1
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Ghai S, Shrestha R, Su KH. HSF1 at the crossroads of chemoresistance: from current insights to future horizons in cell death mechanisms. Front Cell Dev Biol 2025; 12:1500880. [PMID: 39850800 PMCID: PMC11754285 DOI: 10.3389/fcell.2024.1500880] [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: 09/24/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Heat Shock Factor 1 (HSF1) is a major transcriptional factor regulating the heat shock response and has become a potential target for overcoming cancer chemoresistance. This review comprehensively examines HSF1's role in chemoresistance and its potential as a therapeutic target in cancer. We explore the complex, intricate mechanism that regulates the activation of HSF1, HSF1's function in promoting resistance to chemotherapy, and the strategies used to manipulate HSF1 for therapeutic benefit. In addition, we discuss emerging research implicating HSF1's roles in autophagy, apoptosis, DNA damage repair, drug efflux, and thus chemoresistance. This article highlights the significance of HSF1 in cancer chemoresistance and its potential as a target for enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
| | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, United States
| |
Collapse
|
3
|
Al-Dewik N, Abuarja T, Younes S, Nasrallah G, Alsharshani M, Ibrahim FE, Samara M, Farrell T, Abdulrouf PV, Qoronfleh MW, Al Rifai H. Precision medicine activities and opportunities for shaping maternal and neonatal health in Qatar. Per Med 2024; 21:313-333. [PMID: 39347749 DOI: 10.1080/17410541.2024.2394397] [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: 10/17/2023] [Accepted: 08/16/2024] [Indexed: 10/01/2024]
Abstract
Precision Medicine (PM) is a transformative clinical medicine strategy that aims to revolutionize healthcare by leveraging biological information and biomarkers. In the context of maternal and neonatal health, PM enables personalized care from preconception through the postnatal period. Qatar has emerged as a key player in PM research, with dedicated programs driving advancements and translating cutting-edge research into clinical applications. This article delves into neonatal and maternal health in Qatar, emphasizing PM programs and initiatives that have been implemented. It also features noteworthy clinical cases that demonstrate the effectiveness of precision interventions. Furthermore, the article highlights the role of pharmacogenomics in addressing various maternal health conditions. The review further explores potential advancements in the application of PM in maternal and neonatal healthcare in Qatar.
Collapse
Affiliation(s)
- Nader Al-Dewik
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
- Department of Neonatology, Neonatal Intensive Care Unit, Newborn Screening Unit, Women's Wellness & Research Center, Hamad Medical Corporation, Doha, 3050, Qatar
- Translational Research Institute (TRI), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
- Genomics & Precision Medicine (GPM), College of Health & Life Science (CHLS), Hamad Bin Khalifa University (HBKU), Doha, 34110, Qatar
| | - Tala Abuarja
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Salma Younes
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University (QU), Doha, 2713, Qatar
| | - Gheyath Nasrallah
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University (QU), Doha, 2713, Qatar
| | - Mohamed Alsharshani
- Diagnostic Genetics Division (DGD), Department of Laboratory Medicine & Pathology (DLMP), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Faisal E Ibrahim
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Muthanna Samara
- Department of Psychology, Kingston University London, Kingston upon Thames, London, KT1 2EE, United Kingdom
| | - Thomas Farrell
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - Palli Valapila Abdulrouf
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
| | - M Walid Qoronfleh
- Q3 Research Institute (QRI), Healthcare Research & Policy Division, 7227 Rachel Drive, Ypsilanti, MI 48917, USA
| | - Hilal Al Rifai
- Department of Research & Translational & Precision Medicine Research Lab, Women's Wellness & Research Center (WWRC), Hamad Medical Corporation (HMC), Doha, 3050, Qatar
- Department of Neonatology, Neonatal Intensive Care Unit, Newborn Screening Unit, Women's Wellness & Research Center, Hamad Medical Corporation, Doha, 3050, Qatar
| |
Collapse
|
4
|
Malik S, Sikander M, Wahid M, Dhasmana A, Sarwat M, Khan S, Cobos E, Yallapu MM, Jaggi M, Chauhan SC. Deciphering cellular and molecular mechanism of MUC13 mucin involved in cancer cell plasticity and drug resistance. Cancer Metastasis Rev 2024; 43:981-999. [PMID: 38498072 DOI: 10.1007/s10555-024-10177-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024]
Abstract
There has been a surge of interest in recent years in understanding the intricate mechanisms underlying cancer progression and treatment resistance. One molecule that has recently emerged in these mechanisms is MUC13 mucin, a transmembrane glycoprotein. Researchers have begun to unravel the molecular complexity of MUC13 and its impact on cancer biology. Studies have shown that MUC13 overexpression can disrupt normal cellular polarity, leading to the acquisition of malignant traits. Furthermore, MUC13 has been associated with increased cancer plasticity, allowing cells to undergo epithelial-mesenchymal transition (EMT) and metastasize. Notably, MUC13 has also been implicated in the development of chemoresistance, rendering cancer cells less responsive to traditional treatment options. Understanding the precise role of MUC13 in cellular plasticity, and chemoresistance could pave the way for the development of targeted therapies to combat cancer progression and enhance treatment efficacy.
Collapse
Affiliation(s)
- Shabnam Malik
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Mohammed Sikander
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Mohd Wahid
- Unit of Research and Scientific Studies, College of Nursing and Allied Health Sciences, University of Jazan, Jizan, Saudi Arabia
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Sheema Khan
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Everardo Cobos
- Department of Medicine, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA.
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
5
|
Farooq F, Amin A, Wani UM, Lone A, Qadri RA. Shielding and nurturing: Fibronectin as a modulator of cancer drug resistance. J Cell Physiol 2023; 238:1651-1669. [PMID: 37269547 DOI: 10.1002/jcp.31048] [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: 03/22/2023] [Revised: 05/02/2023] [Accepted: 05/10/2023] [Indexed: 06/05/2023]
Abstract
Resistance to chemotherapy and targeted therapies constitute a common hallmark of most cancers and represent a dominant factor fostering tumor relapse and metastasis. Fibronectin, an abundant extracellular matrix glycoprotein, has long been proposed to play an important role in the pathobiology of cancer. Recent research has unraveled the role of Fibronectin in the onset of chemoresistance against a variety of antineoplastic drugs including DNA-damaging agents, hormone receptor antagonists, tyrosine kinase inhibitors, microtubule destabilizing agents, etc. The current review summarizes the role played by Fibronectin in mediating drug resistance against diverse anticancer drugs. We have also discussed how the aberrant expression of Fibronectin drives the oncogenic signaling pathways ultimately leading to drug resistance through the inhibition of apoptosis, promotion of cancer cell growth and proliferation.
Collapse
Affiliation(s)
- Faizah Farooq
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Asif Amin
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Umer Majeed Wani
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Asif Lone
- Department of Biochemistry, Deshbandu College, University of Delhi, Delhi, India
| | - Raies A Qadri
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
6
|
Hatse S, Serena M, Vulsteke C, Punie K, Neven P, Smeets A, Laenen A, Wildiers H. Impact of baseline telomere length on survival and chemotherapy related toxicity in breast cancer patients receiving (neo)adjuvant anthracycline containing chemotherapy. Transl Oncol 2022; 26:101551. [PMID: 36219936 PMCID: PMC9558049 DOI: 10.1016/j.tranon.2022.101551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/09/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
PURPOSE The aim of this study is to assess baseline mean leukocyte telomere length (TL) as a potential predictive factor for chemotherapy toxicity and a prognostic marker for long-term outcome in early breast cancer (BC) patients. METHODS 445 BC patients were selected, diagnosed between 2007 and 2010 with early BC and treated with (neo)adjuvant fluorouracil, epirubicin and cyclophosphamide (FEC) or with FEC and Docetaxel (FEC-D). RT-qPCR was performed on germline DNA samples collected at diagnosis before any treatment, to measure mean leukocyte TL. Uni- and multivariable logistic regression or Cox proportional hazard regression analyses were carried out to assess correlation between baseline TL and toxicity parameters (derived from the medical chart) or longer-term outcome. RESULTS Baseline TL correlated with age as expected (p = 0.005), but not with febrile neutropenia (n = 97), left ventricular ejection fraction >10% decrease (n = 17) nor other toxicity endpoints measured (all p > 0.05). TL was neither associated with overall survival, breast cancer specific survival or distant disease-free survival (all p > 0.05). CONCLUSIONS Baseline TL is not associated with chemotherapy-related toxicity nor long-term outcome in BC patients.
Collapse
Affiliation(s)
- Sigrid Hatse
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Marta Serena
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Christof Vulsteke
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Kevin Punie
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium; Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium; Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Ann Smeets
- Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Annouschka Laenen
- Interuniversity Centre for Biostatistics and Statistical Bioinformatics, Leuven, Belgium
| | - Hans Wildiers
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium; Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium; Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Azimi S, Esmaeil Lashgarian H, Ghorbanzadeh V, Moradipour A, Pirzeh L, Dariushnejad H. 5-FU and the dietary flavonoid carvacrol: a synergistic combination that induces apoptosis in MCF-7 breast cancer cells. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:253. [PMID: 36224408 DOI: 10.1007/s12032-022-01863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 10/17/2022]
Abstract
Along with the benefits of chemotherapy in the treatment of breast cancer, the side effects of these drugs along with drug resistance make their use complicated. One of the solutions to overcome this problem is the use of herbal products and combination therapy. In this research, we try to investigate the effects of carvacrol, a monoterpene flavonoid, in combination with the chemotherapy drug 5-FU. Combination index method was used for the drug-drug interactions analysis based on the Chou and Talalay method and the data from MTT assays. Apoptosis was assessed by the ELISA cell death method. P-glycoprotein expression was evaluated at the gene level by Real-time PCR. Here, we described the first experimental evidence for the existence of synergism between carvacrol and 5-FU in the in vitro model of breast cancer. MTT assay results showed combination treatment of the cells with carvacrol and 5-FU decreased 5-FU concentrations significantly. Incubation of the cells with carvacrol neutralized P-glycoprotein overexpression in qPCR assay (P ≤ 0.05). Compared with adding verapamil (a P-glycoprotein inhibitor) to 5-FU, the combination of carvacrol and 5-FU caused a further increase in the percentage of apoptotic cells when the cells were treated with both agents. Our results suggest that carvacrol can downregulate P-gp expression and combination therapy with carvacrol and 5-FU is considered a novel approach to improve the efficacy of chemotherapeutics in cancer patients with high P-glycoprotein expression.
Collapse
Affiliation(s)
- Saleh Azimi
- Razi Herbal Medicines Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamed Esmaeil Lashgarian
- Razi Herbal Medicines Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Vajihe Ghorbanzadeh
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ayat Moradipour
- Young Researchers and Elite Club, Ahar Branch, Islamic Azad University, Ahar, Iran
| | - Lale Pirzeh
- Institute for Vascular Signaling, Center for Molecular Medicine, Johann Wolfgang Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfort am Main, Germany
| | - Hassan Dariushnejad
- Razi Herbal Medicines Research Center, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
8
|
Ramos A, Sadeghi S, Tabatabaeian H. Battling Chemoresistance in Cancer: Root Causes and Strategies to Uproot Them. Int J Mol Sci 2021; 22:9451. [PMID: 34502361 PMCID: PMC8430957 DOI: 10.3390/ijms22179451] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
With nearly 10 million deaths, cancer is the leading cause of mortality worldwide. Along with major key parameters that control cancer treatment management, such as diagnosis, resistance to the classical and new chemotherapeutic reagents continues to be a significant problem. Intrinsic or acquired chemoresistance leads to cancer recurrence in many cases that eventually causes failure in the successful treatment and death of cancer patients. Various determinants, including tumor heterogeneity and tumor microenvironment, could cause chemoresistance through a diverse range of mechanisms. In this review, we summarize the key determinants and the underlying mechanisms by which chemoresistance appears. We then describe which strategies have been implemented and studied to combat such a lethal phenomenon in the management of cancer treatment, with emphasis on the need to improve the early diagnosis of cancer complemented by combination therapy.
Collapse
Affiliation(s)
- Alisha Ramos
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore;
| | - Samira Sadeghi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore 138672, Singapore
| | - Hossein Tabatabaeian
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
9
|
Alimardani M, Moghbeli M, Rastgar-Moghadam A, Shandiz FH, Abbaszadegan MR. Single nucleotide polymorphisms as the efficient prognostic markers in breast cancer. Curr Cancer Drug Targets 2021; 21:768-793. [PMID: 34036920 DOI: 10.2174/1568009621666210525151846] [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: 10/24/2020] [Revised: 03/15/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast cancer (BC) is known as the most common malignancy in women. Environmental and genetic factors are associated with BC progression. Genetic polymorphisms have been reported as important risk factors of BC prognosis and drug response. Main body: Therefore, in the present review, we have summarized all single nucleotide polymorphisms (SNPs) which have been significantly associated with drug response in BC patients around the world. We have also categorized the reported SNPs based on their related genes functions to clarify the molecular biology of drug responses in BC. CONCLUSION The majority of SNPs were reported in detoxifying enzymes, which introduced such genes as the main genetic risk factors during BC drug responses. This review paves the way for introducing a prognostic panel of SNPs for the BC patients in the world.
Collapse
Affiliation(s)
- Maliheh Alimardani
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azam Rastgar-Moghadam
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Homaei Shandiz
- Department of Radiotherapy/Oncology, Omid Hospital, Mashhad University of Medical Science, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Pleiotropic Roles of ABC Transporters in Breast Cancer. Int J Mol Sci 2021; 22:ijms22063199. [PMID: 33801148 PMCID: PMC8004140 DOI: 10.3390/ijms22063199] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/16/2022] Open
Abstract
Chemotherapeutics are the mainstay treatment for metastatic breast cancers. However, the chemotherapeutic failure caused by multidrug resistance (MDR) remains a pivotal obstacle to effective chemotherapies of breast cancer. Although in vitro evidence suggests that the overexpression of ATP-Binding Cassette (ABC) transporters confers resistance to cytotoxic and molecularly targeted chemotherapies by reducing the intracellular accumulation of active moieties, the clinical trials that target ABCB1 to reverse drug resistance have been disappointing. Nevertheless, studies indicate that ABC transporters may contribute to breast cancer development and metastasis independent of their efflux function. A broader and more clarified understanding of the functions and roles of ABC transporters in breast cancer biology will potentially contribute to stratifying patients for precision regimens and promote the development of novel therapies. Herein, we summarise the current knowledge relating to the mechanisms, functions and regulations of ABC transporters, with a focus on the roles of ABC transporters in breast cancer chemoresistance, progression and metastasis.
Collapse
|
11
|
Daniyal A, Santoso I, Gunawan NHP, Barliana MI, Abdulah R. Genetic Influences in Breast Cancer Drug Resistance. BREAST CANCER (DOVE MEDICAL PRESS) 2021; 13:59-85. [PMID: 33603458 PMCID: PMC7882715 DOI: 10.2147/bctt.s284453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022]
Abstract
Breast cancer is the most common cancer in adult women aged 20 to 50 years. The therapeutic regimens that are commonly recommended to treat breast cancer are human epidermal growth factor receptor 2 (HER2) targeted therapy, endocrine therapy, and systemic chemotherapy. The selection of pharmacotherapy is based on the characteristics of the tumor and its hormone receptor status, specifically, the presence of HER2, progesterone receptors, and estrogen receptors. Breast cancer pharmacotherapy often gives different results in various populations, which may cause therapeutic failure. Different types of congenital drug resistance in individuals can cause this. Genetic polymorphism is a factor in the occurrence of congenital drug resistance. This review explores the relationship between genetic polymorphisms and resistance to breast cancer therapy. It considers studies published from 2010 to 2020 concerning the relationship of genetic polymorphisms and breast cancer therapy. Several gene polymorphisms are found to be related to longer overall survival, worse relapse-free survival, higher pathological complete response, and increased disease-free survival in breast cancer patients. The presence of these gene polymorphisms can be considered in the treatment of breast cancer in order to shape personalized therapy to yield better results.
Collapse
Affiliation(s)
- Adhitiya Daniyal
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Ivana Santoso
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Nadira Hasna Putri Gunawan
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Melisa Intan Barliana
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Rizky Abdulah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
12
|
Yi J, Chen S, Yi P, Luo J, Fang M, Du Y, Zou L, Fan P. Pyrotinib Sensitizes 5-Fluorouracil-Resistant HER2 + Breast Cancer Cells to 5-Fluorouracil. Oncol Res 2020; 28:519-531. [PMID: 32727638 PMCID: PMC7751227 DOI: 10.3727/096504020x15960154585410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
5-Fluorouracil (5-FU) is a widely used chemotherapeutic agent for breast cancer. However, acquired chemoresistance leads to a loss of its efficacy; methods to reverse are urgently needed. Some studies have shown that pyrotinib, an ErbB receptor tyrosine kinase inhibitor, is effective against HER2+ breast cancer. However, whether pyrotinib sensitizes 5-FU-resistant breast cancer cells to 5-FU is unknown. We hypothesized that the combination of pyrotinib and 5-FU would show synergistic antitumor activity, and pyrotinib could reverse 5-FU resistance in HER2+ breast cancer cells in vitro and in vivo. Our data showed that pyrotinib inhibited the growth of 5-FU-resistant SKBR-3/FU and MDA-MB-453/FU cell lines and the parental cell lines. 5-FU remarkably suppressed the growth of SKBR-3 and MAD-MB-453 cells. However, SKBR-3/FU and MAD-MB-453/FU cells showed resistance to 5-FU. A combination of pyrotinib and 5-FU resulted in the synergistic inhibition of the growth of the 5-FU-resistant SKBR-3/FU and MDA-MB-453/FU cell lines and the parental cell lines. Pyrotinib decreased significantly the IC50 values of 5-FU and the thymidylate synthase (TS) mRNA expression levels in the 5-FU-resistant SKBR-3/FU and MDA-MB-453/FU cell lines and the parental cell lines and increased significantly the intracellular concentration of 5-FU in SKBR-3/FU and MDA-MB-453/FU cells. In addition, pyrotinib reduced the ABCG2 mRNA and protein expression levels in SKBR-3/FU and MDA-MB-453/FU cells and downregulated the protein expression levels of pAKT, pHER2, and pHER4 in all four cell lines. After TS or ABCG2 in 5-FU-resistant breast cancer cells was knocked down, the sensitivity of SKBR-3/FU and MDA-MB-453/FU cells to 5-FU was restored. Moreover, in vivo experiments demonstrated that pyrotinib in combination with 5-FU more effectively inhibited SKBR-3/FU tumor growth than either pyrotinib or 5-FU alone. In conclusion, our findings suggest that pyrotinib could restore sensitivity of 5-FU-resistant HER2+ breast cancer cells to 5-FU through downregulating the expression levels of TS and ABCG2.
Collapse
Affiliation(s)
- Jianing Yi
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| | - Shuai Chen
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| | - Pingyong Yi
- Department of Oncology, Changsha Kexin Cancer HospitalChangshaP. R. China
| | - Jinlin Luo
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| | - Meng Fang
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| | - Yang Du
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| | - Lianhong Zou
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| | - Peizhi Fan
- Surgical Department of Breast and Thyroid Gland, The First Affiliated Hospital of Hunan Normal University/Hunan Provincial Peoples HospitalChangshaP. R. China
| |
Collapse
|
13
|
Rooney SR, Shelton EL, Aka I, Shaffer CM, Clyman RI, Dagle JM, Ryckman K, Lewis TR, Reese J, Van Driest SL, Kannankeril PJ. CYP2C9*2 is associated with indomethacin treatment failure for patent ductus arteriosus. Pharmacogenomics 2019; 20:939-946. [PMID: 31486736 PMCID: PMC6817966 DOI: 10.2217/pgs-2019-0079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Aims: To identify clinical andgenetic factors associated with indomethacin treatment failure in preterm neonates with patent ductus arteriosus (PDA). Patients & Methods: This is a multicenter cohort study of 144 preterm infants (22-32 weeks gestational age) at three centers who received at least one treatment course of indomethacin for PDA. Indomethacin failure was defined as requiring subsequent surgical intervention. Results: In multivariate analysis, gestational age (AOR 0.76, 95% CI 0.60-0.96), surfactant use (AOR 9.77, 95% CI 1.15-83.26), and CYP2C9*2 (AOR 3.74; 95% CI 1.34-10.44) were each associated with indomethacin failure. Conclusion: Age, surfactant use, and CYP2C9*2 influence indomethacin treatment outcome in preterm infants with PDA. This combination of clinical and genetic factors may facilitate targeted indomethacin use for PDA.
Collapse
Affiliation(s)
- Sydney R Rooney
- Vanderbilt University School of Medicine, UCSF, Nashville, TN 37232, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
| | - Ida Aka
- Department of Pediatrics, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
| | - Christian M Shaffer
- Department of Medicine, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
| | - Ronald I Clyman
- Department of Pediatrics & Cardiovascular Research Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - John M Dagle
- Department of Pediatrics, University of Iowa, Iowa City, UMKC, IA 52242, USA
| | - Kelli Ryckman
- Department of Epidemiology, University of Iowa, Iowa City, UMKC, IA 52242, USA
| | - Tamorah R Lewis
- Department of Pediatrics, University of Missouri Kansas City School of Medicine, Kansas City, MO 64110, USA
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
| | - Sara L Van Driest
- Department of Pediatrics, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
- Department of Medicine, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
| | - Prince J Kannankeril
- Department of Pediatrics, Vanderbilt University Medical Center, UCSF, Nashville, TN 37232, USA
| |
Collapse
|
14
|
Campos-Parra AD, López-Urrutia E, Orozco Moreno LT, López-Camarillo C, Meza-Menchaca T, Figueroa González G, Bustamante Montes LP, Pérez-Plasencia C. Long Non-Coding RNAs as New Master Regulators of Resistance to Systemic Treatments in Breast Cancer. Int J Mol Sci 2018; 19:2711. [PMID: 30208633 PMCID: PMC6164317 DOI: 10.3390/ijms19092711] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/28/2022] Open
Abstract
Predicting response to systemic treatments in breast cancer (BC) patients is an urgent, yet still unattained health aim. Easily detectable molecules such as long non-coding RNAs (lncRNAs) are the ideal biomarkers when they act as master regulators of many resistance mechanisms, or of mechanisms that are common to more than one treatment. These kinds of markers are pivotal in quasi-personalized treatment selection, and consequently, in improvement of outcome prediction. In order to provide a better approach to understanding development of disease and resistance to treatments, we reviewed current literature searching for lncRNA-associated systemic BC treatments including endocrine therapies, aromatase inhibitors, selective estrogen receptor modulators (SERMs), trastuzumab, paclitaxel, docetaxel, 5-fluorouracil (5-FU), anthracyclines, and cisplatin. We found that the engagement of lncRNAs in resistance is well described, and that lncRNAs such as urotelial carcinoma-associated 1 (UCA1) and regulator of reprogramming (ROR) are indeed involved in multiple resistance mechanisms, which offers tantalizing perspectives for wide usage of lncRNAs as treatment resistance biomarkers. Thus, we propose this work as the foundation for a wide landscape of functions and mechanisms that link more lncRNAs to resistance to current and new treatments in years of research to come.
Collapse
Affiliation(s)
- Alma D Campos-Parra
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, Tlalpan, C.P. 14080 Ciudad de México, Mexico.
| | - Eduardo López-Urrutia
- Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de México (UNAM), Av. De Los Barrios 1. Col. Los Reyes Iztacala, C.P. 54090 Tlalnepantla, Mexico.
| | - Luz Tonantzin Orozco Moreno
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, Tlalpan, C.P. 14080 Ciudad de México, Mexico.
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290, Del Valle Sur, Benito Juárez, C.P. 03100 Ciudad de México, Mexico.
| | - Thuluz Meza-Menchaca
- Laboratorio de Genómica Humana, Facultad de Medicina, Universidad Veracruzana (UV), Médicos, Unidad del Bosque, Xalapa, C.P. 91010 Veracruz, Mexico.
| | - Gabriela Figueroa González
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, Tlalpan, C.P. 14080 Ciudad de México, Mexico.
| | - Lilia P Bustamante Montes
- Decanato. Ciencias de la salud. Universidad Autónoma de Guadalajara. Av. Patria 1201, Col. Lomas del Valle, C.P. 45129 Zapopan, Mexico.
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, Tlalpan, C.P. 14080 Ciudad de México, Mexico.
- Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de México (UNAM), Av. De Los Barrios 1. Col. Los Reyes Iztacala, C.P. 54090 Tlalnepantla, Mexico.
| |
Collapse
|
15
|
Lewis TR, Shelton EL, Van Driest SL, Kannankeril PJ, Reese J. Genetics of the patent ductus arteriosus (PDA) and pharmacogenetics of PDA treatment. Semin Fetal Neonatal Med 2018; 23:232-238. [PMID: 29510900 PMCID: PMC6098727 DOI: 10.1016/j.siny.2018.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patent ductus arteriosus (PDA) is a frequent, complex, and difficult to treat clinical syndrome among preterm infants in the neonatal intensive care unit. In addition to known clinical risk factors, there are emerging data about genetic predisposition to PDA in both animal and human models. Clinical response and toxicity from drugs used to treat PDA are highly variable. Developmental and genetic aspects of pharmacokinetics and pharmacodynamics influence exposure and response to pharmacologic therapies. Given the variable efficacy and toxicity of known drug therapies, novel therapeutic targets for PDA treatment offer the promise of precision medicine. This review addresses the known genetic contributions to prolonged ductal patency, variability in response to drug therapy for PDA, and potential novel drug targets for future PDA treatment discovery.
Collapse
Affiliation(s)
- Tamorah R Lewis
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, University of Missouri, Kansas City School of Medicine, Kansas City, MO, USA
| | - Elaine L Shelton
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara L Van Driest
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Prince J Kannankeril
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeff Reese
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
16
|
Megías-Vericat JE, Montesinos P, Herrero MJ, Moscardó F, Bosó V, Rojas L, Martínez-Cuadrón D, Rodríguez-Veiga R, Sendra L, Cervera J, Poveda JL, Sanz MÁ, Aliño SF. Impact of NADPH oxidase functional polymorphisms in acute myeloid leukemia induction chemotherapy. THE PHARMACOGENOMICS JOURNAL 2018; 18:301-307. [PMID: 28485375 DOI: 10.1038/tpj.2017.19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/22/2016] [Accepted: 01/09/2017] [Indexed: 02/04/2023]
Abstract
Efficacy and toxicity of anthracycline treatment in acute myeloid leukemia (AML) is mediated by reactive oxygen species (ROS). NADPH oxidase is the major endogenous source of ROS and a key mediator of oxidative cardiac damage. The impact of NADPH oxidase polymorphisms (CYBA:rs4673, NCF4:rs1883112, RAC2:rs13058338) was evaluated in 225 adult de novo AML patients. Variant alleles of NCF4 and RAC2 were related to higher complete remission (P=0.035, P=0.016), and CYBA homozygous variant showed lower overall survival with recessive model (P=0.045). Anthracycline-induced cardiotoxicity was associated to NCF4 homozygous variant (P=0.012) and CYBA heterozygous genotype (P=0.027). Novel associations were found between variant allele of CYBA and lower lung and gastrointestinal toxicities, and a protective effect in nephrotoxicity and RAC2 homozygous variant. Moreover, RAC2 homozygous variant was related to delayed thrombocytopenia recovery. This study supports the interest of NADPH oxidase polymorphisms regarding efficacy and toxicity of AML induction therapy, in a coherent integrated manner.
Collapse
Affiliation(s)
- J E Megías-Vericat
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - P Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - M J Herrero
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Facultad de Medicina, Departamento Farmacología, Universidad de Valencia, Valencia, Spain
| | - F Moscardó
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - V Bosó
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - L Rojas
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Faculty of Medicine, Department of Internal Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - R Rodríguez-Veiga
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - L Sendra
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Facultad de Medicina, Departamento Farmacología, Universidad de Valencia, Valencia, Spain
| | - J Cervera
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - J L Poveda
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - M Á Sanz
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - S F Aliño
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Facultad de Medicina, Departamento Farmacología, Universidad de Valencia, Valencia, Spain
- Unidad de Farmacología Clínica, Área del Medicamento, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|
17
|
Genetic polymorphisms and response to 5-fluorouracil, doxorubicin and cyclophosphamide chemotherapy in breast cancer patients. Oncotarget 2018; 7:66790-66808. [PMID: 27527855 PMCID: PMC5341838 DOI: 10.18632/oncotarget.11053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 07/18/2016] [Indexed: 11/25/2022] Open
Abstract
Clinical resistance to chemotherapy is one of the major problems in breast cancer treatment. In this study we analyzed possible impact of 22 polymorphic variants on the treatment response in 324 breast cancer patients. Selected genes were involved in FAC chemotherapy drugs transport (ABCB1, ABCC2, ABCG2, SLC22A16), metabolism (CYP1B1, CYP2C19, GSTT1, GSTM1, GSTP1, TYMS, MTHFR, DPYD), drug-induced damage repair (ERCC1, ERCC2, XRCC1) and involved in regulation of DNA damage response and cell cycle control (ATM, TP53). Apart from preexisting metastases three polymorphic variants were independent prognostic high risk factors of lack of response to FAC chemotherapy. Our results showed that the response to treatment depended of the variability in genes engaged in drugs’ transport (ABCC2 c.-24C>T, ABCB1 p.Ser893Ala/Thr) and in DNA repair machinery (ERCC2 p.Lys751Gln). Furthermore, the growing number of high-risk genotypes was reflected in gradual increase in risk of the non-responsiveness to treatment- from OR 2.68 for presence of two genotypes to OR 9.93 for carriers of all three negative genotypes in the group of all patients. Similar gene-dosage effect was observed in the subgroup of TNBCs. Also, TFFS significantly shortened with the increasing number of high-risk genotypes, with median of 54.4 months for carriers of one variant, to 51.5 and 34.9 months for the carriers of two and three genotypes, respectively. Our results demonstrate that results of cancer treatment are the effect of many clinical and genetic factors. It seems that multifactorial polymorphic models could be a potentially useful tool in personalization of cancer therapies. The novelty in our model is the over representation of triple negative breast cancer (TNBC) patients among the carriers of all unfavorable polymorphic variants. This finding contributes to the elucidation of the mechanisms of drug resistance in this subgroup of breast cancer patients.
Collapse
|
18
|
ADAM12-L confers acquired 5-fluorouracil resistance in breast cancer cells. Sci Rep 2017; 7:9687. [PMID: 28852196 PMCID: PMC5575004 DOI: 10.1038/s41598-017-10468-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 08/10/2017] [Indexed: 12/17/2022] Open
Abstract
5-FU-based combinatory chemotherapeutic regimens have been routinely used for many years for the treatment of breast cancer patients. Recurrence and chemotherapeutic drug resistance are two of the most prominent factors that underpin the high mortality rates associated with most breast cancers (BC). Increasing evidence indicates that overexpression of ADAMs could correlate with cancer progression. However, the role of ADAMs in the chemoresistance of cancer cells has rarely been reported. In this study, we observed that 5-FU induces expression of the ADAM12 isoform ADAM12-L but not ADAM12-S in BC cells and in recurrent BC tissues. The overexpression of ADAM12-L in BC cells following 5-FU treatment results in the acquisition of resistance to 5-FU. ADAM12-L overexoression also resulted in increased levels of p-Akt but not p-ERK. These alterations enhanced BC cell growth and invasive abilities. Conversely, ADAM12 knockdown attenuated the levels of p-Akt and restored 5-FU sensitivity in 5-FU-resistant BC cells. ADAM12 knockdown also reduced BC cell survival and invasive abilities. These findings suggest that ADAM12-L mediates chemoresistance to 5-FU and 5-FU-induced recurrence of BC by enhancing PI3K/Akt signaling. The results of this study suggest that specific ADAM12-L inhibition could optimize 5-FU-based chemotherapy of BC, thereby preventing BC recurrence in patients.
Collapse
|
19
|
Mechanisms and Genetic Susceptibility of Chemotherapy-Induced Cardiotoxicity in Patients With Breast Cancer. Am J Ther 2017; 24:e3-e11. [PMID: 27145188 DOI: 10.1097/mjt.0000000000000453] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cardiotoxicity remains an important adverse reaction of chemotherapy used in the treatment of breast cancer, leading to increased morbidity and mortality. DATA SOURCES Anthracyclines, taxanes, and trastuzumab are the most commonly used cytotoxic drugs for the treatment of breast cancer. Cardiotoxicity may vary from asymptomatic forms to irreducible heart failure and death. AREAS OF UNCERTAINTY Susceptibility for the occurrence of chemotherapy-induced cardiotoxicity and treatment resistance is multifactorial, with interindividual variability, determined by the interaction between genetic and phenotypic factors. Implementation of pharmacogenomic findings into clinical practice might be useful, to predict cardiotoxicity and to allow appropriate therapeutic measures. RESULTS AND CONCLUSIONS This review will summarize the cellular mechanisms of chemotherapy-induced cardiotoxicity in breast cancer patients and will discuss the role of the genetic susceptibility for cardiac dysfunction.
Collapse
|
20
|
Cascorbi I, Werk AN. Advances and challenges in hereditary cancer pharmacogenetics. Expert Opin Drug Metab Toxicol 2016; 13:73-82. [DOI: 10.1080/17425255.2017.1233965] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Tulsyan S, Mittal RD, Mittal B. The effect of ABCB1 polymorphisms on the outcome of breast cancer treatment. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2016; 9:47-58. [PMID: 27175090 PMCID: PMC4854269 DOI: 10.2147/pgpm.s86672] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The ABCB1 gene encodes a permeability glycoprotein, which is one of the most extensively studied human adenosine-triphosphate (ATP)-dependent efflux transporters. Permeability glycoprotein is expressed in the apical membranes of tissues such as intestine, liver, blood–brain barrier, kidney, placenta, and testis and contributes to intracellular drug disposition. It is also highly expressed in tumor cells conferring drug resistance, which is one of the major problems in the efficacy of cancer chemotherapy treatment. ABCB1 is highly polymorphic, and three well-known single-nucleotide polymorphisms such as 1236C>T, 2677G>T/A, and 3435C>T have been found to be associated with altered messenger RNA levels, protein folding, and drug pharmacokinetics. Many association studies and meta-analyses have demonstrated the clinical impact of ABCB1 polymorphisms in breast cancer treatment outcomes with respect to therapeutic response, chemotoxicity, and overall survival. Therefore, the aim of this review was to evaluate the effects of ABCB1 polymorphisms on the outcome of breast cancer treatment which, in future, would be important for tailoring individualized anticancer therapy.
Collapse
Affiliation(s)
- Sonam Tulsyan
- Department of Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rama Devi Mittal
- Department of Urology and Renal Transplant, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Balraj Mittal
- Department of Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
22
|
Vulsteke C, Pfeil AM, Maggen C, Schwenkglenks M, Pettengell R, Szucs TD, Lambrechts D, Dieudonné AS, Hatse S, Neven P, Paridaens R, Wildiers H. Clinical and genetic risk factors for epirubicin-induced cardiac toxicity in early breast cancer patients. Breast Cancer Res Treat 2015; 152:67-76. [PMID: 26017071 DOI: 10.1007/s10549-015-3437-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 12/31/2022]
Abstract
Anthracycline-induced cardiotoxicity (ACT) is a well-known serious adverse drug reaction leading to substantial morbidity. The purpose of this study was to assess ACT occurrence and clinical and genetic risk factors in early breast cancer patients. In 6 genes of interest (ABCC1, ABCC2, CYBA, NCF4, RAC2, SLC28A3), 10 single nucleotide polymorphisms (SNPs) involved in ACT were selected based on a literature search. Eight hundred and seventy-seven patients treated between 2000 and 2010 with 3-6 cycles of (neo) adjuvant 5-fluorouracil, epirubicin and cyclophosphamide (FEC) were genotyped for these SNPs using Sequenom MassARRAY. Main outcome measures were asymptomatic decrease of left ventricular ejection fraction (LVEF) > 10 % and cardiac failure grade 3-5 (CTCAE 4.0). To evaluate the impact of these 10 SNPs as well as clinical factors (age, relative dose intensity of epirubicin, left-sided radiotherapy, occurrence of febrile neutropenia, and planned and received cycles of epirubicin) on decrease of LVEF and cardiac failure, we performed uni- and multivariable logistic regression analysis. Additionally, exploratory analyses including 11 additional SNPs related to the metabolism of anthracyclines were performed. After a median follow-up of 3.62 years (range 0.40-9.60), a LVEF decline of > 10 % occurred in 153 patients (17.5 %) and cardiac failure in 16 patients (1.8 %). In multivariable analysis, six cycles of FEC compared to three cycles received and heterozygous carriers of the rs246221 T-allele in ABCC1 relative to homozygous carriers of the T-allele were significantly associated with LVEF decline of > 10 % (OR 1.3, 95 % CI 1.1-1.4, p < 0.001 and OR 1.6, 95 % CI 1.1-2.3, p = 0.02). Radiotherapy for left-sided breast cancer was associated with cardiac failure (OR 3.7, 95 % CI 1.2-11.5, p 0.026). The other 9 SNPs and clinical factors tested were not significantly associated. In our exploratory analysis, no other SNPs related to anthracycline metabolism were retained in the multivariate model for prediction of LVEF decline. ACT in breast cancer patients is related to number of received cycles of epirubicin and left-sided radiotherapy. Additional studies should be performed to independently confirm the potential association between rs246221 in ABCC1 and LVEF.
Collapse
Affiliation(s)
- Christof Vulsteke
- Integrated Cancer Center Ghent, AZ Maria Middelares, Ghent, Belgium.
| | - Alena M Pfeil
- Institute of Pharmaceutical Medicine (ECPM), University of Basel, Basel, Switzerland
| | - Charlotte Maggen
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, Louvain, Belgium
| | | | - Ruth Pettengell
- Cellular and Molecular Medicine, St. George's University of London, London, United Kingdom
| | - Thomas D Szucs
- Institute of Pharmaceutical Medicine (ECPM), University of Basel, Basel, Switzerland
| | - Diether Lambrechts
- Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB), Louvain, Belgium.,Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Louvain, Belgium
| | - Anne-Sophie Dieudonné
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, Louvain, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Herestraat 49, 3000, Louvain, Belgium
| | - Patrick Neven
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, Louvain, Belgium
| | - Robert Paridaens
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Herestraat 49, 3000, Louvain, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Herestraat 49, 3000, Louvain, Belgium
| |
Collapse
|