset of chemo- and radioresistances and cancer stem cell-based recurrence make this problem harder, and this hour needs a novel treatment approach. Competent nanoparticle-based accurate drug delivery and cancer nanotheranostics like photothermal therapy, photodynamic therapy, chemodynamic therapy, and sonodynamic therapy can be the key to solving this problem due to their unique characteristics. These innovative formulations can be a better cargo with fewer side effects than the standard chemotherapy and can eliminate the stability problems associated with cancer immunotherapy. The nanotheranostic systems can kill the tumor cells and the resistant breast cancer stem cells by novel mechanisms like local hyperthermia and reactive oxygen species and prevent tumor recurrence. These theranostic systems can also combine with chemotherapy or immunotherapy approaches. These combining approaches can be the future of anticancer therapy, especially to overcome the breast cancer stem cells mediated chemo- and radioresistances. This review paper discusses several novel theranostic systems and smart nanoparticles, their mechanism of action, and their modifications with time. It explains their relevance and market scope in the current era. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Prithwish Kola
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | | | - Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Rui Luis Reis
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimaraes, Portugal
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
135
|
Xuan DTM, Wu CC, Wang WJ, Hsu HP, Ta HDK, Anuraga G, Chiao CC, Wang CY. Glutamine synthetase regulates the immune microenvironment and cancer development through the inflammatory pathway. Int J Med Sci 2023; 20:35-49. [PMID: 36619229 PMCID: PMC9812810 DOI: 10.7150/ijms.75625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/03/2022] [Indexed: 12/23/2022] Open
Abstract
Although adjuvant tamoxifen therapy is beneficial to estrogen receptor-positive (ER+) breast cancer patients, a significant number of patients still develop metastasis or undergo recurrence. Therefore, identifying novel diagnostic and prognostic biomarkers for these patients is urgently needed. Predictive markers and therapeutic strategies for tamoxifen-resistant ER+ breast cancer are not clear, and micro (mi)RNAs have recently become a focal research point in cancer studies owing to their regulation of gene expressions, metabolism, and many other physiological processes. Therefore, systematic investigation is required to understand the modulation of gene expression in tamoxifen-resistant patients. High-throughput technology uses a holistic approach to observe differences among expression profiles of thousands of genes, which provides a comprehensive level to extensively investigate functional genomics and biological processes. Through a bioinformatics analysis, we revealed that glutamine synthetase/glutamate-ammonia ligase (GLUL) might play essential roles in the recurrence of tamoxifen-resistant ER+ patients. GLUL increases intracellular glutamine usage via glutaminolysis, and further active metabolism-related downstream molecules in cancer cell. However, how GLUL regulates the tumor microenvironment for tamoxifen-resistant ER+ breast cancer remains unexplored. Analysis of MetaCore pathway database demonstrated that GLUL is involved in the cell cycle, immune response, interleukin (IL)-4-induced regulators of cell growth, differentiation, and metabolism-related pathways. Experimental data also confirmed that the knockdown of GLUL in breast cancer cell lines decreased cell proliferation and influenced expressions of specific downstream molecules. Through a Connectivity Map (CMap) analysis, we revealed that certain drugs/molecules, including omeprazole, methacholine chloride, ioversol, fulvestrant, difenidol, cycloserine, and MK-801, may serve as potential treatments for tamoxifen-resistant breast cancer patients. These drugs may be tested in combination with current therapies in tamoxifen-resistant breast cancer patients. Collectively, our study demonstrated the crucial roles of GLUL, which provide new targets for the treatment of tamoxifen-resistant breast cancer patients.
Collapse
Affiliation(s)
- Do Thi Minh Xuan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Chung-Che Wu
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jan Wang
- Research Center for Cancer Biology, China Medical University, Taichung 40676, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hoang Dang Khoa Ta
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan
| | - Gangga Anuraga
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan.,Department of Statistics, Faculty of Science and Technology, PGRI Adi Buana University, East Java, Surabaya 60234, Indonesia
| | - Chung-Chieh Chiao
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Yang Wang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science, Taipei Medical University, Taipei 11031, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
136
|
Appiah CO, Singh M, May L, Bakshi I, Vaidyanathan A, Dent P, Ginder G, Grant S, Bear H, Landry J. The epigenetic regulation of cancer cell recovery from therapy exposure and its implications as a novel therapeutic strategy for preventing disease recurrence. Adv Cancer Res 2023; 158:337-385. [PMID: 36990536 DOI: 10.1016/bs.acr.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The ultimate goal of cancer therapy is the elimination of disease from patients. Most directly, this occurs through therapy-induced cell death. Therapy-induced growth arrest can also be a desirable outcome, if prolonged. Unfortunately, therapy-induced growth arrest is rarely durable and the recovering cell population can contribute to cancer recurrence. Consequently, therapeutic strategies that eliminate residual cancer cells reduce opportunities for recurrence. Recovery can occur through diverse mechanisms including quiescence or diapause, exit from senescence, suppression of apoptosis, cytoprotective autophagy, and reductive divisions resulting from polyploidy. Epigenetic regulation of the genome represents a fundamental regulatory mechanism integral to cancer-specific biology, including the recovery from therapy. Epigenetic pathways are particularly attractive therapeutic targets because they are reversible, without changes in DNA, and are catalyzed by druggable enzymes. Previous use of epigenetic-targeting therapies in combination with cancer therapeutics has not been widely successful because of either unacceptable toxicity or limited efficacy. The use of epigenetic-targeting therapies after a significant interval following initial cancer therapy could potentially reduce the toxicity of combination strategies, and possibly exploit essential epigenetic states following therapy exposure. This review examines the feasibility of targeting epigenetic mechanisms using a sequential approach to eliminate residual therapy-arrested populations, that might possibly prevent recovery and disease recurrence.
Collapse
Affiliation(s)
- Christiana O Appiah
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States; Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, United States
| | - Manjulata Singh
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Lauren May
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Ishita Bakshi
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Ashish Vaidyanathan
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Gordon Ginder
- Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Steven Grant
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States; Department of Internal Medicine, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States; Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States; Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, Richmond, VA, United States
| | - Harry Bear
- Department of Surgery, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, VA, United States; Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Massey Cancer Center, Richmond, Richmond, VA, United States
| | - Joseph Landry
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| |
Collapse
|
137
|
Thimotheo Batista JP, Santos Marzano LA, Menezes Silva RA, de Sá Rodrigues KE, Simões E Silva AC. Chemotherapy and Anticancer Drugs Adjustment in Obesity: A Narrative Review. Curr Med Chem 2023; 30:1003-1028. [PMID: 35946096 DOI: 10.2174/0929867329666220806140204] [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: 10/18/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Obese individuals have higher rates of cancer incidence and cancer- related mortality. The worse chemotherapy outcomes observed in this subset of patients are multifactorial, including the altered physiology in obesity and its impact on pharmacokinetics, the possible increased risk of underdosing, and treatment-related toxicity. AIMS The present review aimed to discuss recent data on physiology, providing just an overall perspective and pharmacokinetic alterations in obesity concerning chemotherapy. We also reviewed the controversies of dosing adjustment strategies in adult and pediatric patients, mainly addressing the use of actual total body weight and ideal body weight. METHODS This narrative review tried to provide the best evidence to support antineoplastic drug dosing strategies in children, adolescents, and adults. RESULTS Cardiovascular, hepatic, and renal alterations of obesity can affect the distribution, metabolism, and clearance of drugs. Anticancer drugs have a narrow therapeutic range, and variations in dosing may result in either toxicity or underdosing. Obese patients are underrepresented in clinical trials that focus on determining recommendations for chemotherapy dosing and administration in clinical practice. After considering associated comorbidities, the guidelines recommend that chemotherapy should be dosed according to body surface area (BSA) calculated with actual total body weight, not an estimate or ideal weight, especially when the intention of therapy is the cure. CONCLUSION The actual total body weight dosing appears to be a better approach to dosing anticancer drugs in both adults and children when aiming for curative results, showing no difference in toxicity and no limitation in treatment outcomes compared to adjusted doses.
Collapse
Affiliation(s)
- João Pedro Thimotheo Batista
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil
| | - Lucas Alexandre Santos Marzano
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil
| | - Renata Aguiar Menezes Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil
| | - Karla Emília de Sá Rodrigues
- Departmento de Pediatria, Faculdade de Medicina, Universidade Federal de Minas Gerais, CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efgênia, Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil.,Departmento de Pediatria, Faculdade de Medicina, Universidade Federal de Minas Gerais, CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efgênia, Belo Horizonte, MG, Brazil
| |
Collapse
|
138
|
Chen D, Liu X, Lu X, Tian J. Nanoparticle drug delivery systems for synergistic delivery of tumor therapy. Front Pharmacol 2023; 14:1111991. [PMID: 36874010 PMCID: PMC9978018 DOI: 10.3389/fphar.2023.1111991] [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: 11/30/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Nanoparticle drug delivery systems have proved anti-tumor effects; however, they are not widely used in tumor therapy due to insufficient ability to target specific sites, multidrug resistance to anti-tumor drugs, and the high toxicity of the drugs. With the development of RNAi technology, nucleic acids have been delivered to target sites to replace or correct defective genes or knock down specific genes. Also, synergistic therapeutic effects can be achieved for combined drug delivery, which is more effective for overcoming multidrug resistance of cancer cells. These combination therapies achieve better therapeutic effects than delivering nucleic acids or chemotherapeutic drugs alone, so the scope of combined drug delivery has also been expanded to three aspects: drug-drug, drug-gene, and gene-gene. This review summarizes the recent advances of nanocarriers to co-delivery agents, including i) the characterization and preparation of nanocarriers, such as lipid-based nanocarriers, polymer nanocarriers, and inorganic delivery carriers; ii) the advantages and disadvantages of synergistic delivery approaches; iii) the effectual delivery cases that are applied in the synergistic delivery systems; and iv) future perspectives in the design of nanoparticle drug delivery systems to co-deliver therapeutic agents.
Collapse
Affiliation(s)
- Daoyuan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Xuecun Liu
- Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Xiaoyan Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
139
|
Hussen BM, Mohamadtahr S, Abdullah SR, Hidayat HJ, Rasul MF, Hama Faraj GS, Ghafouri-Fard S, Taheri M, Khayamzadeh M, Jamali E. Exosomal circular RNAs: New player in breast cancer progression and therapeutic targets. Front Genet 2023; 14:1126944. [PMID: 36926585 PMCID: PMC10011470 DOI: 10.3389/fgene.2023.1126944] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Breast cancer is the most prevalent type of malignancy among women. Exosomes are extracellular vesicles of cell membrane origin that are released via exocytosis. Their cargo contains lipids, proteins, DNA, and different forms of RNA, including circular RNAs. Circular RNAs are new class of non-coding RNAs with a closed-loop shape involved in several types of cancer, including breast cancer. Exosomes contained a lot of circRNAs which are called exosomal circRNAs. By interfering with several biological pathways, exosomal circRNAs can have either a proliferative or suppressive role in cancer. The involvement of exosomal circRNAs in breast cancer has been studied with consideration to tumor development and progression as well as its effects on therapeutic resistance. However, its exact mechanism is still unclear, and there have not been available clinical implications of exo-circRNAs in breast cancer. Here, we highlight the role of exosomal circRNAs in breast cancer progression and to highlight the most recent development and potential of circRNAas therapeutic targets and diagnostics for breast cancer.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Medical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Sayran Mohamadtahr
- Department of Medical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | | | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University-Erbil, Erbil, Iraq
| | - Mohammad Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Iraq
| | - Goran Sedeeq Hama Faraj
- Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaimany, Iraq
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.,Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Khayamzadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Academy of Medical Sciences, Tehran, Iran
| | - Elena Jamali
- Department of Pathology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
140
|
Długosz-Pokorska A, Perlikowska R, Janecki T, Janecka A. New Uracil Analog with Exocyclic Methylidene Group Can Reverse Resistance to Taxol in MCF-7 Cancer Cells. Biologics 2023; 17:69-83. [PMID: 37213261 PMCID: PMC10198386 DOI: 10.2147/btt.s405080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/10/2023] [Indexed: 05/23/2023]
Abstract
Introduction Taxol (Tx), a microtubule-stabilizing drug, has been widely used as a chemotherapeutic in several types of cancer. However, the development of resistance limited its application. One of the strategies used to prevent the emergence of drug resistance is combination treatment, involving at least two drugs. The aim of the current study was to assess if a new uracil analog, 3-p-bromophenyl-1-ethyl-5-methylidenedihydrouracil (U-359) can prevent the development of Tx resistance in breast cancer cells. Methods The cytotoxicity of the new drug was tested in MCF-7 (hormone receptor (ER, PR) positive cell-line) and MCF-10A cell lines using MTT method. For the detection of apoptosis and necrosis, the Wright and Giemsa staining was used. Gene expression was measured by real-time PCR, and changes in the protein levels were evaluated by ELISA and bioluminescent method. Results We investigated the effect of Tx and U-359 on cancer MCF-7 and normal MCF-10A cells alone and in combination. Tx co-administered with U-359 inhibited proliferation of MCF-7 cells to 7% while the level of ATPase drastically decreased to 14%, compared with effects produced by Tx alone. The apoptosis process was induced through the mitochondrial pathway. These effects were not seen in MCF-10A cells, showing the wide safety margin. The obtained results have shown that U-359 produced a synergistic effect with Tx probably by reducing Tx resistance in MCF-7 cells. To elucidate the possible mechanism of resistance, expression of tubulin III (TUBIII), responsible for microtubule stabilization and tau and Nlp proteins, responsible for microtubule dynamics, was assessed. Conclusion Combination of Tx with U-359 reduced overexpression of TUBIII and Nlp. Thus, U-359 may stand for a potential reversal agent for the treatment of MDR in cancer cells.
Collapse
Affiliation(s)
- Angelika Długosz-Pokorska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
- Correspondence: Angelika Długosz-Pokorska, Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, Lodz, 92-215, Poland, Tel +48 42 2725706, Fax +48 42 678 42 77, Email
| | - Renata Perlikowska
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Tomasz Janecki
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
141
|
Hanes R, Ayuda-Durán P, Rønneberg L, Nakken S, Hovig E, Zucknick M, Enserink JM. screenwerk: a modular tool for the design and analysis of drug combination screens. Bioinformatics 2022; 39:6961189. [PMID: 36573326 PMCID: PMC9825784 DOI: 10.1093/bioinformatics/btac840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/14/2022] [Accepted: 12/26/2022] [Indexed: 12/28/2022] Open
Abstract
MOTIVATION There is a rapidly growing interest in high-throughput drug combination screening to identify synergizing drug interactions for treatment of various maladies, such as cancer and infectious disease. This creates the need for pipelines that can be used to design such screens, perform quality control on the data and generate data files that can be analyzed by synergy-finding bioinformatics applications. RESULTS screenwerk is an open-source, end-to-end modular tool available as an R-package for the design and analysis of drug combination screens. The tool allows for a customized build of pipelines through its modularity and provides a flexible approach to quality control and data analysis. screenwerk is adaptable to various experimental requirements with an emphasis on precision medicine. It can be coupled to other R packages, such as bayesynergy, to identify synergistic and antagonistic drug interactions in cell lines or patient samples. screenwerk is scalable and provides a complete solution for setting up drug sensitivity screens, read raw measurements and consolidate different datasets, perform various types of quality control and analyze, report and visualize the results of drug sensitivity screens. AVAILABILITY AND IMPLEMENTATION The R-package and technical documentation is available at https://github.com/Enserink-lab/screenwerk; the R source code is publicly available at https://github.com/Enserink-lab/screenwerk under GNU General Public License v3.0; bayesynergy is accessible at https://github.com/ocbe-uio/bayesynergy. Selected modules are available through Galaxy, an open-source platform for FAIR data analysis at https://oncotools.elixir.no. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Robert Hanes
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway,Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway,Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
| | - Pilar Ayuda-Durán
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway,Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Leiv Rønneberg
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, 0317 Oslo, Norway,MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - Sigve Nakken
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway,Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo 0379, Norway,Department of Informatics, Centre for Bioinformatics, University of Oslo, Oslo 0372, Norway
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo 0379, Norway,Department of Informatics, Centre for Bioinformatics, University of Oslo, Oslo 0372, Norway
| | - Manuela Zucknick
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, 0317 Oslo, Norway
| | | |
Collapse
|
142
|
Highly Biocompatible Apigenin-Loaded Silk Fibroin Nanospheres: Preparation, Characterization, and Anti-Breast-Cancer Activity. Polymers (Basel) 2022; 15:polym15010023. [PMID: 36616371 PMCID: PMC9823476 DOI: 10.3390/polym15010023] [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: 11/16/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is among the most common fatal diseases among women. Low-toxicity apigenin (AGN) is of interest due to its good antitumor activity, but its clinical application is severely limited due to its poor water solubility and low bioavailability. An effective strategy to enhance the anti-breast-cancer activity of AGN is to develop it as a nanodelivery system. Silk fibroin (SF) is an ideal drug carrier with good biocompatibility, biodegradability, and a simple extraction process. This paper develops a novel and efficient apigenin-loaded silk fibroin nanodelivery system (SF-AGN) by nanoprecipitation with SF as a carrier. The system was characterized in terms of morphology, zeta potential, particle size, ultraviolet (UV), infrared (IR), and synchronous thermal analyses (TG-DSC), and the in vitro cytotoxicity and in vivo pharmacokinetics were examined. Finally, the chronic toxicity of SF-AGN in mice was studied. The SF-AGN nanodelivery system has good dispersibility, a hydrated particle size of 163.35 nm, a zeta potential of -18.5 mV, an average drug loading of 6.20%, and good thermal stability. MTT studies showed that SF-AGN significantly enhanced the inhibitory effect of AGN on 4T1 and MDA-MB-231 cells. Pharmacokinetic studies have demonstrated that SF-AGN can dramatically improve the bioavailability of AGN. The results of toxicity experiments showed that SF-AGN is biocompatible and does not alter normal tissues or organs. In sum, the SF-AGN nanodelivery system is a promising drug-delivery system for the clinical treatment of breast cancer.
Collapse
|
143
|
Current evidence on circRNAs as potential theranostic markers for detecting chemoresistance in breast cancer: a systematic review and meta‑analysis. Sci Rep 2022; 12:22016. [PMID: 36539545 PMCID: PMC9768200 DOI: 10.1038/s41598-022-26220-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
This study assessed the value of circRNAs (circular RNAs) as prognostic markers in BC (breast cancer). We searched pertinent studies on the PubMed, Embase, and Web of Science online databases published according to PRISMA guidelines. A random-effects model for meta-analysis was used to assess the combined effect size of the HRs (hazard ratios) of the included studies. The heterogeneity test used Cochran's Q-test and I2 statistics. Thirty of the 520 trials retrieved were included in the systematic review. A total of 11 chemotherapeutic agents were used in the included studies. A total of 30 studies on 30 circRNAs were included in the systematic review. Of the 30 relevant circRNAs, 28 were upregulated and two were downregulated in breast cancer versus normal samples, and both were associated with increased drug resistance. Nine of 30 studies were used for the meta-analysis. The results of the meta-analysis showed that the groups with circRNA upregulation and circRNA downregulation showed the same prognostic risk (HR = 1.37, 95% Cl: 0.80-2.36, I2 = 63.7%). The results of subgroup analysis showed that both upregulated circRNAs (HR = 2.24, 95% Cl: 1.34-3.75, I2 = 0%) and downregulated circRNAs (HR = 0.61, 95% Cl: 0.45-0.83, I2 = 0%) were associated with poor BC prognosis. Collectively, the results of all relevant articles collected indicated that circRNAs showed good potential as possible clinical biomarkers of chemoresistance in BC patients.
Collapse
|
144
|
Marinho PML, Lima RB, Santos JCDO, Santos DKDC, Silva GM, Kameo SY, Sawada NO. Clinical-Epidemiological Profile and Health-Related Quality of Life of Women with Breast Cancer During Chemotherapy Treatment: Observational Study. REVISTA BRASILEIRA DE CANCEROLOGIA 2022. [DOI: 10.32635/2176-9745.rbc.2022v68n4.3164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction: Breast cancer may affect different profiles of women worldwide. In addition, chemotherapy to treat breast neoplasms directly affects health-related quality of life. Objective: To describe the clinical-epidemiological profile and to compare the general and specific health-related quality of life of women with breast cancer during chemotherapy. Method: In an observational and prospective study, 140 women with breast cancer in northeastern Brazil were evaluated in the intermediate cycle and at the end of chemotherapy. Quality of life was assessed using a general instrument (EORTC-QLQ-C30) and a specific module (EORTC-QLQ-BR23). Data were compared with a paired non-parametric test, with a significance level of 5%. Results: Regarding the clinical-epidemiological profile, the median age was 50 years, 82.9% were black or mixed-race, and 95% lived in Sergipe, Brazil. In addition, 50.7% had no occupation, the median family income was one minimum wage, as well as the median of education was eight years of study and only 40.7% have completed high school. Considering the data from the C30 and BR23 questionnaires, it was observed that most items and scales worsened at the end of chemotherapy when compared to the intermediate cycle. Among the items and scales with significant differences, most had moderate or high effect sizes. Conclusion: It is possible to conclude that the clinical-epidemiological profile was unfavorable and chemotherapy reduced several aspects of the health-related quality of life of women with breast cancer.
Collapse
|
145
|
Barros LLE, Natale PD, Donoso MT, Mendoza IYQ, Souza ADD, Ercole FF, Simino GPR. Efeitos adversos locorregionais da terapêutica oncológica em mulheres com câncer de mama avançado. REME: REVISTA MINEIRA DE ENFERMAGEM 2022. [DOI: 10.35699/2316-9389.2022.40564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
Abstract
Objetivo: identificar os efeitos adversos locorregionais da administração da terapêutica oncológica endovenosa em mulheres com câncer de mama avançado. Método: revisão integrativa da literatura, que utilizou as bases de dados PubMed/MEDLINE, CINAHL, LILACS e EMBASE, sem recorte temporal, além de busca reversa dos artigos selecionados, atualizada até maio de 2022 A população contemplou mulheres com câncer de mama avançado submetidas à intervenção com terapêutica oncológica endovenosa com quimioterapia ou hormonioterapia ou anticorpo monoclonal, e o desfecho avaliou efeitos adversos locorregionais Resultados: identificaram-se 2.789 estudos, e a amostra final foi composta por 8 ensaios clínicos e 1 estudo observacional retrospectivo, sendo todos estudos internacionais e publicados no período de 1986 a 2018. Predominantemente, as pacientes tinham câncer de mama em estádio IV, idade de 50 anos ou mais e múltiplas metástases. Os efeitos adversos locorregionais foram: flebite, ulceração e/ou necrose, dor, eritema e reação no local da injeção não especificada. Os estudos não trazem detalhamento do tipo de cateter venoso, osmolaridade dos fármacos e cuidados preventivos para diminuição desses efeitos adversos. Conclusão: as evidências desses artigos mostraram que os efeitos adversos locorregionais estão presentes em estudos de eficácia dos fármacos oncológicos em mulheres com câncer de mama avançado. No entanto, destaca-se que a segurança da administração dos fármacos oncológicos não se apresenta elucidada nessa revisão, indicando necessidade de estudos de acompanhamento dos efeitos adversos.
Collapse
|
146
|
Zhou W, Ma X, Wang J, Xu X, Koivisto O, Feng J, Viitala T, Zhang H. Co-delivery CPT and PTX prodrug with a photo/thermo-responsive nanoplatform for triple-negative breast cancer therapy. SMART MEDICINE 2022; 1:e20220036. [PMID: 39188747 PMCID: PMC11235718 DOI: 10.1002/smmd.20220036] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is still the most aggressive cancer in women. Combination chemotherapy holds great potential for cancer therapy; however, the off-target and side effects of free chemotherapy administration remain a major challenge. In this study, we developed a photo/thermo-responsive nanoplatform that can be used for TNBC treatment via photothermic therapy in combination with multidrug therapy. By conjugating the chemotherapy drug PTX prodrug on the surface of mesoporous silica-coated gold nanorod nanoparticles and then loading another chemotherapy drug, CPT, the Au@MSN-PTX@CPT nanoparticles exhibited great photothermal response, redox response drug release and cancer cell inhibition abilities. Otherwise, we further coated the Au@MSN-PTX@CPT nanoparticle with a temperature-sensitive polymer poly(N-isopropylacrylamide-co-methacrylic acid) (p(NIPAM-co-MAAc)), and the polymer-coated Au@MSN-PTX@TPT@polymer nanoparticles showed perfect near-infrared (NIR) light controlled drug release. Finally, the Au@MSN-PTX@CPT@polymer nanoparticles were injected into the 4T1 breast cancer mouse model. The Au@MSN-PTX@CPT@polymer nanoparticles preferably accumulated at the tumor site and had reduced chemotherapy injuries and great antitumor activity when combined with 650 nm laser treatment. In summary, our developed Au@MSN-PTX@CPT@polymer nanoparticles served as a good method for controlled chemodrug delivery and provided a good choice for TNBC combination therapy.
Collapse
Affiliation(s)
- Wenhui Zhou
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
- Southern Medical University Affiliated Fengxian HospitalShanghaiChina
| | - Xiaodong Ma
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Jie Wang
- Southern Medical University Affiliated Fengxian HospitalShanghaiChina
| | - Xiaoyu Xu
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Oliver Koivisto
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Jing Feng
- Southern Medical University Affiliated Fengxian HospitalShanghaiChina
- Longgang District People's Hospital of ShenzhenShenzhenChina
| | - Tapani Viitala
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Hongbo Zhang
- Pharmaceutical Sciences LaboratoryÅbo Akademi UniversityTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| |
Collapse
|
147
|
Shechter O, Sausen DG, Gallo ES, Dahari H, Borenstein R. Epstein-Barr Virus (EBV) Epithelial Associated Malignancies: Exploring Pathologies and Current Treatments. Int J Mol Sci 2022; 23:14389. [PMID: 36430864 PMCID: PMC9699474 DOI: 10.3390/ijms232214389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
Epstein-Barr virus (EBV) is one of eight known herpesviruses with the potential to infect humans. Globally, it is estimated that between 90-95% of the population has been infected with EBV. EBV is an oncogenic virus that has been strongly linked to various epithelial malignancies such as nasopharyngeal and gastric cancer. Recent evidence suggests a link between EBV and breast cancer. Additionally, there are other, rarer cancers with weaker evidence linking them to EBV. In this review, we discuss the currently known epithelial malignancies associated with EBV. Additionally, we discuss and establish which treatments and therapies are most recommended for each cancer associated with EBV.
Collapse
Affiliation(s)
- Oren Shechter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Daniel G. Sausen
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Elisa S. Gallo
- Tel-Aviv Sourasky Medical Center, Division of Dermatology, Tel-Aviv 6423906, Israel
| | - Harel Dahari
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Ronen Borenstein
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
148
|
The Potential of NORAD-PUMILIO- RALGAPB Regulatory Axis as a Biomarker in Breast Cancer. Noncoding RNA 2022; 8:ncrna8060076. [PMID: 36412911 PMCID: PMC9680495 DOI: 10.3390/ncrna8060076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022] Open
Abstract
Introduction: Long non-coding RNAs (LncRNA) represent a heterogeneous family of RNAs that have emerged as regulators of various biological processes through their association with proteins in ribonucleoproteins complexes. The dynamic of these interactions can affect cell metabolism, including cancer development. Annually, breast cancer causes thousands of deaths worldwide, and searching for new biomarkers is pivotal for better diagnosis and treatment. Methods: Based on in silico prediction analysis, we focus on LncRNAs that have binding sites for PUMILIO, an RBP family involved in post-transcriptional regulation and associated with cancer progression. We compared the expression levels of these LncRNAs in breast cancer and non-tumor samples from the TCGA database. We analyzed the impact of overall and disease-free survival associated with the expression of the LncRNAs and co-expressed genes and targets of PUMILIO proteins. Results: Our results found NORAD as the most relevant LncRNA with a PUMILIO binding site in breast cancer, differently expressed between Luminal A and Basal subtypes. Additionally, NORAD was co-expressed in a Basal-like subtype (0.55) with the RALGAPB gene, a target gene of PUMILIO related to chromosome stability during cell division. Conclusion: These data suggest that this molecular axis may provide insights for developing novel therapeutic strategies for breast cancer.
Collapse
|
149
|
Liu L, Chen Y, Chen L, Shi Y, Fang J, Zhao M, Wang M. Preparation and pharmacodynamics of Niclosamide-hydroxypropyl-β-cyclodextrin inclusion complex. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
150
|
Ding P, Gao Y, Wang J, Xiang H, Zhang C, Wang L, Ji G, Wu T. Progress and challenges of multidrug resistance proteins in diseases. Am J Cancer Res 2022; 12:4483-4501. [PMID: 36381332 PMCID: PMC9641395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/09/2022] [Indexed: 06/16/2023] Open
Abstract
Chemotherapy remains the first choice for patients with advanced cancers when other treatments are ineffective. Multidrug resistance (MDR) is an unavoidable factor that negatively affects the effectiveness of cancer chemotherapy drugs. Researchers are trying to reduce MDR, improve the effectiveness of chemotherapeutic drugs, and alleviate patient suffering to positively contribute to disease treatment. MDR also occurs in inflammation and genetic disorders, which increases the difficulty of clinically beneficial treatments. The ATP-binding cassette (ABC) is an active transporter that plays an important role in the barrier and secretory functions of many normal cells. As the C subfamily in the ABC family, multidrug resistance proteins (MRPs/ABCCs) export a variety of antitumour drugs and are expressed in a variety of cancers. The present review summarises the role of MRPs in cancer and other diseases and recent research progress of MRP inhibitors to better examine the mechanism and function of MRPs, and establish a good relationship with clinical treatment.
Collapse
Affiliation(s)
- Peilun Ding
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Ying Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Caiyun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| |
Collapse
|