1
|
Bajtai E, Kiss C, Bakos É, Langó T, Lovrics A, Schád É, Tisza V, Hegedűs K, Fürjes P, Szabó Z, Tusnády GE, Szakács G, Tantos Á, Spisák S, Tóvári J, Füredi A. Therapy-induced senescence is a transient drug resistance mechanism in breast cancer. Mol Cancer 2025; 24:128. [PMID: 40312750 PMCID: PMC12044945 DOI: 10.1186/s12943-025-02310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/23/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Therapy-induced senescence (TIS) is considered a permanent cell cycle arrest following DNA-damaging treatments; however, its irreversibility has recently been challenged. Here, we demonstrate that escape from TIS is universal across breast cancer cells. Moreover, TIS provides a reversible drug resistance mechanism that ensures the survival of the population, and could contribute to relapse. METHODS TIS was induced in four different breast cancer cell line with high-dose chemotherapy and cultured until cells escaped TIS. Parental, TIS and repopulating cells were analyzed by bulk and single-cell RNA sequencing and surface proteomics. A genetically engineered mouse model of triple-negative breast cancer was used to prove why current senolytics cannot overcome TIS in tumors. RESULTS Screening the toxicity of a diverse panel of FDA-approved anticancer drugs revealed that TIS meditates resistance to half of these compounds, despite their distinct mechanism of action. Bulk and single-cell RNA sequencing, along with surface proteome analysis, showed that while parental and repopulating cells are almost identical, TIS cells are significantly different from both, highlighting their transient nature. Furthermore, investigating dozens of known drug resistance mechanisms offered no explanation for this unique drug resistance pattern. Additionally, TIS cells expressed a gene set associated with immune evasion and a potential KRAS-driven escape mechanism from TIS. CONCLUSION Our results reveal that TIS, as a transient drug resistance mechanism, could contribute to overcome the immune response and to relapse by reverting to a proliferative stage.
Collapse
Affiliation(s)
- Eszter Bajtai
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Semmelweis University Doctoral School, Budapest, 1085, Hungary
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
- National Laboratory for Drug Research and Development, Budapest, 1117, Hungary
| | - Csaba Kiss
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Doctoral School of Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Éva Bakos
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Tamás Langó
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Anna Lovrics
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Éva Schád
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Viktória Tisza
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Károly Hegedűs
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Semmelweis University Doctoral School, Budapest, 1085, Hungary
| | - Péter Fürjes
- Institute of Technical Physics and Materials Science, HUN-REN Centre of Energy Research, Budapest, 1121, Hungary
| | - Zoltán Szabó
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, 6725, Hungary
| | - Gábor E Tusnády
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Department of Bioinformatics, Semmelweis University, Budapest, 1085, Hungary
| | - Gergely Szakács
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
- Center for Cancer Research, Medical University of Vienna, Vienna, 1090, Austria
| | - Ágnes Tantos
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Sándor Spisák
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
| | - József Tóvári
- Semmelweis University Doctoral School, Budapest, 1085, Hungary.
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary.
| | - András Füredi
- Institute of Molecular Life Sciences, Center of Excellence of The Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
- Semmelweis University Doctoral School, Budapest, 1085, Hungary.
- National Laboratory for Drug Research and Development, Budapest, 1117, Hungary.
- Institute of Technical Physics and Materials Science, HUN-REN Centre of Energy Research, Budapest, 1121, Hungary.
- Physiological Controls Research Center, University Research and Innovation Center, Obuda University, Budapest, 1034, Hungary.
| |
Collapse
|
2
|
Chen L, Cen Y, Qian K, Yang W, Zhou W, Yang Y. MMP1-induced NF-κB activation promotes epithelial-mesenchymal transition and sacituzumab govitecan resistance in hormone receptor-positive breast cancer. Cell Death Dis 2025; 16:346. [PMID: 40287412 PMCID: PMC12033297 DOI: 10.1038/s41419-025-07615-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Sacituzumab govitecan (SG), a novel antibody-drug conjugate (ADC), shows promise in the treatment of breast cancer (BC); however, drug resistance limits its clinical application. Matrix metalloproteinase 1 (MMP1), which is overexpressed in many tumor types, plays a key role in tumor metastasis and drug resistance. The involvement of MMP1 in SG resistance in metastatic hormone receptor-positive (HR + ) BC has not been previously reported. In this study, we employed various in vitro and in vivo approaches to investigate the role of MMP1 in SG resistance in BC. MMP1 expression was manipulated in different BC cell lines through lentiviral transfection and small interfering RNA techniques. Key methodologies included Western blot, quantitative reverse transcription PCR, and RNA sequencing to assess marker expression and identify differentially expressed genes. Functional assays were conducted to evaluate cell viability, proliferation, invasion, and migration. In vivo, a cell-derived xenograft model in nude mice was utilized to assess tumor growth and drug response. Bioinformatics analyses further explored MMP1 expression and its clinical relevance across different cancer types. Our findings indicate that MMP1 is overexpressed by approximately 30-fold in HR + BC tissues and is associated with poorer prognosis among HR + BC patients. Furthermore, our analysis reveals that HR + BC with high MMP1 expression displays resistance to SG, supporting the hypothesis that MMP1 plays a key role in regulating ADC resistance. Mechanistic studies demonstrate that MMP1 can activate the NF-κB pathway, which subsequently influences the epithelial-mesenchymal transition, thereby contributing to SG resistance. Ultimately, our research underscores the potential of MMP1 as a therapeutic target and biomarker, facilitating personalized treatment strategies that could enhance patient outcomes in BC therapy.
Collapse
Affiliation(s)
- Letian Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinghuan Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Keyang Qian
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Wang Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenbin Zhou
- Division of Breast Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Yaping Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Zhang X, Zhuang M, Zhang H, Zhu Y, Yang J, Wu X, Yu X, Tao J, Liu X. Melatonin-mediated cGAS-STING signal in senescent macrophages promote TNBC chemotherapy resistance and drive the SASP. J Biol Chem 2025; 301:108438. [PMID: 40127867 DOI: 10.1016/j.jbc.2025.108438] [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: 10/11/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
The build-up of senescent cells in tissues is a key indicator of aging, associated with negative prognosis and therapy resistance. Despite immune dysfunction related to aging, also known as immunosenescence, is recognized as a factor in this process, the exact mechanisms are still unclear. In this study, we reported that melatonin deficiency accelerated macrophage senescence in triple-negative breast cancer, whereas melatonin could defend macrophages against senescence through the Nfatc1-Trim26-cgas-Sting pathway. Mechanistically, melatonin enhanced the nuclear translocation of Nfatc1 and elevated Trim26 transcription levels. Trim26, functioning as an E3 ligase, ubiquitinates cgas, thereby inhibiting the activation of the cgas-Sing pathway and consequently preventing cell senescence. Conversely, melatonin deficiency induced cgas-Sting pathway activation to promote macrophage aging. Our results show that melatonin inhibited macrophage senescence and improved chemotherapy responsiveness, with further enhancement when combined with the cgas inhibitor (G150). Overall, our findings indicated that melatonin protects macrophages from immunosenescence, suggesting its therapeutic potential for enhancing chemotherapy response.
Collapse
Affiliation(s)
- Xiaoqiang Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Minyu Zhuang
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Hongfei Zhang
- Department of Ultrasound in Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yanhui Zhu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Junzhe Yang
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xian Wu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xiafei Yu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China.
| | - Jing Tao
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiaoan Liu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
4
|
Khalizieva A, Moser SC, Bouwman P, Jonkers J. BRCA1 and BRCA2: from cancer susceptibility to synthetic lethality. Genes Dev 2025; 39:86-108. [PMID: 39510841 PMCID: PMC11789497 DOI: 10.1101/gad.352083.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The discovery of BRCA1 and BRCA2 as tumor susceptibility genes and their role in genome maintenance has transformed our understanding of hereditary breast and ovarian cancer. This review traces the evolution of BRCA1/2 research over the past 30 years, highlighting key discoveries in the field and their contributions to tumor development. Additionally, we discuss current preventive measures for BRCA1/2 mutation carriers and targeted treatment options based on the concept of synthetic lethality. Finally, we explore the challenges of acquired therapy resistance and discuss potential alternative avenues for targeting BRCA1/2 mutant tumors.
Collapse
Affiliation(s)
- Anna Khalizieva
- Division of Molecular Pathology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Division of Cell Systems and Drug Safety, Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sarah C Moser
- Division of Molecular Pathology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Peter Bouwman
- Division of Cell Systems and Drug Safety, Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| |
Collapse
|
5
|
Duan Y, Deng M, Liu B, Meng X, Liao J, Qiu Y, Wu Z, Lin J, Dong Y, Duan Y, Sun Y. Mitochondria targeted drug delivery system overcoming drug resistance in intrahepatic cholangiocarcinoma by reprogramming lipid metabolism. Biomaterials 2024; 309:122609. [PMID: 38754290 DOI: 10.1016/j.biomaterials.2024.122609] [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: 04/08/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The challenge of drug resistance in intrahepatic cholangiocarcinoma (ICC) is intricately linked with lipid metabolism reprogramming. The hepatic lipase (HL) and the membrane receptor CD36 are overexpressed in BGJ398-resistant ICC cells, while they are essential for lipid uptake, further enhancing lipid utilization in ICC. Herein, a metal-organic framework-based drug delivery system (OB@D-pMOF/CaP-AC, DDS), has been developed. The specifically designed DDS exhibits a successive targeting property, enabling it to precisely target ICC cells and their mitochondria. By specifically targeting the mitochondria, DDS produces reactive oxygen species (ROS) through its sonodynamic therapy effect, achieving a more potent reduction in ATP levels compared to non-targeted approaches, through the impairment of mitochondrial function. Additionally, the DDS strategically minimizes lipid uptake through the incorporation of the anti-HL drug, Orlistat, and anti-CD36 monoclonal antibody, reducing lipid-derived energy production. This dual-action strategy on both mitochondria and lipids can hinder energy utilization to restore drug sensitivity to BGJ398 in ICC. Moreover, an orthotopic mice model of drug-resistant ICC was developed, which serves as an exacting platform for evaluating the multifunction of designed DDS. Upon in vivo experiments with this model, the DDS demonstrated exceptional capabilities in suppressing tumor growth, reprogramming lipid metabolism and improving immune response, thereby overcoming drug resistance. These findings underscore the mitochondria-targeted DDS as a promising and innovative solution in ICC drug resistance.
Collapse
Affiliation(s)
- Yi Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Mengqiong Deng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Bin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xianwei Meng
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jinghan Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yijie Qiu
- Department of Ultrasound, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092, Shanghai, China
| | - Zhihua Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Jiangtao Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yi Dong
- Department of Ultrasound, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092, Shanghai, China.
| | - Yourong Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Ying Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Sarwar S, Morozov VM, Newcomb MA, Yan B, Brant JO, Opavsky R, Guryanova OA, Ishov AM. Overcoming ABCB1 mediated multidrug resistance in castration resistant prostate cancer. Cell Death Dis 2024; 15:558. [PMID: 39090086 PMCID: PMC11294535 DOI: 10.1038/s41419-024-06949-3] [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: 04/08/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. PCa that relapses after hormonal therapies, referred to as castration resistant PCa (CRPC), often presents with metastases (mCRPC) that are the major cause of mortality. The few available therapies for mCRPC patients include taxanes docetaxel (DTX) and cabazitaxel (CBZ). However, development of resistance limits their clinical use. Mechanistically, resistance arises through upregulation of multidrug resistance (MDR) proteins such as MDR1/ABCB1, making ABCB1 an attractive therapeutic target. Yet, ABCB1 inhibitors failed to be clinically useful due to low specificity and toxicity issues. To study taxanes resistance, we produced CBZ resistant C4-2B cells (RC4-2B) and documented resistance to both CBZ and DTX in cell culture and in 3D prostaspheres settings. RNAseq identified increased expression of ABCB1 in RC4-2B, that was confirmed by immunoblotting and immunofluorescent analysis. ABCB1-specific inhibitor elacridar reversed CBZ and DTX resistance in RC4-2B cells, confirming ABCB1-mediated resistance mechanism. In a cell-based screen using a curated library of cytotoxic drugs, we found that DNA damaging compounds Camptothecin (CPT) and Cytarabine (Ara-C) overcame resistance as seen by similar cytotoxicity in parental C4-2B and resistant RC4-2B. Further, these compounds were cytotoxic to multiple PC cells resistant to taxanes with high ABCB1 expression and, therefore, can be used to conquer the acquired resistance to taxanes in PCa. Finally, inhibition of cyclin-dependent kinases 4/6 (CDK4/6) with small molecule inhibitors (CDK4/6i) potentiated cytotoxic effect of CPT or Ara-C in both parental and resistant cells. Overall, our findings indicate that DNA damaging agents CPT and Ara-C alone or in combination with CDK4/6i can be suggested as a new treatment regimen in CRPC patients, including those that are resistant to taxanes.
Collapse
Affiliation(s)
- Sadia Sarwar
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Viacheslav M Morozov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Mallory A Newcomb
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Bowen Yan
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jason O Brant
- Department of Biostatistics, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Rene Opavsky
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Olga A Guryanova
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
- University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Alexander M Ishov
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA.
- University of Florida Health Cancer Center, Gainesville, FL, USA.
| |
Collapse
|
7
|
Lumpp T, Stößer S, Fischer F, Hartwig A, Köberle B. Role of Epigenetics for the Efficacy of Cisplatin. Int J Mol Sci 2024; 25:1130. [PMID: 38256203 PMCID: PMC10816946 DOI: 10.3390/ijms25021130] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The clinical utility of the chemotherapeutic agent cisplatin is restricted by cancer drug resistance, which is either intrinsic to the tumor or acquired during therapy. Epigenetics is increasingly recognized as a factor contributing to cisplatin resistance and hence influences drug efficacy and clinical outcomes. In particular, epigenetics regulates gene expression without changing the DNA sequence. Common types of epigenetic modifications linked to chemoresistance are DNA methylation, histone modification, and non-coding RNAs. This review provides an overview of the current findings of various epigenetic modifications related to cisplatin efficacy in cell lines in vitro and in clinical tumor samples. Furthermore, it discusses whether epigenetic alterations might be used as predictors of the platinum agent response in order to prevent avoidable side effects in patients with resistant malignancies. In addition, epigenetic targeting therapies are described as a possible strategy to render cancer cells more susceptible to platinum drugs.
Collapse
Affiliation(s)
| | | | | | | | - Beate Köberle
- Department Food Chemistry and Toxicology, Institute of Applied Biosciences, Karlsruhe Institute of Technology, Adenauerring 20a, 76131 Karlsruhe, Germany; (T.L.); (S.S.); (F.F.); (A.H.)
| |
Collapse
|
8
|
Szebényi K, Füredi A, Bajtai E, Sama SN, Csiszar A, Gombos B, Szabó P, Grusch M, Szakács G. Effective targeting of breast cancer by the inhibition of P-glycoprotein mediated removal of toxic lipid peroxidation byproducts from drug tolerant persister cells. Drug Resist Updat 2023; 71:101007. [PMID: 37741091 DOI: 10.1016/j.drup.2023.101007] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
Therapy resistance has long been considered to occur through the selection of pre-existing clones equipped to survive and quickly regrow, or through the acquisition of mutations during chemotherapy. Here we show that following in vitro treatment by chemotherapy, epithelial breast cancer cells adopt a transient drug tolerant phenotype characterized by cell cycle arrest, epithelial-to-mesenchymal transition (EMT) and the reversible upregulation of the multidrug resistance (MDR) efflux transporter P-glycoprotein (P-gp). The drug tolerant persister (DTP) state is reversible, as cells eventually resume proliferation, giving rise to a cell population resembling the initial, drug-naïve cell lines. However, recovery after doxorubicin treatment is almost completely eliminated when DTP cells are cultured in the presence of the P-gp inhibitor Tariquidar. Mechanistically, P-gp contributes to the survival of DTP cells by removing reactive oxygen species-induced lipid peroxidation products resulting from doxorubicin exposure. In vivo, prolonged administration of Tariquidar during doxorubicin treatment holidays resulted in a significant increase of the overall survival of Brca1-/-;p53-/- mammary tumor bearing mice. These results indicate that prolonged administration of a P-gp inhibitor during drug holidays would likely benefit patients without the risk of aggravated side effects related to the concomitantly administered toxic chemotherapy. Effective targeting of DTPs through the inhibition of P-glycoprotein may result in a paradigm shift, changing the focus from countering drug resistance mechanisms to preventing or delaying therapy resistance.
Collapse
Affiliation(s)
- Kornélia Szebényi
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria; Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary.
| | - András Füredi
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria; Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Institute of Technical Physics and Materials Science, Centre of Energy Research, Budapest, Hungary
| | - Eszter Bajtai
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria; Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Doctoral School of Pathological Sciences, Semmelweis University, Budapest, Hungary
| | - Sai Nagender Sama
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Agnes Csiszar
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Balázs Gombos
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Doctoral School of Molecular Medicine, Semmelweis University, Budapest, Hungary
| | - Pál Szabó
- Centre for Structural Study, Research Centre for Natural Sciences, Budapest, Hungary
| | - Michael Grusch
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Gergely Szakács
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria; Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary.
| |
Collapse
|
9
|
Uceda-Castro R, Margarido AS, Song JY, de Gooijer MC, Messal HA, Chambers CR, Nobis M, Çitirikkaya CH, Hahn K, Seinstra D, Herrmann D, Timpson P, Wesseling P, van Tellingen O, Vennin C, van Rheenen J. BCRP drives intrinsic chemoresistance in chemotherapy-naïve breast cancer brain metastasis. SCIENCE ADVANCES 2023; 9:eabp9530. [PMID: 37851804 PMCID: PMC10584345 DOI: 10.1126/sciadv.abp9530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 09/14/2023] [Indexed: 10/20/2023]
Abstract
Although initially successful, treatments with chemotherapy often fail because of the recurrence of chemoresistant metastases. Since these tumors develop after treatment, resistance is generally thought to occur in response to chemotherapy. However, alternative mechanisms of intrinsic chemoresistance in the chemotherapy-naïve setting may exist but remain poorly understood. Here, we study drug-naïve murine breast cancer brain metastases (BCBMs) to identify how cancer cells growing in a secondary site can acquire intrinsic chemoresistance without cytotoxic agent exposure. We demonstrate that drug-naïve murine breast cancer cells that form cancer lesions in the brain undergo vascular mimicry and concomitantly express the adenosine 5'-triphosphate-binding cassette transporter breast cancer resistance protein (BCRP), a common marker of brain endothelial cells. We reveal that expression of BCRP by the BCBM tumor cells protects them against doxorubicin and topotecan. We conclude that BCRP overexpression can cause intrinsic chemoresistance in cancer cells growing in metastatic sites without prior chemotherapy exposure.
Collapse
Affiliation(s)
- Rebeca Uceda-Castro
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Andreia S. Margarido
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Mark C. de Gooijer
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| | - Hendrik A. Messal
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Cecilia R. Chambers
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Max Nobis
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Ceren H. Çitirikkaya
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Kerstin Hahn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Danielle Seinstra
- Department of Pathology, Amsterdam University Medical Centers/VUmc and Brain Tumor Center Amsterdam, Amsterdam, Netherlands
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers/VUmc and Brain Tumor Center Amsterdam, Amsterdam, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Claire Vennin
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
10
|
Beneyton A, Nonfoux L, Gagné JP, Rodrigue A, Kothari C, Atalay N, Hendzel M, Poirier G, Masson JY. The dynamic process of covalent and non-covalent PARylation in the maintenance of genome integrity: a focus on PARP inhibitors. NAR Cancer 2023; 5:zcad043. [PMID: 37609662 PMCID: PMC10440794 DOI: 10.1093/narcan/zcad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023] Open
Abstract
Poly(ADP-ribosylation) (PARylation) by poly(ADP-ribose) polymerases (PARPs) is a highly regulated process that consists of the covalent addition of polymers of ADP-ribose (PAR) through post-translational modifications of substrate proteins or non-covalent interactions with PAR via PAR binding domains and motifs, thereby reprogramming their functions. This modification is particularly known for its central role in the maintenance of genomic stability. However, how genomic integrity is controlled by an intricate interplay of covalent PARylation and non-covalent PAR binding remains largely unknown. Of importance, PARylation has caught recent attention for providing a mechanistic basis of synthetic lethality involving PARP inhibitors (PARPi), most notably in homologous recombination (HR)-deficient breast and ovarian tumors. The molecular mechanisms responsible for the anti-cancer effect of PARPi are thought to implicate both catalytic inhibition and trapping of PARP enzymes on DNA. However, the relative contribution of each on tumor-specific cytotoxicity is still unclear. It is paramount to understand these PAR-dependent mechanisms, given that resistance to PARPi is a challenge in the clinic. Deciphering the complex interplay between covalent PARylation and non-covalent PAR binding and defining how PARP trapping and non-trapping events contribute to PARPi anti-tumour activity is essential for developing improved therapeutic strategies. With this perspective, we review the current understanding of PARylation biology in the context of the DNA damage response (DDR) and the mechanisms underlying PARPi activity and resistance.
Collapse
Affiliation(s)
- Adèle Beneyton
- CHU de Québec Research Center, HDQ Pavilion, Oncology Division, Laval University Cancer Research Center, 9 McMahon, Québec City, QC G1R 3S3, Canada
| | - Louis Nonfoux
- CHU de Québec Research Center, HDQ Pavilion, Oncology Division, Laval University Cancer Research Center, 9 McMahon, Québec City, QC G1R 3S3, Canada
- CHU de Québec Research Center, CHUL Pavilion, Oncology Division, Laval University Cancer Research Center, 2705 Boulevard Laurier, Québec City, QC G1V 4G2, Canada
| | - Jean-Philippe Gagné
- CHU de Québec Research Center, CHUL Pavilion, Oncology Division, Laval University Cancer Research Center, 2705 Boulevard Laurier, Québec City, QC G1V 4G2, Canada
| | - Amélie Rodrigue
- CHU de Québec Research Center, HDQ Pavilion, Oncology Division, Laval University Cancer Research Center, 9 McMahon, Québec City, QC G1R 3S3, Canada
| | - Charu Kothari
- CHU de Québec Research Center, CHUL Pavilion, Oncology Division, Laval University Cancer Research Center, 2705 Boulevard Laurier, Québec City, QC G1V 4G2, Canada
| | - Nurgul Atalay
- CHU de Québec Research Center, HDQ Pavilion, Oncology Division, Laval University Cancer Research Center, 9 McMahon, Québec City, QC G1R 3S3, Canada
- CHU de Québec Research Center, CHUL Pavilion, Oncology Division, Laval University Cancer Research Center, 2705 Boulevard Laurier, Québec City, QC G1V 4G2, Canada
| | - Michael J Hendzel
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AlbertaT6G 1Z2, Canada
| | - Guy G Poirier
- CHU de Québec Research Center, CHUL Pavilion, Oncology Division, Laval University Cancer Research Center, 2705 Boulevard Laurier, Québec City, QC G1V 4G2, Canada
| | - Jean-Yves Masson
- CHU de Québec Research Center, HDQ Pavilion, Oncology Division, Laval University Cancer Research Center, 9 McMahon, Québec City, QC G1R 3S3, Canada
| |
Collapse
|
11
|
Gottesman MM, Robey RW, Ambudkar SV. New mechanisms of multidrug resistance: an introduction to the Cancer Drug Resistance special collection. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:590-595. [PMID: 37842242 PMCID: PMC10571052 DOI: 10.20517/cdr.2023.86] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 10/17/2023]
Abstract
Cancer Drug Resistance publishes contributions to understanding the biology and consequences of mechanisms that interfere with successful treatment of cancer. Since virtually all patients who die of metastatic cancer have multidrug-resistant tumors, improved treatment will require an understanding of the mechanisms of resistance to design therapies that circumvent these mechanisms, exploit these mechanisms, or inactivate these multidrug resistance mechanisms. One example of a resistance mechanism is the expression of ATP-binding cassette efflux pumps, but unfortunately, inhibition of these transporters has not proved to be the solution to overcome multidrug resistance in cancer. Other mechanisms that confer multidrug resistance, and the confluence of multiple different mechanisms (multifactorial multidrug resistance) have been identified, and it is the goal of this Special Collection to expand this catalog of potential multidrug resistance mechanisms, to explore novel ways to overcome resistance, and to present thoughtful reviews on the problem of multidrug resistance in cancer.
Collapse
Affiliation(s)
- Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
12
|
Gómez-Vecino A, Corchado-Cobos R, Blanco-Gómez A, García-Sancha N, Castillo-Lluva S, Martín-García A, Mendiburu-Eliçabe M, Prieto C, Ruiz-Pinto S, Pita G, Velasco-Ruiz A, Patino-Alonso C, Galindo-Villardón P, Vera-Pedrosa ML, Jalife J, Mao JH, Macías de Plasencia G, Castellanos-Martín A, Sáez-Freire MDM, Fraile-Martín S, Rodrigues-Teixeira T, García-Macías C, Galvis-Jiménez JM, García-Sánchez A, Isidoro-García M, Fuentes M, García-Cenador MB, García-Criado FJ, García-Hernández JL, Hernández-García MÁ, Cruz-Hernández JJ, Rodríguez-Sánchez CA, García-Sancho AM, Pérez-López E, Pérez-Martínez A, Gutiérrez-Larraya F, Cartón AJ, García-Sáenz JÁ, Patiño-García A, Martín M, Alonso-Gordoa T, Vulsteke C, Croes L, Hatse S, Van Brussel T, Lambrechts D, Wildiers H, Chang H, Holgado-Madruga M, González-Neira A, Sánchez PL, Pérez Losada J. Intermediate Molecular Phenotypes to Identify Genetic Markers of Anthracycline-Induced Cardiotoxicity Risk. Cells 2023; 12:1956. [PMID: 37566035 PMCID: PMC10417374 DOI: 10.3390/cells12151956] [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: 04/17/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Cardiotoxicity due to anthracyclines (CDA) affects cancer patients, but we cannot predict who may suffer from this complication. CDA is a complex trait with a polygenic component that is mainly unidentified. We propose that levels of intermediate molecular phenotypes (IMPs) in the myocardium associated with histopathological damage could explain CDA susceptibility, so variants of genes encoding these IMPs could identify patients susceptible to this complication. Thus, a genetically heterogeneous cohort of mice (n = 165) generated by backcrossing were treated with doxorubicin and docetaxel. We quantified heart fibrosis using an Ariol slide scanner and intramyocardial levels of IMPs using multiplex bead arrays and QPCR. We identified quantitative trait loci linked to IMPs (ipQTLs) and cdaQTLs via linkage analysis. In three cancer patient cohorts, CDA was quantified using echocardiography or Cardiac Magnetic Resonance. CDA behaves as a complex trait in the mouse cohort. IMP levels in the myocardium were associated with CDA. ipQTLs integrated into genetic models with cdaQTLs account for more CDA phenotypic variation than that explained by cda-QTLs alone. Allelic forms of genes encoding IMPs associated with CDA in mice, including AKT1, MAPK14, MAPK8, STAT3, CAS3, and TP53, are genetic determinants of CDA in patients. Two genetic risk scores for pediatric patients (n = 71) and women with breast cancer (n = 420) were generated using machine-learning Least Absolute Shrinkage and Selection Operator (LASSO) regression. Thus, IMPs associated with heart damage identify genetic markers of CDA risk, thereby allowing more personalized patient management.
Collapse
Affiliation(s)
- Aurora Gómez-Vecino
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - Adrián Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, 28040 Madrid, Spain;
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 24040 Madrid, Spain
| | - Ana Martín-García
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Servicio de Cardiología, Hospital Universitario de Salamanca, Universidad de Salamanca (CIBER.CV), 37007 Salamanca, Spain
| | - Marina Mendiburu-Eliçabe
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - Carlos Prieto
- Servicio de Bioinformática, Nucleus, Universidad de Salamanca, 37007 Salamanca, Spain;
| | - Sara Ruiz-Pinto
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (S.R.-P.); (G.P.); (A.V.-R.)
| | - Guillermo Pita
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (S.R.-P.); (G.P.); (A.V.-R.)
| | - Alejandro Velasco-Ruiz
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (S.R.-P.); (G.P.); (A.V.-R.)
| | - Carmen Patino-Alonso
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Estadística, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Purificación Galindo-Villardón
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Estadística, Universidad de Salamanca, 37007 Salamanca, Spain
- Escuela Superior Politécnica del Litoral, ESPOL, Centro de Estudios e Investigaciones Estadísticas, Campus Gustavo Galindo, Km. 30.5 Via Perimetral, Guayaquil P.O. Box 09-01-5863, Ecuador
| | | | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, 28029 Madrid, Spain; (M.L.V.-P.); (J.J.)
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA;
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 92720, USA
| | - Guillermo Macías de Plasencia
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Servicio de Cardiología, Hospital Universitario de Salamanca, Universidad de Salamanca (CIBER.CV), 37007 Salamanca, Spain
| | - Andrés Castellanos-Martín
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - María del Mar Sáez-Freire
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | - Susana Fraile-Martín
- Servicio de Patología Molecular Comparada, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca, 37007 Salamanca, Spain; (S.F.-M.); (T.R.-T.); (C.G.-M.)
| | - Telmo Rodrigues-Teixeira
- Servicio de Patología Molecular Comparada, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca, 37007 Salamanca, Spain; (S.F.-M.); (T.R.-T.); (C.G.-M.)
| | - Carmen García-Macías
- Servicio de Patología Molecular Comparada, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca, 37007 Salamanca, Spain; (S.F.-M.); (T.R.-T.); (C.G.-M.)
| | - Julie Milena Galvis-Jiménez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Instituto Nacional de Cancerología de Colombia, Bogotá 111511-110411001, Colombia
| | - Asunción García-Sánchez
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Servicio de Bioquímica Clínica, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - María Isidoro-García
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Servicio de Bioquímica Clínica, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Manuel Fuentes
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
- Unidad de Proteómica y Servicio General de Citometría de Flujo, Nucleus, Universidad de Salamanca, 37007 Salamanca, Spain
| | - María Begoña García-Cenador
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Cirugía, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Francisco Javier García-Criado
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Cirugía, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Juan Luis García-Hernández
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| | | | - Juan Jesús Cruz-Hernández
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
- Servicio de Oncología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - César Augusto Rodríguez-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
- Servicio de Oncología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Alejandro Martín García-Sancho
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Servicio de Hematología, Hospital Universitario de Salamanca, CIBERONC, 37007 Salamanca, Spain;
| | - Estefanía Pérez-López
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Servicio de Hematología, Hospital Universitario de Salamanca, CIBERONC, 37007 Salamanca, Spain;
| | - Antonio Pérez-Martínez
- Department of Paediatric Hemato-Oncology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | - Federico Gutiérrez-Larraya
- Department of Paediatric Cardiology, Hospital Universitario La Paz, 28046 Madrid, Spain; (F.G.-L.); (A.J.C.)
| | - Antonio J. Cartón
- Department of Paediatric Cardiology, Hospital Universitario La Paz, 28046 Madrid, Spain; (F.G.-L.); (A.J.C.)
| | - José Ángel García-Sáenz
- Medical Oncology Service, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, 28040 Madrid, Spain;
| | - Ana Patiño-García
- Department of Pediatrics, Solid Tumor Program, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, IdisNA, 31008 Pamplona, Spain;
| | - Miguel Martín
- Department of Medicine, Gregorio Marañón Health Research Institute (IISGM), Centro de Investigación Biomédica en Red Oncológica (CIBERONC), Universidad Complutense, 28007 Madrid, Spain;
| | - Teresa Alonso-Gordoa
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
| | - Christof Vulsteke
- Department of Molecular Imaging, Pathology, Radiotherapy and Oncology (MIPRO), Center for Oncological Research (CORE), Antwerp University, 2610 Antwerp, Belgium; (C.V.); (L.C.)
- Department of Oncology, Integrated Cancer Center in Ghent, AZ Maria Middelares, 9000 Ghent, Belgium
| | - Lieselot Croes
- Department of Molecular Imaging, Pathology, Radiotherapy and Oncology (MIPRO), Center for Oncological Research (CORE), Antwerp University, 2610 Antwerp, Belgium; (C.V.); (L.C.)
- Department of Oncology, Integrated Cancer Center in Ghent, AZ Maria Middelares, 9000 Ghent, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology (LEO), Department of Oncology, Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium;
| | - Thomas Van Brussel
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium; (T.V.B.); (D.L.)
- Laboratory of Translational Genetics, Department of Human Genetics, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
| | - Diether Lambrechts
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium; (T.V.B.); (D.L.)
- Laboratory of Translational Genetics, Department of Human Genetics, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Unit, Leuven Cancer Institute, and Laboratory of Experimental Oncology (LEO), Department of Oncology, Leuven Cancer Institute and University Hospital Leuven, Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium;
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA;
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 92720, USA
| | - Marina Holgado-Madruga
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, 37007 Salamanca, Spain
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain
| | - Anna González-Neira
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (S.R.-P.); (G.P.); (A.V.-R.)
| | - Pedro L. Sánchez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Servicio de Cardiología, Hospital Universitario de Salamanca, Universidad de Salamanca (CIBER.CV), 37007 Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Jesús Pérez Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (A.G.-V.); (R.C.-C.); (A.B.-G.); (N.G.-S.); (M.M.-E.); (A.C.-M.); (M.d.M.S.-F.); (J.M.G.-J.); (M.F.); (J.L.G.-H.); (J.J.C.-H.); (C.A.R.-S.); (A.M.G.-S.); (E.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.-G.); (C.P.-A.); (P.G.-V.); (G.M.d.P.); (A.G.-S.); (M.I.-G.); (M.B.G.-C.); (F.J.G.-C.)
| |
Collapse
|
13
|
Bhin J, Paes Dias M, Gogola E, Rolfs F, Piersma SR, de Bruijn R, de Ruiter JR, van den Broek B, Duarte AA, Sol W, van der Heijden I, Andronikou C, Kaiponen TS, Bakker L, Lieftink C, Morris B, Beijersbergen RL, van de Ven M, Jimenez CR, Wessels LFA, Rottenberg S, Jonkers J. Multi-omics analysis reveals distinct non-reversion mechanisms of PARPi resistance in BRCA1- versus BRCA2-deficient mammary tumors. Cell Rep 2023; 42:112538. [PMID: 37209095 DOI: 10.1016/j.celrep.2023.112538] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/16/2023] [Accepted: 05/03/2023] [Indexed: 05/22/2023] Open
Abstract
BRCA1 and BRCA2 both function in DNA double-strand break repair by homologous recombination (HR). Due to their HR defect, BRCA1/2-deficient cancers are sensitive to poly(ADP-ribose) polymerase inhibitors (PARPis), but they eventually acquire resistance. Preclinical studies yielded several PARPi resistance mechanisms that do not involve BRCA1/2 reactivation, but their relevance in the clinic remains elusive. To investigate which BRCA1/2-independent mechanisms drive spontaneous resistance in vivo, we combine molecular profiling with functional analysis of HR of matched PARPi-naive and PARPi-resistant mouse mammary tumors harboring large intragenic deletions that prevent reactivation of BRCA1/2. We observe restoration of HR in 62% of PARPi-resistant BRCA1-deficient tumors but none in the PARPi-resistant BRCA2-deficient tumors. Moreover, we find that 53BP1 loss is the prevalent resistance mechanism in HR-proficient BRCA1-deficient tumors, whereas resistance in BRCA2-deficient tumors is mainly induced by PARG loss. Furthermore, combined multi-omics analysis identifies additional genes and pathways potentially involved in modulating PARPi response.
Collapse
Affiliation(s)
- Jinhyuk Bhin
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Department of Biomedical System Informatics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Mariana Paes Dias
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Ewa Gogola
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Frank Rolfs
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; OncoProteomics Laboratory, Department Medical Oncology, Amsterdam UMC, 1081HV Amsterdam, the Netherlands
| | - Sander R Piersma
- OncoProteomics Laboratory, Department Medical Oncology, Amsterdam UMC, 1081HV Amsterdam, the Netherlands
| | - Roebi de Bruijn
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Julian R de Ruiter
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Bram van den Broek
- Division of Cell Biology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Alexandra A Duarte
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Wendy Sol
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Ingrid van der Heijden
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Christina Andronikou
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3088 Bern, Switzerland; Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Taina S Kaiponen
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3088 Bern, Switzerland; Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Lara Bakker
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Ben Morris
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging, Preclinical Intervention Unit, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands
| | - Connie R Jimenez
- OncoProteomics Laboratory, Department Medical Oncology, Amsterdam UMC, 1081HV Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands.
| | - Sven Rottenberg
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands; Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3088 Bern, Switzerland; Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland.
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, 1066CX Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Sargazi Z, Yazdani Y, Tahavvori A, Youshanlouei HR, Alivirdiloo V, Beilankouhi EAV, Valilo M. NFR2/ABC transporter axis in drug resistance of breast cancer cells. Mol Biol Rep 2023; 50:5407-5414. [PMID: 37081307 DOI: 10.1007/s11033-023-08384-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/07/2023] [Indexed: 04/22/2023]
Abstract
Breast cancer is one of the most serious malignancies among women, accounting for about 12% of all cancers. The inherent complexity and heterogeneity of breast cancer results in failure to respond to treatment in the advanced stages of the disease. Breast cancer is caused by several genetic and environmental factors. One of the significant factors involved in the development of breast cancer is oxidative stress, which is generally regulated by nuclear factor erythroid 2-related factor 2 (NRF2). The level of NRF2 expression is low in healthy cells, which maintains the balance of the antioxidant system; however, its expression is higher in cancer cells, which have correlation characteristics such as angiogenesis, stem cell formation, drug resistance, and metastasis. Drug resistance increases with the upregulation of NRF2 expression, which contributes to cell protection. NRF2 controls this mechanism by increasing the expression of ATP-binding cassettes (ABCs). Considering the growing number of studies in this field, we aimed to investigate the relationship between NRF2 and ABCs, as well as their role in the development of drug resistance in breast cancer.
Collapse
Affiliation(s)
- Zinat Sargazi
- Department of Anatomical Sciences, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Yalda Yazdani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Tahavvori
- Department of internal medicine, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamed Rahmani Youshanlouei
- Department of internal medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Alivirdiloo
- Medical Doctor Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | | | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
15
|
Das S, Kundu M, Hassan A, Parekh A, Jena BC, Mundre S, Banerjee I, Yetirajam R, Das CK, Pradhan AK, Das SK, Emdad L, Mitra P, Fisher PB, Mandal M. A novel computational predictive biological approach distinguishes Integrin β1 as a salient biomarker for breast cancer chemoresistance. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166702. [PMID: 37044238 DOI: 10.1016/j.bbadis.2023.166702] [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: 11/09/2022] [Revised: 03/11/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023]
Abstract
Chemoresistance is a primary cause of breast cancer treatment failure, and protein-protein interactions significantly contribute to chemoresistance during different stages of breast cancer progression. In pursuit of novel biomarkers and relevant protein-protein interactions occurring during the emergence of breast cancer chemoresistance, we used a computational predictive biological (CPB) approach. CPB identified associations of adhesion molecules with proteins connected with different breast cancer proteins associated with chemoresistance. This approach identified an association of Integrin β1 (ITGB1) with chemoresistance and breast cancer stem cell markers. ITGB1 activated the Focal Adhesion Kinase (FAK) pathway promoting invasion, migration, and chemoresistance in breast cancer by upregulating Erk phosphorylation. FAK also activated Wnt/Sox2 signaling, which enhanced self-renewal in breast cancer. Activation of the FAK pathway by ITGB1 represents a novel mechanism linked to breast cancer chemoresistance, which may lead to novel therapies capable of blocking breast cancer progression by intervening in ITGB1-regulated signaling pathways.
Collapse
Affiliation(s)
- Subhayan Das
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Moumita Kundu
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Atif Hassan
- Department of Computer Science & Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Aditya Parekh
- Anant National University, Ahmedabad, Gujarat, India
| | - Bikash Ch Jena
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Swati Mundre
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Indranil Banerjee
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; School of Pharmacy, Sister Nivedita University (Techno India Group), Kolkata, West Bengal, India
| | - Rajesh Yetirajam
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Chandan K Das
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Pralay Mitra
- Department of Computer Science & Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
16
|
Cai Z, Huan ML, Zhang YW, Zhao TT, Han TY, He W, Zhou SY, Zhang BL. Tumor targeted combination therapeutic system for the effective treatment of drug resistant triple negative breast cancer. Int J Pharm 2023; 636:122821. [PMID: 36914017 DOI: 10.1016/j.ijpharm.2023.122821] [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: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023]
Abstract
Breast cancer has become the malignant tumor with the largest incidence, especially the drug resistant triple negative breast cancer (TNBC). The combination therapeutic system can play a better role in resisting drug resistant TNBC. In this study, dopamine and tumor targeted folic acid modified dopamine were synthesized as carrier materials to construct melanin-like tumor targeted combination therapeutic system. The optimized nanoparticles of CPT/Fe@PDA-FA10 with efficient loading of camptothecin and iron was achieved, which showed tumor targeted delivery ability, pH sensitive controlled release, effective photothermal conversion performance and excellent anti-tumor efficacy in vitro and in vivo. CPT/Fe@PDA-FA10 plus laser could significantly kill the drug resistant tumor cells, inhibit the growth of the orthotopic drug resistant triple negative breast cancer through apoptosis/ferroptosis/photothermal treatment, and had no significant side effects on the main tissues and organs. This strategy provided a new idea for the construction and clinical application of triple-combination therapeutic system as effective treatment for drug resistant triple negative breast cancer.
Collapse
Affiliation(s)
- Zedong Cai
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Meng-Lei Huan
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yao-Wen Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Ting-Ting Zhao
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Tian-Yan Han
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wei He
- Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China; Department of Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Si-Yuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China.
| |
Collapse
|
17
|
Gómez-Vecino A, Corchado-Cobos R, Blanco-Gómez A, García-Sancha N, Castillo-Lluva S, Martín-García A, Mendiburu-Eliçabe M, Prieto C, Ruiz-Pinto S, Pita G, Velasco-Ruiz A, Patino-Alonso C, Galindo-Villardón P, Vera-Pedrosa ML, Jalife J, Mao JH, de Plasencia GM, Castellanos-Martín A, Freire MDMS, Fraile-Martín S, Rodrigues-Teixeira T, García-Macías C, Galvis-Jiménez JM, García-Sánchez A, Isidoro-García M, Fuentes M, García-Cenador MB, García-Criado FJ, García JL, Hernández-García MÁ, Hernández JJC, Rodríguez-Sánchez CA, Martín-Ruiz A, Pérez-López E, Pérez-Martínez A, Gutiérrez-Larraya F, Cartón AJ, García-Sáenz JÁ, Patiño-García A, Martín M, Gordoa TA, Vulsteke C, Croes L, Hatse S, Brussel TV, Lambrechts D, Wildiers H, Hang C, Holgado-Madruga M, González-Neira A, Sánchez PL, Losada JP. Intermediate molecular phenotypes to identify genetic markers of anthracycline-induced cardiotoxicity risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522844. [PMID: 36712139 PMCID: PMC9881971 DOI: 10.1101/2023.01.05.522844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cardiotoxicity due to anthracyclines (CDA) affects cancer patients, but we cannot predict who may suffer from this complication. CDA is a complex disease whose polygenic component is mainly unidentified. We propose that levels of intermediate molecular phenotypes in the myocardium associated with histopathological damage could explain CDA susceptibility; so that variants of genes encoding these intermediate molecular phenotypes could identify patients susceptible to this complication. A genetically heterogeneous cohort of mice generated by backcrossing (N = 165) was treated with doxorubicin and docetaxel. Cardiac histopathological damage was measured by fibrosis and cardiomyocyte size by an Ariol slide scanner. We determine intramyocardial levels of intermediate molecular phenotypes of CDA associated with histopathological damage and quantitative trait loci (ipQTLs) linked to them. These ipQTLs seem to contribute to the missing heritability of CDA because they improve the heritability explained by QTL directly linked to CDA (cda-QTLs) through genetic models. Genes encoding these molecular subphenotypes were evaluated as genetic markers of CDA in three cancer patient cohorts (N = 517) whose cardiac damage was quantified by echocardiography or Cardiac Magnetic Resonance. Many SNPs associated with CDA were found using genetic models. LASSO multivariate regression identified two risk score models, one for pediatric cancer patients and the other for women with breast cancer. Molecular intermediate phenotypes associated with heart damage can identify genetic markers of CDA risk, thereby allowing a more personalized patient management. A similar strategy could be applied to identify genetic markers of other complex trait diseases.
Collapse
Affiliation(s)
- Aurora Gómez-Vecino
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Adrián Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, 28040, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Ana Martín-García
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Cardiología, Hospital Universitario de Salamanca, Universidad de Salamanca, and CIBER.CV, Salamanca, 37007, Spain
| | - Marina Mendiburu-Eliçabe
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Carlos Prieto
- Servicio de Bioinformática, Nucleus, Universidad de Salamanca, Salamanca, 37007, Spain
| | - Sara Ruiz-Pinto
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029, Spain
| | - Guillermo Pita
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029, Spain
| | - Alejandro Velasco-Ruiz
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029, Spain
| | - Carmen Patino-Alonso
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Estadística, Universidad de Salamanca, Salamanca, 37007, Spain; and Centro de Investigación Institucional (CII). Universidad Bernardo O’Higgins, 1497. Santiago, Chile
| | - Purificación Galindo-Villardón
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Estadística, Universidad de Salamanca, Salamanca, 37007, Spain; and Centro de Investigación Institucional (CII). Universidad Bernardo O’Higgins, 1497. Santiago, Chile
| | | | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, 28029, Spain
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Guillermo Macías de Plasencia
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Cardiología, Hospital Universitario de Salamanca, Universidad de Salamanca, and CIBER.CV, Salamanca, 37007, Spain
| | - Andrés Castellanos-Martín
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - María del Mar Sáez Freire
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | - Susana Fraile-Martín
- Servicio de Patología Molecular Comparada, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca, Salamanca, 37007, Spain
| | - Telmo Rodrigues-Teixeira
- Servicio de Patología Molecular Comparada, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca, Salamanca, 37007, Spain
| | - Carmen García-Macías
- Servicio de Patología Molecular Comparada, Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca, Salamanca, 37007, Spain
| | - Julie Milena Galvis-Jiménez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Instituto Nacional de Cancerología de Colombia, Bogotá D. C., Colombia
| | - Asunción García-Sánchez
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Bioquímica Clínica, Hospital Universitario de Salamanca, Salamanca, 37007, Spain
| | - María Isidoro-García
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Bioquímica Clínica, Hospital Universitario de Salamanca, Salamanca, 37007, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007, Spain
| | - Manuel Fuentes
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007, Spain
- Unidad de Proteómica y Servicio General de Citometría de Flujo, Nucleus, Universidad de Salamanca, 37007, Spain
| | - María Begoña García-Cenador
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Cirugía, Universidad de Salamanca. Salamanca, 37007, Spain
| | - Francisco Javier García-Criado
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Cirugía, Universidad de Salamanca. Salamanca, 37007, Spain
| | - Juan Luis García
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| | | | - Juan Jesús Cruz Hernández
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007, Spain
- Servicio de Oncología, Hospital Universitario de Salamanca, Salamanca, 37007, Spain
| | - César Augusto Rodríguez-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007, Spain
- Servicio de Oncología, Hospital Universitario de Salamanca, Salamanca, 37007, Spain
| | - Alejandro Martín-Ruiz
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, CIBERONC, Salamanca, 37007, Spain
| | - Estefanía Pérez-López
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, CIBERONC, Salamanca, 37007, Spain
| | - Antonio Pérez-Martínez
- Department of Paediatric Hemato-Oncology, Hospital Universitario La Paz, Madrid, 28046, Spain
| | | | - Antonio J. Cartón
- Department of Paediatric Hemato-Oncology, Hospital Universitario La Paz, Madrid, 28046, Spain
| | - José Ángel García-Sáenz
- Medical Oncology Service, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, 28040, Spain
| | - Ana Patiño-García
- Department of Pediatrics, University Clinic of Navarra, Solid Tumor Program, CIMA, Universidad de Navarra, IdisNA, Pamplona, 31008, Spain
| | - Miguel Martín
- Gregorio Marañón Health Research Institute (IISGM), CIBERONC, Department of Medicine, Universidad Complutense, Madrid, 28007, Spain
| | - Teresa Alonso Gordoa
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Christof Vulsteke
- Department of Molecular Imaging, Pathology, Radiotherapy and Oncology (MIPRO), Center for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
- Department of Oncology, Integrated Cancer Center in Ghent, AZ Maria Middelares, Ghent, Belgium
| | - Lieselot Croes
- Department of Molecular Imaging, Pathology, Radiotherapy and Oncology (MIPRO), Center for Oncological Research (CORE), Antwerp University, Antwerp, Belgium
- Department of Oncology, Integrated Cancer Center in Ghent, AZ Maria Middelares, Ghent, 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, Leuven, Belgium
| | - Thomas Van Brussel
- VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Diether Lambrechts
- VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Centre, University Hospitals Leuven, Leuven Cancer Institute, and Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, Leuven, Belgium
| | - Chang Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Marina Holgado-Madruga
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, 37007, Salamanca. Spain
- Instituto de Neurociencias de Castilla y León (INCyL), Salamanca, 37007, Spain
| | - Anna González-Neira
- Human Genotyping Unit-CeGen, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), 28029, Spain
| | - Pedro L Sánchez
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
- Servicio de Cardiología, Hospital Universitario de Salamanca, Universidad de Salamanca, and CIBER.CV, Salamanca, 37007, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, 37007, Spain
| | - Jesús Pérez Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, 37007, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, 37007, Spain
| |
Collapse
|
18
|
Widmer CA, Klebic I, Domanitskaya N, Decollogny M, Howald D, Siffert M, Essers P, Nowicka Z, Stokar-Regenscheit N, van de Ven M, de Korte-Grimmerink R, Galván JA, Pritchard CE, Huijbers IJ, Fendler W, Vens C, Rottenberg S. Loss of the volume-regulated anion channel components LRRC8A and LRRC8D limits platinum drug efficacy. CANCER RESEARCH COMMUNICATIONS 2022; 2:1266-1281. [PMID: 36467895 PMCID: PMC7613873 DOI: 10.1158/2767-9764.crc-22-0208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In recent years platinum (Pt) drugs have been found to be especially efficient to treat patients with cancers that lack a proper DNA damage response, e.g. due to dysfunctional BRCA1. Despite this knowledge, we are still missing helpful markers to predict Pt response in the clinic. We have previously shown that volume-regulated anion channels, containing the subunits LRRC8A and LRRC8D, promote the uptake of cisplatin and carboplatin in BRCA1-proficient cell lines. Here, we show that the loss of LRRC8A or LRRC8D significantly reduces the uptake of cis- and carboplatin in BRCA1;p53-deficient mouse mammary tumor cells. This results in reduced DNA damage and in vivo drug resistance. In contrast to Lrrc8a, the deletion of the Lrrc8d gene does not affect the viability and fertility of mice. Interestingly, Lrrc8d-/- mice tolerate a two-fold cisplatin maximum-tolerable dose. This allowed us to establish a mouse model for intensified Pt-based chemotherapy, and we found that an increased cisplatin dose eradicates BRCA1;p53-deficient tumors, whereas eradication is not possible in WT mice. Moreover, we show that decreased expression of LRRC8A/D in head and neck squamous cell carcinoma patients, who are treated with a Pt-based chemoradiotherapy, leads to decreased overall survival of the patients. In particular, high cumulative cisplatin dose treatments lost their efficacy in patients with a low LRRC8A/D expression in their cancers. Our data therefore suggest that LRRC8A and LRRC8D should be included in a prospective trial to predict the success of intensified cis- or car-boplatin-based chemotherapy.
Collapse
Affiliation(s)
- Carmen A. Widmer
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ismar Klebic
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- COMPATH, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Natalya Domanitskaya
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Morgane Decollogny
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Denise Howald
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Myriam Siffert
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Paul Essers
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Zuzanna Nowicka
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | | | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging Research (MCCA), Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renske de Korte-Grimmerink
- Mouse Clinic for Cancer and Aging Research (MCCA), Preclinical Intervention Unit, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - José A. Galván
- Translational Research Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Colin E.J. Pritchard
- Mouse Clinic for Cancer and Aging Research (MCCA), Transgenic Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ivo J. Huijbers
- Mouse Clinic for Cancer and Aging Research (MCCA), Transgenic Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Conchita Vens
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Corresponding Author: Sven Rottenberg, Institute of Animal Pathology and Bern Center for Precision Medicine, Länggassstrasse 122, Bern 3012, Switzerland. Phone: +41-(0)31-6842395; E-mail:
| |
Collapse
|
19
|
PARP-inhibition reprograms macrophages toward an anti-tumor phenotype. Cell Rep 2022; 41:111462. [PMID: 36223740 PMCID: PMC9727835 DOI: 10.1016/j.celrep.2022.111462] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 07/14/2022] [Accepted: 09/16/2022] [Indexed: 01/17/2023] Open
Abstract
Poly(ADP)ribosylation inhibitors (PARPis) are toxic to cancer cells with homologous recombination (HR) deficiency but not to HR-proficient cells in the tumor microenvironment (TME), including tumor-associated macrophages (TAMs). As TAMs can promote or inhibit tumor growth, we set out to examine the effects of PARP inhibition on TAMs in BRCA1-related breast cancer (BC). The PARPi olaparib causes reprogramming of TAMs toward higher cytotoxicity and phagocytosis. A PARPi-related surge in NAD+ increases glycolysis, blunts oxidative phosphorylation, and induces reverse mitochondrial electron transport (RET) with an increase in reactive oxygen species (ROS) and transcriptional reprogramming. This reprogramming occurs in the absence or presence of PARP1 or PARP2 and is partially recapitulated by addition of NAD derivative methyl-nicotinamide (MNA). In vivo and ex vivo, the effect of olaparib on TAMs contributes to the anti-tumor efficacy of the PARPi. In vivo blockade of the "don't-eat-me signal" with CD47 antibodies in combination with olaparib improves outcomes in a BRCA1-related BC model.
Collapse
|
20
|
Modi A, Roy D, Sharma S, Vishnoi JR, Pareek P, Elhence P, Sharma P, Purohit P. ABC transporters in breast cancer: their roles in multidrug resistance and beyond. J Drug Target 2022; 30:927-947. [PMID: 35758271 DOI: 10.1080/1061186x.2022.2091578] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
ATP-binding cassette (ABC) transporters are membrane-spanning proteins involved in cholesterol homeostasis, transport of various molecules in and out of cells and organelles, oxidative stress, immune recognition, and drug efflux. They are long implicated in the development of multidrug resistance in cancer chemotherapy. Existing clinical and molecular evidence has also linked ABC transporters with cancer pathogenesis, prognostics, and therapy. In this review, we aim to provide a comprehensive update on all ABC transporters and their roles in drug resistance in breast cancer (BC). For solid tumours such as BC, various ABC transporters are highly expressed in less differentiated subtypes and metastases. ABCA1, ABCB1 and ABCG2 are key players in BC chemoresistance. Restraining these transporters has evolved as a possible mechanism to reverse this phenomenon. Further, ABCB1 and ABCC1 are important in BC prognosis. Newer therapeutic approaches have been developed to target all these molecules to dysregulate their effect, reduce cell viability, induce apoptosis, and increase drug sensitivity. In the future, targeted therapy for specific genetic variations and upstream or downstream molecules can help improve patient prognosis.
Collapse
Affiliation(s)
- Anupama Modi
- Department of Biochemistry, AIIMS, Jodhpur, India
| | - Dipayan Roy
- Department of Biochemistry, AIIMS, Jodhpur, India.,Indian Institute of Technology (IIT) Madras, Chennai, India
| | | | | | - Puneet Pareek
- Department of Radiation Oncology, AIIMS, Jodhpur, India
| | - Poonam Elhence
- Department of Pathology and Laboratory Medicine, AIIMS, Jodhpur, India
| | | | | |
Collapse
|
21
|
Effective targeting of breast cancer stem cells by combined inhibition of Sam68 and Rad51. Oncogene 2022; 41:2196-2209. [PMID: 35217791 PMCID: PMC8993694 DOI: 10.1038/s41388-022-02239-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 01/07/2022] [Accepted: 02/09/2022] [Indexed: 12/17/2022]
Abstract
Breast cancer (BC) is the second cause of cancer-related deceases in the worldwide female population. Despite the successful treatment advances, 25% of BC develops resistance to current therapeutic regimens, thereby remaining a major hurdle for patient management. Current therapies, targeting the molecular events underpinning the adaptive resistance, still require effort to improve BC treatment. Using BC sphere cells (BCSphCs) as a model, here we showed that BC stem-like cells express high levels of Myc, which requires the presence of the multifunctional DNA/RNA binding protein Sam68 for the DNA-damage repair. Analysis of a cohort of BC patients displayed that Sam68 is an independent negative factor correlated with the progression of the disease. Genetic inhibition of Sam68 caused a defect in PARP-induced PAR chain synthesis upon DNA-damaging insults, resulting in cell death of TNBC cells. In contrast, BC stem-like cells were able to survive due to an upregulation of Rad51. Importantly, the inhibition of Rad51 showed synthetic lethal effect with the silencing of Sam68, hampering the cell viability of patient-derived BCSphCs and stabilizing the growth of tumor xenografts, including those TNBC carrying BRCA mutation. Moreover, the analysis of Myc, Sam68 and Rad51 expression demarcated a signature of a poor outcome in a large cohort of BC patients. Thus, our findings suggest the importance of targeting Sam68-PARP1 axis and Rad51 as potential therapeutic candidates to counteract the expansion of BC cells with an aggressive phenotype.
Collapse
|
22
|
Shah S, Rachmat R, Enyioma S, Ghose A, Revythis A, Boussios S. BRCA Mutations in Prostate Cancer: Assessment, Implications and Treatment Considerations. Int J Mol Sci 2021; 22:12628. [PMID: 34884434 PMCID: PMC8657599 DOI: 10.3390/ijms222312628] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer ranks fifth in cancer-related mortality in men worldwide. DNA damage is implicated in cancer and DNA damage response (DDR) pathways are in place against this to maintain genomic stability. Impaired DDR pathways play a role in prostate carcinogenesis and germline or somatic mutations in DDR genes have been found in both primary and metastatic prostate cancer. Among these, BRCA mutations have been found to be especially clinically relevant with a role for germline or somatic testing. Prostate cancer with DDR defects may be sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors which target proteins in a process called PARylation. Initially they were used to target BRCA-mutated tumor cells in a process of synthetic lethality. However, recent studies have found potential for PARP inhibitors in a variety of other genetic settings. In this review, we explore the mechanisms of DNA repair, potential for genomic analysis of prostate cancer and therapeutics of PARP inhibitors along with their safety profile.
Collapse
Affiliation(s)
- Sidrah Shah
- Department of Palliative Care, Guy’s and St Thomas’ Hospital, Great Maze Pond, London SE1 9RT, UK;
| | - Rachelle Rachmat
- Department of Radiology, Guy’s and St Thomas’ Hospital, Great Maze Pond, London SE1 9RT, UK;
| | - Synthia Enyioma
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
| | - Aruni Ghose
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, W Smithfield, London EC1A 7BE, UK;
- Faculty of Life Sciences & Medicine, King’s College London, London WC2R 2LS, UK
| | - Antonios Revythis
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
23
|
Mir C, Garcia-Mayea Y, Garcia L, Herrero P, Canela N, Tabernero R, Lorente J, Castellvi J, Allonca E, García-Pedrero J, Rodrigo JP, Carracedo Á, LLeonart ME. SDCBP Modulates Stemness and Chemoresistance in Head and Neck Squamous Cell Carcinoma through Src Activation. Cancers (Basel) 2021; 13:cancers13194952. [PMID: 34638436 PMCID: PMC8508472 DOI: 10.3390/cancers13194952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Drug resistance is the principal limiting factor to achieving good survival rates in patients with cancer. The identification of potential biomarkers for diagnosis and prognostic prediction, as well as the design of new molecular-targeted treatments, will be essential to improving head and neck squamous cell carcinoma (HNSCC) patient outcomes. In this sense, the sensitization of resistant cells and cancer stem cells (CSCs) represents a major challenge in cancer therapy. We conducted a proteomic study involving cisplatin-resistance and CSCs with the aim to unravel the molecular and cellular mechanisms by which tumor cells acquire resistance to chemotherapy. Syntenin-1 (SDCBP) was identified as an important protein involved in the chemoresistance and stemness of HNSCC tumors. Abstract To characterize the mechanisms that govern chemoresistance, we performed a comparative proteomic study analyzing head and neck squamous cell carcinoma (HNSCC) cells: CCL-138 (parental), CCL-138-R (cisplatin-resistant), and cancer stem cells (CSCs). Syntenin-1 (SDCBP) was upregulated in CCL-138-R cells and CSCs over parental cells. SDCBP depletion sensitized biopsy-derived and established HNSCC cell lines to cisplatin (CDDP) and reduced CSC markers, Src activation being the main SDCBP downstream target. In mice, SDCBP-depleted cells formed tumors with decreased mitosis, Ki-67 positivity, and metastasis over controls. Moreover, the fusocellular pattern of CCL-138-R cell-derived tumors reverted to a more epithelial morphology upon SDCBP silencing. Importantly, SDCBP expression was associated with Src activation, poor differentiated tumor grade, advanced tumor stage, and shorter survival rates in a series of 382 HNSCC patients. Our results reveal that SDCBP might be a promising therapeutic target for effectively eliminating CSCs and CDDP resistance.
Collapse
Affiliation(s)
- Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain; (C.M.); (Y.G.-M.); (L.G.); (J.C.)
- Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain; (C.M.); (Y.G.-M.); (L.G.); (J.C.)
| | - Laia Garcia
- Biomedical Research in Cancer Stem Cells Group, Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain; (C.M.); (Y.G.-M.); (L.G.); (J.C.)
| | - Pol Herrero
- Eurecat, Centre Tecnològic de Catalunya–Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (P.H.); (N.C.)
| | - Nuria Canela
- Eurecat, Centre Tecnològic de Catalunya–Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain; (P.H.); (N.C.)
| | - Rocío Tabernero
- Otorhinolaryngology Department, Hospital Vall d’Hebron (HUVH), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (R.T.); (J.L.)
| | - Juan Lorente
- Otorhinolaryngology Department, Hospital Vall d’Hebron (HUVH), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain; (R.T.); (J.L.)
| | - Josep Castellvi
- Biomedical Research in Cancer Stem Cells Group, Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain; (C.M.); (Y.G.-M.); (L.G.); (J.C.)
| | - Eva Allonca
- Department of Otolaryngology, Hospital Universitario Central de Asturias (HUCA), Instituto de Investigación Sanitaria del Principado de Asturias, IUOPA, University of Oviedo, 33011 Oviedo, Spain or (E.A.); (J.G.-P.); (J.P.R.)
| | - Juana García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias (HUCA), Instituto de Investigación Sanitaria del Principado de Asturias, IUOPA, University of Oviedo, 33011 Oviedo, Spain or (E.A.); (J.G.-P.); (J.P.R.)
- Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain
| | - Juan Pablo Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias (HUCA), Instituto de Investigación Sanitaria del Principado de Asturias, IUOPA, University of Oviedo, 33011 Oviedo, Spain or (E.A.); (J.G.-P.); (J.P.R.)
- Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain
| | - Ángel Carracedo
- Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complejo Hospitalario Universitario de Santiago, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Matilde Esther LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain; (C.M.); (Y.G.-M.); (L.G.); (J.C.)
- Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-93-4894169
| |
Collapse
|
24
|
Pulver EM, Mukherjee C, van de Kamp G, Roobol SJ, Rother MB, van der Gulden H, de Bruijn R, Lattanzio MV, van der Burg E, Drenth AP, Verkaik NS, Hahn K, Klarenbeek S, de Korte-Grimmerink R, van de Ven M, Pritchard CEJ, Huijbers IJ, Xia B, van Gent DC, Essers J, van Attikum H, Ray Chaudhuri A, Bouwman P, Jonkers J. A BRCA1 coiled-coil domain variant disrupting PALB2 interaction promotes the development of mammary tumors and confers a targetable defect in homologous recombination repair. Cancer Res 2021; 81:6171-6182. [PMID: 34548335 PMCID: PMC7612117 DOI: 10.1158/0008-5472.can-21-1415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022]
Abstract
The BRCA1 tumor suppressor gene encodes a multi-domain protein for which several functions have been described. These include a key role in homologous recombination repair (HRR) of DNA double-strand breaks (DSB), which is shared with two other high-risk hereditary breast cancer suppressors, BRCA2 and PALB2. Although both BRCA1 and BRCA2 interact with PALB2, BRCA1 missense variants affecting its PALB2-interacting coiled-coil domain are considered variants of uncertain clinical significance (VUS). Using genetically engineered mice, we show here that a BRCA1 coiled-coil domain VUS, Brca1 p.L1363P, disrupts the interaction with PALB2 and leads to embryonic lethality. Brca1 p.L1363P led to a similar acceleration in the development of Trp53-deficient mammary tumors as Brca1 loss, but the tumors showed distinct histopathological features, with more stable DNA copy number profiles in Brca1 p.L1363P tumors. Nevertheless, Brca1 p.L1363P mammary tumors were HRR-incompetent and responsive to cisplatin and PARP inhibition. Overall, these results provide the first direct evidence that a BRCA1 missense variant outside of the RING and BRCT domains increases the risk of breast cancer.
Collapse
Affiliation(s)
- Emilia M Pulver
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute
| | | | | | - Stefan J Roobol
- Molecular Genetics, Radiology and Nuclear Medicine, Erasmus MC
| | | | - Hanneke van der Gulden
- Division of Molecular Pathology and Cancer Genomics Centre, The Netherlands Cancer Institute
| | - Roebi de Bruijn
- Division of Molecular Pathology, The Netherlands Cancer Institute
| | | | | | | | | | - Kerstin Hahn
- Molecular Pathology, Oncode Institute-Netherlands Cancer Institute
| | | | - Renske de Korte-Grimmerink
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging (MCCA) Preclinical Intervention Unit, The Netherlands Cancer Institute
| | | | - Ivo J Huijbers
- Transgenic core facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute
| | - Bing Xia
- Radiation Oncology, Rutgers Cancer Institute of New Jersey
| | - Dik C van Gent
- Molecular Genetics, Erasmus MC University Medical Center Rotterdam and Oncode Institute, The Netherlands
| | | | | | | | - Peter Bouwman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute
| |
Collapse
|
25
|
Franchet C, Hoffmann JS, Dalenc F. Recent Advances in Enhancing the Therapeutic Index of PARP Inhibitors in Breast Cancer. Cancers (Basel) 2021; 13:cancers13164132. [PMID: 34439286 PMCID: PMC8392832 DOI: 10.3390/cancers13164132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Two to three percent of breast cancer patients harbor germline mutation of either BRCA1 or BRCA2 genes. Their tumor cells are deficient in homologous recombination, a BRCA-dependent DNA repair machinery. These deficient cells survive thanks to the PARP-mediated alternative pathway. Therefore, PARP inhibitors have already shown some level of efficiency in the treatment of metastatic breast cancer patients. Unfortunately, some tumor cells inevitably resist PARP inhibitors by different mechanisms. In this review, we (i) present the notion of homologous recombination deficiency and its evaluation methods, (ii) detail the PARP inhibitor clinical trials in breast cancer, (iii) briefly describe the mechanisms to PARP inhibitors resistance, and (iv) discuss some strategies currently under evaluation to enhance the therapeutic index of PARP inhibitors in breast cancer. Abstract As poly-(ADP)-ribose polymerase (PARP) inhibition is synthetic lethal with the deficiency of DNA double-strand (DSB) break repair by homologous recombination (HR), PARP inhibitors (PARPi) are currently used to treat breast cancers with mutated BRCA1/2 HR factors. Unfortunately, the increasingly high rate of PARPi resistance in clinical practice has dented initial hopes. Multiple resistance mechanisms and acquired vulnerabilities revealed in vitro might explain this setback. We describe the mechanisms and vulnerabilities involved, including newly identified modes of regulation of DSB repair that are now being tested in large cohorts of patients and discuss how they could lead to novel treatment strategies to improve the therapeutic index of PARPi.
Collapse
Affiliation(s)
- Camille Franchet
- Laboratoire de Pathologie and Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 Av. Irène Joliot-Curie, 31100 Toulouse, France;
| | - Jean-Sébastien Hoffmann
- Laboratoire d’Excellence Toulouse Cancer (TOUCAN), Laboratoire de Pathologie, Institut Universitaire du Cancer-Toulouse, 31037 Toulouse, France;
| | - Florence Dalenc
- Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 Av. Irène Joliot-Curie, 31100 Toulouse, France
- Correspondence:
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW To summarize recently discovered PARP inhibitor resistance mechanisms and highlight the clinical relevance of these findings to date. RECENT FINDINGS A predominant mechanism of acquired PARP inhibitor resistance in homologous recombination-deficient cancers is the acquisition of homologous recombination proficiency as a consequence of secondary genetic or epigenetic events, such as secondary mutations in BRCA1 or BRCA2, or reversal of BRCA1 promoter methylation that restores homologous recombination and leads to PARP inhibitor resistance. Multiple other potential mechanisms of acquired resistance to PARP inhibitors including loss of DNA end resection inhibition (53BP1/REV7/RIF1/Sheldin) or DNA replication fork protection (PTIP/EZH2), but also increased drug efflux or induction of a reversible senescent or mesenchymal cell state have been described in ovarian cancer models. However, only few of these mechanisms have been identified in clinical samples. SUMMARY Multiple adaptive responses following PARP inhibitor treatment have been identified. Further research is needed to better understand what role these mechanisms play for clinical PARP inhibitor resistance and how these mechanisms may render ovarian cancer cells susceptible to subsequent novel combination therapies.
Collapse
|
27
|
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: 41] [Impact Index Per Article: 10.3] [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
|
28
|
Roarty K, Echeverria GV. Laboratory Models for Investigating Breast Cancer Therapy Resistance and Metastasis. Front Oncol 2021; 11:645698. [PMID: 33777805 PMCID: PMC7988094 DOI: 10.3389/fonc.2021.645698] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/28/2021] [Indexed: 01/16/2023] Open
Abstract
While numerous therapies are highly efficacious in early-stage breast cancers and in particular subsets of breast cancers, therapeutic resistance and metastasis unfortunately arise in many patients. In many cases, tumors that are resistant to standard of care therapies, as well as tumors that have metastasized, are treatable but incurable with existing clinical strategies. Both therapy resistance and metastasis are multi-step processes during which tumor cells must overcome diverse environmental and selective hurdles. Mechanisms by which tumor cells achieve this are numerous and include acquisition of invasive and migratory capabilities, cell-intrinsic genetic and/or epigenetic adaptations, clonal selection, immune evasion, interactions with stromal cells, entering a state of dormancy or senescence, and maintaining self-renewal capacity. To overcome therapy resistance and metastasis in breast cancer, the ability to effectively model each of these mechanisms in the laboratory is essential. Herein we review historic and the current state-of-the-art laboratory model systems and experimental approaches used to investigate breast cancer metastasis and resistance to standard of care therapeutics. While each model system has inherent limitations, they have provided invaluable insights, many of which have translated into regimens undergoing clinical evaluation. We will discuss the limitations and advantages of a variety of model systems that have been used to investigate breast cancer metastasis and therapy resistance and outline potential strategies to improve experimental modeling to further our knowledge of these processes, which will be crucial for the continued development of effective breast cancer treatments.
Collapse
Affiliation(s)
- Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Gloria V Echeverria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States.,Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States.,Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
29
|
Abstract
Breast cancer is the most common malignancy in women. Basic and translational breast cancer research relies heavily on experimental animal models. Ideally, such models for breast cancer should have commonality with human breast cancer in terms of tumor etiology, biological behavior, pathology, and response to therapeutics. This review introduces current progress in different breast cancer experimental animal models and analyzes their characteristics, advantages, disadvantages, and potential applications. Finally, we propose future research directions for breast cancer animal models.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Wei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ce-Shi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
30
|
Abstract
Platinum (Pt) compounds entered the clinic as anticancer agents when cisplatin was approved in 1978. More than 40 years later, even in the era of precision medicine and immunotherapy, Pt drugs remain among the most widely used anticancer drugs. As Pt drugs mainly target DNA, it is not surprising that recent insights into alterations of DNA repair mechanisms provide a useful explanation for their success. Many cancers have defective DNA repair, a feature that also sheds new light on the mechanisms of secondary drug resistance, such as the restoration of DNA repair pathways. In addition, genome-wide functional screening approaches have revealed interesting insights into Pt drug uptake. About half of cisplatin and carboplatin but not oxaliplatin may enter cells through the widely expressed volume-regulated anion channel (VRAC). The analysis of this heteromeric channel in tumour biopsies may therefore be a useful biomarker to stratify patients for initial Pt treatments. Moreover, Pt-based approaches may be improved in the future by the optimization of combinations with immunotherapy, management of side effects and use of nanodelivery devices. Hence, Pt drugs may still be part of the standard of care for several cancers in the coming years.
Collapse
Affiliation(s)
- Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
| | - Carmen Disler
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Paola Perego
- Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
31
|
BRCAness as a prognostic indicator in patients with early breast cancer. Sci Rep 2020; 10:21173. [PMID: 33273622 PMCID: PMC7713365 DOI: 10.1038/s41598-020-78016-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 10/14/2020] [Indexed: 11/28/2022] Open
Abstract
BRCAness is defined as a phenotypic copy of germline BRCA mutations, which describes presence of homologous recombination defects in sporadic cancers. We detected BRCAness by multiplex ligation-dependent probe amplification (MLPA) and explored whether BRCAness can be used as a predictor of prognosis. BRCAness status was classified for total 121 breast cancer patients. Forty-eight patients (39.7%) were identified as BRCAness positive. Tumors of BRCAness were more likely to be hormone receptors negative (95.8% vs. 50.7%, P < 0.001), nuclear grade III (76.1% vs. 48.4%, P = 0.001) and triple-negative breast cancer subtype (91.6% vs. 42.5%, P < 0.001). Five-year disease free survival (DFS) (54.0% vs. 88.0%, P < 0.001) and overall survival (OS) (76.3% vs. 93.1%, P = 0.002) were significantly lower in BRCAness patients. In neoadjuvant chemotherapy subgroup analysis, clinical response rate for taxane-based regimen was significantly lower in BRCAness patients (58.3% vs. 77.8%, P = 0.041). Cox regression multivariate analysis showed that BRCAness was the independent prognostic factor for DFS (HR 2.962, 95%CI 1.184–7.412, P = 0.020), but not for OS (HR 2.681, 95%CI 0.618–11.630, P = 0.188). BRCAness is associated with specific characteristics and may suggest resistance to taxane-based chemotherapy. BRCAness can be used as a negative prognostic indicator for breast cancer.
Collapse
|
32
|
Borst P. Looking back at multidrug resistance (MDR) research and ten mistakes to be avoided when writing about ABC transporters in MDR. FEBS Lett 2020; 594:4001-4011. [PMID: 33111311 DOI: 10.1002/1873-3468.13972] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/31/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022]
Abstract
This paper presents a personal, selective, and sometimes critical retrospective of the history of ABC transporters in multidrug resistance (MDR) of cancer cells, overrepresenting discoveries of some early pioneers, long forgotten, and highlights of research in Amsterdam, mainly focussing on discoveries made with disruptions of ABC genes in mice (KO mice) and on the role of ABC transporters in causing drug resistance in a mouse model of mammary cancer. The history is complemented by a list of erroneous concepts often found in papers and grant applications submitted anno 2020.
Collapse
Affiliation(s)
- Piet Borst
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Trusler O, Goodwin J, Laslett AL. BRCA1 and BRCA2 associated breast cancer and the roles of current modelling systems in drug discovery. Biochim Biophys Acta Rev Cancer 2020; 1875:188459. [PMID: 33129865 DOI: 10.1016/j.bbcan.2020.188459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/08/2023]
Abstract
For a drug candidate to be fully developed takes years and investment of hundreds of millions of dollars. There is no doubt that drug development is difficult and risky, but vital to protecting against devastating disease. This difficulty is clearly evident in BRCA1 and BRCA2 related breast cancer, with current treatment options largely confined to invasive surgical procedures, as well as chemotherapy and radiotherapy regimes which damage healthy tissue and can leave remnant disease. Consequently, patient survival and relapse rates are far from ideal, and new candidate treatments are needed. The preclinical stages of drug discovery are crucial to get right for translation to hospital beds. Disease models must take advantage of current technologies and be accurate for rapid and translatable treatments. Careful selection of cell lines must be coupled with high throughput techniques, with promising results trialled further in highly accurate humanised patient derived xenograft models. Traditional adherent drug screening should transition to 3D culture systems amenable to high throughput techniques if the gap between in vitro and in vivo studies is to be partially bridged. The possibility of organoid, induced pluripotent stem cell, and conditionally reprogrammed in vitro models is tantalising, however protocols are yet to be fully established. This review of BRCA1 and BRCA2 cancer biology and current modelling systems will hopefully guide the design of future drug discovery endeavours and highlight areas requiring improvement.
Collapse
Affiliation(s)
- Oliver Trusler
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia; Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Jacob Goodwin
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia; Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Andrew L Laslett
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia; Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia.
| |
Collapse
|
34
|
Olatunde OZ, Yong J, Lu C. The Progress of the Anticancer Agents Related to the Microtubules Target. Mini Rev Med Chem 2020; 20:2165-2192. [PMID: 32727327 DOI: 10.2174/1389557520666200729162510] [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: 03/31/2020] [Revised: 05/11/2020] [Accepted: 05/22/2020] [Indexed: 11/22/2022]
Abstract
Anticancer drugs based on the microtubules target are potent mitotic spindle poison agents, which interact directly with the microtubules, and were classified as microtubule-stabilizing agents and microtubule-destabilizing agents. Researchers have worked tremendously towards the improvements of anticancer drugs, in terms of improving the efficacy, solubility and reducing the side effects, which brought about advancement in chemotherapy. In this review, we focused on describing the discovery, structures and functions of the microtubules as well as the progress of anticancer agents related to the microtubules, which will provide adequate references for researchers.
Collapse
Affiliation(s)
- Olagoke Zacchaeus Olatunde
- CAS Key Laboratory of Desing and Assembly of Functional Nanostructures, and Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structures of Matter, Chinese Academy of Sciences. Fuzhou, Fujian, 350002, China
| | - Jianping Yong
- Xiamen Institute of Rare-Earth Materials, Chinese Academy of Sciences, Xiamen, Fujian, 361021, China
| | - Canzhong Lu
- CAS Key Laboratory of Desing and Assembly of Functional Nanostructures, and Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structures of Matter, Chinese Academy of Sciences. Fuzhou, Fujian, 350002, China
| |
Collapse
|
35
|
Cipponi A, Goode DL, Bedo J, McCabe MJ, Pajic M, Croucher DR, Rajal AG, Junankar SR, Saunders DN, Lobachevsky P, Papenfuss AT, Nessem D, Nobis M, Warren SC, Timpson P, Cowley M, Vargas AC, Qiu MR, Generali DG, Keerthikumar S, Nguyen U, Corcoran NM, Long GV, Blay JY, Thomas DM. MTOR signaling orchestrates stress-induced mutagenesis, facilitating adaptive evolution in cancer. Science 2020; 368:1127-1131. [PMID: 32499442 DOI: 10.1126/science.aau8768] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/09/2019] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
In microorganisms, evolutionarily conserved mechanisms facilitate adaptation to harsh conditions through stress-induced mutagenesis (SIM). Analogous processes may underpin progression and therapeutic failure in human cancer. We describe SIM in multiple in vitro and in vivo models of human cancers under nongenotoxic drug selection, paradoxically enhancing adaptation at a competing intrinsic fitness cost. A genome-wide approach identified the mechanistic target of rapamycin (MTOR) as a stress-sensing rheostat mediating SIM across multiple cancer types and conditions. These observations are consistent with a two-phase model for drug resistance, in which an initially rapid expansion of genetic diversity is counterbalanced by an intrinsic fitness penalty, subsequently normalizing to complete adaptation under the new conditions. This model suggests synthetic lethal strategies to minimize resistance to anticancer therapy.
Collapse
Affiliation(s)
- Arcadi Cipponi
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia. .,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David L Goode
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Justin Bedo
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Computing and Information Systems, the University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Mark J McCabe
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Alvaro Gonzalez Rajal
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Simon R Junankar
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, NSW, Australia
| | | | - Anthony T Papenfuss
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Computing and Information Systems, the University of Melbourne, Parkville, VIC, Australia.,Peter MacCallum Cancer Centre, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Danielle Nessem
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Max Nobis
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Sean C Warren
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Mark Cowley
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Ana C Vargas
- Douglass Hanly Moir Pathology, Turramurra, NSW, Australia
| | - Min R Qiu
- St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.,Anatomical and Molecular Oncology Pathology, SYDPATH, St. Vincent's Hospital, Darlinghurst, NSW, Australia
| | - Daniele G Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Breast Cancer Unit and Translational Research Unit, ASST Cremona, Cremona, Italy
| | - Shivakumar Keerthikumar
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Uyen Nguyen
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Niall M Corcoran
- Division of Urology, Royal Melbourne Hospital, Parkville, VIC, Australia.,Department of Urology, Peninsula Health, Frankston, VIC, Australia.,Department of Surgery, University of Melbourne, VIC, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Wollstonecraft, NSW, Australia.,The University of Sydney, Sydney, NSW, Australia.,Royal North Shore Hospital and Mater Hospital, Sydney, NSW, Australia.,Crown Princess Mary Cancer Centre Westmead Hospital, Sydney, NSW, Australia
| | - Jean-Yves Blay
- Centre Leon Berard and Université Claude Bernard Lyon, Lyon, France.,UNICANCER, Paris, France
| | - David M Thomas
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia. .,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
36
|
Tarsounas M, Sung P. The antitumorigenic roles of BRCA1-BARD1 in DNA repair and replication. Nat Rev Mol Cell Biol 2020; 21:284-299. [PMID: 32094664 PMCID: PMC7204409 DOI: 10.1038/s41580-020-0218-z] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 11/09/2022]
Abstract
The tumour suppressor breast cancer type 1 susceptibility protein (BRCA1) promotes DNA double-strand break (DSB) repair by homologous recombination and protects DNA replication forks from attrition. BRCA1 partners with BRCA1-associated RING domain protein 1 (BARD1) and other tumour suppressor proteins to mediate the initial nucleolytic resection of DNA lesions and the recruitment and regulation of the recombinase RAD51. The discovery of the opposing functions of BRCA1 and the p53-binding protein 1 (53BP1)-associated complex in DNA resection sheds light on how BRCA1 influences the choice of homologous recombination over non-homologous end joining and potentially other mutagenic pathways of DSB repair. Understanding the functional crosstalk between BRCA1-BARD1 and its cofactors and antagonists will illuminate the molecular basis of cancers that arise from a deficiency or misregulation of chromosome damage repair and replication fork maintenance. Such knowledge will also be valuable for understanding acquired tumour resistance to poly(ADP-ribose) polymerase (PARP) inhibitors and other therapeutics and for the development of new treatments. In this Review, we discuss recent advances in elucidating the mechanisms by which BRCA1-BARD1 functions in DNA repair, replication fork maintenance and tumour suppression, and its therapeutic relevance.
Collapse
Affiliation(s)
- Madalena Tarsounas
- Genome Stability and Tumourigenesis Group, Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
37
|
Celecoxib Prevents Doxorubicin-Induced Multidrug Resistance in Canine and Mouse Lymphoma Cell Lines. Cancers (Basel) 2020; 12:cancers12051117. [PMID: 32365663 PMCID: PMC7280963 DOI: 10.3390/cancers12051117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 11/30/2022] Open
Abstract
Background: Treatment of malignancies is still a major challenge in human and canine cancer, mostly due to the emergence of multidrug resistance (MDR). One of the main contributors of MDR is the overexpression P-glycoprotein (Pgp), which recognizes and extrudes various chemotherapeutics from cancer cells. Methods: To study mechanisms underlying the development of drug resistance, we established an in vitro treatment protocol to rapidly induce Pgp-mediated MDR in cancer cells. Based on a clinical observation showing that a 33-day-long, unplanned drug holiday can reverse the MDR phenotype of a canine diffuse large B-cell lymphoma patient, our aim was to use the established assay to prevent the emergence of drug resistance in the early stages of treatment. Results: We showed that an in vitro drug holiday results in the decrease of Pgp expression in MDR cell lines. Surprisingly, celecoxib, a known COX-2 inhibitor, prevented the emergence of drug-induced MDR in murine and canine lymphoma cell lines. Conclusions: Our findings suggest that celecoxib could significantly improve the efficiency of chemotherapy by preventing the development of MDR in B-cell lymphoma.
Collapse
|
38
|
Triple-Negative Breast Cancer: A Review of Conventional and Advanced Therapeutic Strategies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17062078. [PMID: 32245065 PMCID: PMC7143295 DOI: 10.3390/ijerph17062078] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) cells are deficient in estrogen, progesterone and ERBB2 receptor expression, presenting a particularly challenging therapeutic target due to their highly invasive nature and relatively low response to therapeutics. There is an absence of specific treatment strategies for this tumor subgroup, and hence TNBC is managed with conventional therapeutics, often leading to systemic relapse. In terms of histology and transcription profile these cancers have similarities to BRCA-1-linked breast cancers, and it is hypothesized that BRCA1 pathway is non-functional in this type of breast cancer. In this review article, we discuss the different receptors expressed by TNBC as well as the diversity of different signaling pathways targeted by TNBC therapeutics, for example, Notch, Hedgehog, Wnt/b-Catenin as well as TGF-beta signaling pathways. Additionally, many epidermal growth factor receptor (EGFR), poly (ADP-ribose) polymerase (PARP) and mammalian target of rapamycin (mTOR) inhibitors effectively inhibit the TNBCs, but they face challenges of either resistance to drugs or relapse. The resistance of TNBC to conventional therapeutic agents has helped in the advancement of advanced TNBC therapeutic approaches including hyperthermia, photodynamic therapy, as well as nanomedicine-based targeted therapeutics of drugs, miRNA, siRNA, and aptamers, which will also be discussed. Artificial intelligence is another tool that is presented to enhance the diagnosis of TNBC.
Collapse
|
39
|
Becker S, Kiecke C, Schäfer E, Sinzig U, Deuper L, Trigo-Mourino P, Griesinger C, Koch R, Rydzynska Z, Chapuy B, von Bonin F, Kube D, Venkataramani V, Bohnenberger H, Leha A, Flach J, Dierks S, Bastians H, Maruschak B, Bojarczuk K, Taveira MDO, Trümper L, Wulf GM, Wulf GG. Destruction of a Microtubule-Bound MYC Reservoir during Mitosis Contributes to Vincristine's Anticancer Activity. Mol Cancer Res 2020; 18:859-872. [PMID: 32161139 DOI: 10.1158/1541-7786.mcr-19-1203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/11/2020] [Accepted: 03/09/2020] [Indexed: 11/16/2022]
Abstract
Tightly regulated activity of the transcription factor MYC is essential for orderly cell proliferation. Upon deregulation, MYC elicits and promotes cancer progression. Proteasomal degradation is an essential element of MYC regulation, initiated by phosphorylation at Serine62 (Ser62) of the MB1 region. Here, we found that Ser62 phosphorylation peaks in mitosis, but that a fraction of nonphosphorylated MYC binds to the microtubules of the mitotic spindle. Consequently, the microtubule-destabilizing drug vincristine decreases wild-type MYC stability, whereas phosphorylation-deficient MYC is more stable, contributing to vincristine resistance and induction of polyploidy. PI3K inhibition attenuates postmitotic MYC formation and augments the cytotoxic effect of vincristine. IMPLICATIONS: The spindle's function as a docking site for MYC during mitosis may constitute a window of specific vulnerability to be exploited for cancer treatment.
Collapse
Affiliation(s)
- Sabrina Becker
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Christina Kiecke
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Eva Schäfer
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Ursula Sinzig
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Lena Deuper
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Pablo Trigo-Mourino
- Max-Planck Institute for Biophysical Chemistry, Goettingen, Germany.,Analytical Research and Development, Merck & Co., Inc., Kenilworth, New Jersey
| | | | - Raphael Koch
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Zuzanna Rydzynska
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Bjoern Chapuy
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Frederike von Bonin
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Dieter Kube
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Vivek Venkataramani
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | | | - Andreas Leha
- Department of Medical Statistics, University Medicine Goettingen, Goettingen, Germany
| | - Johanna Flach
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Sascha Dierks
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Holger Bastians
- Department of Experimental Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Brigitte Maruschak
- Institute for Neuropathology, University Medicine Goettingen, Goettingen, Germany
| | - Kamil Bojarczuk
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany.,Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | | | - Lorenz Trümper
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Gerburg M Wulf
- Department of Medicine, BIDMC/Harvard Medical School, Boston, Massachusetts
| | - Gerald G Wulf
- Department of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany.
| |
Collapse
|
40
|
Hámori L, Kudlik G, Szebényi K, Kucsma N, Szeder B, Póti Á, Uher F, Várady G, Szüts D, Tóvári J, Füredi A, Szakács G. Establishment and Characterization of a Brca1 -/-, p53 -/- Mouse Mammary Tumor Cell Line. Int J Mol Sci 2020; 21:ijms21041185. [PMID: 32053991 PMCID: PMC7072850 DOI: 10.3390/ijms21041185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/25/2020] [Accepted: 02/01/2020] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most commonly occurring cancer in women and the second most common cancer overall. By the age of 80, the estimated risk for breast cancer for women with germline BRCA1 or BRCA2 mutations is around 80%. Genetically engineered BRCA1-deficient mouse models offer a unique opportunity to study the pathogenesis and therapy of triple negative breast cancer. Here we present a newly established Brca1−/−, p53−/− mouse mammary tumor cell line, designated as CST. CST shows prominent features of BRCA1-mutated triple-negative breast cancers including increased motility, high proliferation rate, genome instability and sensitivity to platinum chemotherapy and PARP inhibitors (olaparib, veliparib, rucaparib and talazoparib). Genomic instability of CST cells was confirmed by whole genome sequencing, which also revealed the presence of COSMIC (Catalogue of Somatic Mutations in Cancer) mutation signatures 3 and 8 associated with homologous recombination (HR) deficiency. In vitro sensitivity of CST cells was tested against 11 chemotherapy agents. Tumors derived from orthotopically injected CST-mCherry cells in FVB-GFP mice showed sensitivity to cisplatin, providing a new model to study the cooperation of BRCA1-KO, mCherry-positive tumor cells and the GFP-expressing stromal compartment in therapy resistance and metastasis formation. In summary, we have established CST cells as a new model recapitulating major characteristics of BRCA1-negative breast cancers.
Collapse
Affiliation(s)
- Lilla Hámori
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - Gyöngyi Kudlik
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - Kornélia Szebényi
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
- Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Nóra Kucsma
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - Bálint Szeder
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - Ádám Póti
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - Ferenc Uher
- Central Hospital of Southern Pest—National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary;
| | - György Várady
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - Dávid Szüts
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, 1122, Budapest, Hungary;
| | - András Füredi
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
- Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: (A.F.); (G.S.)
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, 1117 Budapest, Hungary; (L.H.); (G.K.); (K.S.); (N.K.); (B.S.); (Á.P.); (G.V.); (D.S.)
- Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: (A.F.); (G.S.)
| |
Collapse
|
41
|
Kannan P, Füredi A, Dizdarevic S, Wanek T, Mairinger S, Collins J, Falls T, van Dam RM, Maheshwari D, Lee JT, Szakács G, Langer O. In vivo characterization of [ 18F]AVT-011 as a radiotracer for PET imaging of multidrug resistance. Eur J Nucl Med Mol Imaging 2019; 47:2026-2035. [PMID: 31729540 PMCID: PMC7299908 DOI: 10.1007/s00259-019-04589-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/22/2019] [Indexed: 11/30/2022]
Abstract
Purpose Multidrug resistance (MDR) impedes cancer treatment. Two efflux transporters from the ATP-binding cassette (ABC) family, ABCB1 and ABCG2, may contribute to MDR by restricting the entry of therapeutic drugs into tumor cells. Although a higher expression of these transporters has been correlated with an unfavorable response to chemotherapy, transporter expression does not necessarily correlate with function. In this study, we characterized the pharmacological properties of [18F]AVT-011, a new PET radiotracer for imaging transporter-mediated MDR in tumors. Methods AVT-011 was radiolabeled with 18F and evaluated with PET imaging in preclinical models. Transport of [18F]AVT-011 by ABCB1 and/or ABCG2 was assessed by measuring its uptake in the brains of wild-type, Abcb1a/b−/−, and Abcg2−/− mice at baseline and after administration of the ABCB1 inhibitor tariquidar (n = 5/group). Metabolism and biodistribution of [18F]AVT-011 were also measured. To measure ABCB1 function in tumors, we performed PET experiments using both [18F]AVT-011 and [18F]FDG in mice bearing orthotopic breast tumors (n = 7–10/group) expressing clinically relevant levels of ABCB1. Results At baseline, brain uptake was highest in Abcb1a/b−/− mice. After tariquidar administration, brain uptake increased 3-fold and 8-fold in wild-type and Abcg2−/− mice, respectively, but did not increase further in Abcb1a/b−/− mice. At 30 min after injection, the radiotracer was > 90% in its parent form and had highest uptake in organs of the hepatobiliary system. Compared with that in drug-sensitive tumors, uptake of [18F]AVT-011 was 32% lower in doxorubicin-resistant tumors with highest ABCB1 expression and increased by 40% with tariquidar administration. Tumor uptake of [18F]FDG did not significantly differ among groups. Conclusion [18F]AVT-011 is a dual ABCB1/ABCG2 substrate radiotracer that can quantify transporter function at the blood-brain barrier and in ABCB1-expressing tumors, making it potentially suitable for clinical imaging of ABCB1-mediated MDR in tumors. Electronic supplementary material The online version of this article (10.1007/s00259-019-04589-w) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pavitra Kannan
- CRUK and MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK. .,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - András Füredi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Sabina Dizdarevic
- Brighton and Sussex University Hospitals, NHS Trust and Brighton and Sussex Medical School, Brighton, UK
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Jeffrey Collins
- Crump Institute for Molecular Imaging and Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Theresa Falls
- Crump Institute for Molecular Imaging and Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - R Michael van Dam
- Crump Institute for Molecular Imaging and Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Jason T Lee
- Crump Institute for Molecular Imaging and Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Stanford Center for Innovations in In vivo Imaging, Stanford University School of Medicine, Stanford, CA, USA
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Noordermeer SM, van Attikum H. PARP Inhibitor Resistance: A Tug-of-War in BRCA-Mutated Cells. Trends Cell Biol 2019; 29:820-834. [PMID: 31421928 DOI: 10.1016/j.tcb.2019.07.008] [Citation(s) in RCA: 308] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Poly-(ADP)-ribose polymerase (PARP) inhibition is synthetic lethal with deficiency for homologous recombination (HR), a pathway essential for DNA double-strand break repair. PARP inhibitors (PARPi) therefore hold great promise for the treatment of tumors with disruptive mutations in BRCA1/2 or other HR factors. Unfortunately, PARPi resistance has proved to be a major problem in the clinic. Knowledge about PARPi resistance is expanding quickly, revealing four main mechanisms that alter drug availability, affect (de)PARylation enzymes, restore HR, or restore replication fork stability. We discuss how studies on resistance mechanisms have yielded important insights into the regulation of DNA double-strand break (DSB) repair and replication fork protection, and how these studies could pave the way for novel treatment options to target resistance mechanisms or acquired vulnerabilities.
Collapse
Affiliation(s)
- Sylvie M Noordermeer
- Leiden University Medical Center, Department of Human Genetics, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands.
| | - Haico van Attikum
- Leiden University Medical Center, Department of Human Genetics, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| |
Collapse
|
43
|
Puppe J, Opdam M, Schouten PC, Jóźwiak K, Lips E, Severson T, van de Ven M, Brambillasca C, Bouwman P, van Tellingen O, Bernards R, Wesseling J, Eichler C, Thangarajah F, Malter W, Pandey GK, Ozretić L, Caldas C, van Lohuizen M, Hauptmann M, Rhiem K, Hahnen E, Reinhardt HC, Büttner R, Mallmann P, Schömig-Markiefka B, Schmutzler R, Linn S, Jonkers J. EZH2 Is Overexpressed in BRCA1-like Breast Tumors and Predictive for Sensitivity to High-Dose Platinum-Based Chemotherapy. Clin Cancer Res 2019; 25:4351-4362. [PMID: 31036541 DOI: 10.1158/1078-0432.ccr-18-4024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/25/2019] [Accepted: 04/24/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE BRCA1-deficient breast cancers carry a specific DNA copy-number signature ("BRCA1-like") and are hypersensitive to DNA double-strand break (DSB) inducing compounds. Here, we explored whether (i) EZH2 is overexpressed in human BRCA1-deficient breast tumors and might predict sensitivity to DSB-inducing drugs; (ii) EZH2 inhibition potentiates cisplatin efficacy in Brca1-deficient murine mammary tumors. EXPERIMENTAL DESIGN EZH2 expression was analyzed in 497 breast cancers using IHC or RNA sequencing. We classified 370 tumors by copy-number profiles as BRCA1-like or non-BRCA1-like and examined its association with EZH2 expression. Additionally, we assessed BRCA1 loss through mutation or promoter methylation status and investigated the predictive value of EZH2 expression in a study population of breast cancer patients treated with adjuvant high-dose platinum-based chemotherapy compared with standard anthracycline-based chemotherapy. To explore whether EZH2 inhibition by GSK126 enhances sensitivity to platinum drugs in EZH2-overexpressing breast cancers we used a Brca1-deficient mouse model. RESULTS The highest EZH2 expression was found in BRCA1-associated tumors harboring a BRCA1 mutation, BRCA1-promoter methylation or were classified as BRCA1 like. We observed a greater benefit from high-dose platinum-based chemotherapy in BRCA1-like and non-BRCA1-like patients with high EZH2 expression. Combined treatment with the EZH2 inhibitor GSK126 and cisplatin decreased cell proliferation and improved survival in Brca1-deficient mice in comparison with single agents. CONCLUSIONS Our findings demonstrate that EZH2 is expressed at significantly higher levels in BRCA1-deficient breast cancers. EZH2 overexpression can identify patients with breast cancer who benefit significantly from intensified DSB-inducing platinum-based chemotherapy independent of BRCA1-like status. EZH2 inhibition improves the antitumor effect of platinum drugs in Brca1-deficient breast tumors in vivo.
Collapse
Affiliation(s)
- Julian Puppe
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
- Department of Obstetrics and Gynecology, Medical Faculty, University Hospital Cologne, Cologne, Germany
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne, Germany
| | - Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Philip C Schouten
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Katarzyna Jóźwiak
- Department of Epidemiology and Biostatistics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Esther Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tesa Severson
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marieke van de Ven
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Chiara Brambillasca
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Peter Bouwman
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - René Bernards
- Oncode Institute, Utrecht, the Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christian Eichler
- Department of Obstetrics and Gynecology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Fabinshy Thangarajah
- Department of Obstetrics and Gynecology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Wolfram Malter
- Department of Obstetrics and Gynecology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Gaurav Kumar Pandey
- Oncode Institute, Utrecht, the Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Luka Ozretić
- Department of Pathology, University Hospital of Cologne, Cologne, Germany
| | | | - Maarten van Lohuizen
- Oncode Institute, Utrecht, the Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michael Hauptmann
- Department of Epidemiology and Biostatistics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kerstin Rhiem
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne, Germany
| | - Eric Hahnen
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne, Germany
| | | | - Reinhard Büttner
- Department of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Peter Mallmann
- Department of Obstetrics and Gynecology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | | | - Rita Schmutzler
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne, Germany
| | - Sabine Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
44
|
Geenen JJJ, Linn SC, Beijnen JH, Schellens JHM. PARP Inhibitors in the Treatment of Triple-Negative Breast Cancer. Clin Pharmacokinet 2019; 57:427-437. [PMID: 29063517 DOI: 10.1007/s40262-017-0587-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer is a heterogeneous disease, manifesting in a broad differentiation in phenotypes and morphologic profiles, resulting in variable clinical behavior. Between 10 and 20% of all breast cancers are triple negative. Triple-negative breast cancer (TNBC) lacks the expression of human epidermal growth factor receptor 2 (HER2) and hormone receptors; therefore, to date, chemotherapy remains the backbone of treatment. TNBC tends to be aggressive and has a high histological grade, resulting in a poor 5-year prognosis. It has a high prevalence of BRCA1 mutations and an increased Ki-67 expression. This subtype usually responds well to taxanes and/or platinum compounds and poly (ADP-ribose) polymerase (PARP) inhibitors. Studies with PARP inhibitors have demonstrated promising results in the treatment of BRCA-mutated breast and ovarian cancer, and PARP inhibitors have been studied as monotherapy and in combination with cytotoxic therapy or radiotherapy. PARP inhibitor efficacy on poly (ADP-ribose) polymer (PAR) formation in vivo can be quantified by pharmacodynamic assays that measure PAR activity in peripheral blood mononuclear cells (PBMC). Biomarkers such as TP53, ATM, PALB2 and RAD51C might be prognostic or predictive indicators for treatment response, and could also provide targets for novel treatment strategies. In summary, this review provides an overview of the treatment options for basal-like TNBC, including PARP inhibitors, and focuses on the pharmacotherapeutic options in these patients.
Collapse
Affiliation(s)
- Jill J J Geenen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Sabine C Linn
- Department of Molecular Pathology, Antoni van Leeuwenhoek Hospital, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Division of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Department of Pathology, Utrecht University Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Department of Pharmacy, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands. .,Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands. .,Division of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands. .,Department of Pharmacy, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands. .,Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
45
|
Tacconi EMC, Badie S, De Gregoriis G, Reisländer T, Lai X, Porru M, Folio C, Moore J, Kopp A, Baguña Torres J, Sneddon D, Green M, Dedic S, Lee JW, Batra AS, Rueda OM, Bruna A, Leonetti C, Caldas C, Cornelissen B, Brino L, Ryan A, Biroccio A, Tarsounas M. Chlorambucil targets BRCA1/2-deficient tumours and counteracts PARP inhibitor resistance. EMBO Mol Med 2019; 11:e9982. [PMID: 31273933 PMCID: PMC6609913 DOI: 10.15252/emmm.201809982] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 01/03/2023] Open
Abstract
Due to compromised homologous recombination (HR) repair, BRCA1- and BRCA2-mutated tumours accumulate DNA damage and genomic rearrangements conducive of tumour progression. To identify drugs that target specifically BRCA2-deficient cells, we screened a chemical library containing compounds in clinical use. The top hit was chlorambucil, a bifunctional alkylating agent used for the treatment of chronic lymphocytic leukaemia (CLL). We establish that chlorambucil is specifically toxic to BRCA1/2-deficient cells, including olaparib-resistant and cisplatin-resistant ones, suggesting the potential clinical use of chlorambucil against disease which has become resistant to these drugs. Additionally, chlorambucil eradicates BRCA2-deficient xenografts and inhibits growth of olaparib-resistant patient-derived tumour xenografts (PDTXs). We demonstrate that chlorambucil inflicts replication-associated DNA double-strand breaks (DSBs), similarly to cisplatin, and we identify ATR, FANCD2 and the SNM1A nuclease as determinants of sensitivity to both drugs. Importantly, chlorambucil is substantially less toxic to normal cells and tissues in vitro and in vivo relative to cisplatin. Because chlorambucil and cisplatin are equally effective inhibitors of BRCA2-compromised tumours, our results indicate that chlorambucil has a higher therapeutic index than cisplatin in targeting BRCA-deficient tumours.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/deficiency
- BRCA2 Protein/deficiency
- Cell Line, Tumor
- Chlorambucil/pharmacology
- Cricetinae
- Drug Delivery Systems
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Male
- Mice
- Mice, SCID
- Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors
- Peroxisome Proliferator-Activated Receptors/metabolism
- Phthalazines/pharmacology
- Piperazines/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Eliana MC Tacconi
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Sophie Badie
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Giuliana De Gregoriis
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Timo Reisländer
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Xianning Lai
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Manuela Porru
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Cecilia Folio
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - John Moore
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Arnaud Kopp
- Institut de Génétique et de Biologie Cellulaire et Moléculaire (IGBMC)Inserm U1258, CNRS (UMR 7104)Université de StrasbourgIllkirchFrance
| | - Júlia Baguña Torres
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Deborah Sneddon
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Marcus Green
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Simon Dedic
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Jonathan W Lee
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Ankita Sati Batra
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Oscar M Rueda
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Alejandra Bruna
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Carlo Leonetti
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Carlos Caldas
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Bart Cornelissen
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Laurent Brino
- Institut de Génétique et de Biologie Cellulaire et Moléculaire (IGBMC)Inserm U1258, CNRS (UMR 7104)Université de StrasbourgIllkirchFrance
| | - Anderson Ryan
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Annamaria Biroccio
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Madalena Tarsounas
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| |
Collapse
|
46
|
Aalbersberg EA, Rossi MM, de Wit – van der Veen LJ, Walraven I, van den Heuvel MM, Sonke JJ, Belderbos JS, Vogel WV. Pre-hydration in cisplatin-based CCRT: Effects on tumour concentrations and treatment outcome. Radiother Oncol 2019; 134:30-36. [DOI: 10.1016/j.radonc.2019.01.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 10/27/2022]
|
47
|
Multiple ABCB1 transcriptional fusions in drug resistant high-grade serous ovarian and breast cancer. Nat Commun 2019; 10:1295. [PMID: 30894541 PMCID: PMC6426934 DOI: 10.1038/s41467-019-09312-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/28/2019] [Indexed: 12/24/2022] Open
Abstract
ABCB1 encodes Multidrug Resistance protein (MDR1), an ATP-binding cassette member involved in the cellular efflux of chemotherapeutic drugs. Here we report that ovarian and breast samples from chemotherapy treated patients are positive for multiple transcriptional fusions involving ABCB1, placing it under the control of a strong promoter while leaving its open reading frame intact. We identified 15 different transcriptional fusion partners involving ABCB1, as well as patients with multiple distinct fusion events. The partner gene selected depended on its structure, promoter strength, and chromosomal proximity to ABCB1. Fusion positivity was strongly associated with the number of lines of MDR1-substrate chemotherapy given. MDR1 inhibition in a fusion positive ovarian cancer cell line increased sensitivity to paclitaxel more than 50-fold. Convergent evolution of ABCB1 fusion is therefore frequent in chemotherapy resistant recurrent ovarian cancer. As most currently approved PARP inhibitors (PARPi) are MDR1 substrates, prior chemotherapy may precondition resistance to PARPi. ABCB1 encodes Multidrug Resistance Protein which promotes efflux of chemotherapeutic and targeted agents. Here, in breast and ovarian cancer the authors identify multiple transcriptional fusion partners involving ABCB1 that are associated with treatment failure and previous treatment regimens.
Collapse
|
48
|
ERβ modulates genistein’s cisplatin-enhancing activities in breast cancer MDA-MB-231 cells via P53-independent pathway. Mol Cell Biochem 2019; 456:205-216. [DOI: 10.1007/s11010-019-03505-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/01/2019] [Indexed: 11/25/2022]
|
49
|
Mentoor I, Engelbrecht AM, Nell T. Fatty acids: Adiposity and breast cancer chemotherapy, a bad synergy? Prostaglandins Leukot Essent Fatty Acids 2019; 140:18-33. [PMID: 30553399 DOI: 10.1016/j.plefa.2018.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023]
Abstract
Globally, breast cancer continues to be a major concern in women's health. Lifestyle related risk factors, specifically excess adipose tissue (adiposity) has reached epidemic proportions and has been identified as a major risk factor in the development of breast cancer. Dysfunctional adipose tissue has evoked research focusing on its association with metabolic-related conditions, breast cancer risk and progression. Adipose dysfunction in coordination with immune cells and inflammation, are responsible for accelerated cell growth and survival of cancer cells. Recently, evidence also implicates adiposity as a potential risk factor for chemotherapy resistance. Chemotherapeutic agents have been shown to negatively impact adipose tissue. Since adipose tissue is a major storage site for fatty acids, it is not unlikely that these negative effects may disrupt adipose tissue homeostasis. It is therefore argued that fatty acid composition may be altered due to the chemotherapeutic pharmacokinetics, which in turn could have severe health related outcomes. The underlying molecular mechanisms elucidating the effects of fatty acid composition in adiposity-linked drug resistance are still unclear and under explored. This review focuses on the potential role of adiposity in breast cancer and specifically emphasizes the role of fatty acids in cancer progression and treatment resistance.
Collapse
Affiliation(s)
- Ilze Mentoor
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - A-M Engelbrecht
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa
| | - Theo Nell
- Department of Physiological Sciences, Faculty of Sciences, Stellenbosch University Main Campus, Stellenbosch 7600, Western Cape, Republic of South Africa.
| |
Collapse
|
50
|
Tsai HC, Chang AC, Tsai CH, Huang YL, Gan L, Chen CK, Liu SC, Huang TY, Fong YC, Tang CH. CCN2 promotes drug resistance in osteosarcoma by enhancing ABCG2 expression. J Cell Physiol 2018; 234:9297-9307. [PMID: 30317661 DOI: 10.1002/jcp.27611] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023]
Abstract
In recent years, osteosarcoma survival rates have failed to improve significantly with conventional treatment modalities because of the development of chemotherapeutic resistance. The human breast cancer resistance protein/ATP binding cassette subfamily G member 2 (BCRP/ABCG2), a member of the ATP-binding cassette family, uses ATP hydrolysis to expel xenobiotics and chemotherapeutics from cells. CCN family member 2 (CCN2) is a secreted protein that modulates the biological function of cancer cells, enhanced ABCG2 protein expression and activation in this study via the α6β1 integrin receptor and increased osteosarcoma cell viability. CCN2 treatment downregulated miR-519d expression, which promoted ABCG2 expression. In a mouse xenograft model, knockdown of CCN2 expression increased the therapeutic effect of doxorubicin, which was reversed by ABCG2 overexpression. Our data show that CCN2 increases ABCG2 expression and promotes drug resistance through the α6β1 integrin receptor, whereas CCN2 downregulates miR-519d. CCN2 inhibition may represent a new therapeutic concept in osteosarcoma.
Collapse
Affiliation(s)
- Hsiao-Chi Tsai
- Department of Scientific Education, Qinghai Red Cross Hospital, Qinghai, China
| | - An-Chen Chang
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- School of Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Lijun Gan
- Department of Cardiology, Qinghai Red Cross Hospital, Qinghai, China
| | - Chi-Kuan Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Pathology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shih-Chia Liu
- Department of Orthopaedics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Te-Yang Huang
- Department of Orthopaedics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Chin Fong
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan.,Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|