1
|
Gedeonová D, Bianchi C, Štembírek J, Hrdinka M, Chyra Z, Buchtová M, Hurník P, Blažek T, Režnarová J. BRCA1 and BRCA2 as prognostic markers in oral squamous cell carcinoma: a minireview. Front Oncol 2025; 15:1528822. [PMID: 40224184 PMCID: PMC11986421 DOI: 10.3389/fonc.2025.1528822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC), a subset of head and neck cancers, primarily originates in the epithelial tissues of the oral cavity. Despite advancements in treatment, the mortality rate for OSCC remains around 50%, underscoring the urgent need for improved prognostic markers. This review explores the role of the BRCA1 and BRCA2 genes-traditionally associated with breast and ovarian cancers-in the context of OSCC. We discuss the molecular pathways involving BRCA genes, their potential as diagnostics and prognostic biomarkers, and their implications for personalized treatment strategies, including addressing chemotherapy resistance. Furthermore, this review emphasizes the significance of genome stability in cancer progression and examines both current and emerging methodologies for detecting BRCA mutations in OSCC patients. Despite limited prevalence of BRCA mutations in OSCC compared to other cancers, their role in DNA repair and therapeutic response underscores their potential as clinical biomarkers. However, standardized, multicenter studies are still needed to validate their utility in OSCC management. A better understanding of the role of BRCA genes in OSCC could pave the way for more effective therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Dominika Gedeonová
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Department of Craniofacial Surgery, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Claretta Bianchi
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Health Research Centre, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jan Štembírek
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Department of Craniofacial Surgery, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| | - Matouš Hrdinka
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Health Research Centre, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Zuzana Chyra
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czechia
- Department of Hematology, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Marcela Buchtová
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Pavel Hurník
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Tomáš Blažek
- Clinic of Oncology, University Hospital Ostrava, Ostrava, Czechia
| | - Jana Režnarová
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Health Research Centre, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| |
Collapse
|
2
|
Reddy T, Puri A, Guzman-Rojas L, Thomas C, Qian W, Zhou J, Zhao H, Mahboubi B, Oo A, Cho YJ, Kim B, Thaiparambil J, Rosato R, Martinez KO, Chervo MF, Ayerbe C, Giese N, Wink D, Lockett S, Wong S, Chang J, Krishnamurthy S, Yam C, Moulder S, Piwnica-Worms H, Meric-Bernstam F, Chang J. NOS inhibition sensitizes metaplastic breast cancer to PI3K inhibition and taxane therapy via c-JUN repression. Nat Commun 2024; 15:10737. [PMID: 39737957 PMCID: PMC11685991 DOI: 10.1038/s41467-024-54651-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/19/2024] [Indexed: 01/01/2025] Open
Abstract
Metaplastic breast cancer (MpBC) is a highly chemoresistant subtype of breast cancer with no standardized therapy options. A clinical study in anthracycline-refractory MpBC patients suggested that nitric oxide synthase (NOS) inhibitor NG-monomethyl-l-arginine (L-NMMA) may augment anti-tumor efficacy of taxane. We report that NOS blockade potentiated response of human MpBC cell lines and tumors to phosphoinositide 3-kinase (PI3K) inhibitor alpelisib and taxane. Mechanistically, NOS blockade leads to a decrease in the S-nitrosylation of c-Jun NH2-terminal kinase (JNK)/c-Jun complex to repress its transcriptional output, leading to enhanced tumor differentiation and associated chemosensitivity. As a result, combined NOS and PI3K inhibition with taxane targets MpBC stem cells and improves survival in patient-derived xenograft models relative to single-/dual-agent therapy. Similarly, biopsies from MpBC tumors that responded to L-NMMA+taxane therapy showed a post-treatment reversal of epithelial-to-mesenchymal transition and decreased stemness. Our findings suggest that combined inhibition of iNOS and PI3K is a unique strategy to decrease chemoresistance and improve clinical outcomes in MpBC.
Collapse
Affiliation(s)
- Tejaswini Reddy
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Akshjot Puri
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | | | - Christoforos Thomas
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Wei Qian
- Houston Methodist Research Institute, Houston, TX, USA
| | - Jianying Zhou
- Houston Methodist Research Institute, Houston, TX, USA
| | - Hong Zhao
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Bijan Mahboubi
- Adams School of Dentistry, University of North Carolina, Chapel Hill, USA
| | - Adrian Oo
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Young-Jae Cho
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Baek Kim
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | | | | - Camila Ayerbe
- McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Noah Giese
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - David Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institute of Health, Frederick, MD, USA
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Stephen Wong
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neal Cancer Center, Houston, TX, USA
| | - Jeffrey Chang
- McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Clinton Yam
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Jenny Chang
- Houston Methodist Research Institute, Houston, TX, USA.
- Houston Methodist Neal Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Bi R, Chen L, Huang M, Qiao Z, Li Z, Fan G, Wang Y. Emerging strategies to overcome PARP inhibitors' resistance in ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189221. [PMID: 39571765 DOI: 10.1016/j.bbcan.2024.189221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
The utilization of PARP inhibitors (PARPis) has significantly improved the prognosis for ovarian cancer patients. However, as the use of PARPis increases, the issue of PARPi resistance has become more prominent. Prolonged usage of PARPis can lead to the development of resistance in ovarian cancer, often mediated by mechanisms such as homologous recombination (HR) recovery, ultimately resulting in cancer relapse. Overcoming PARPi resistance in ovarian cancer is a pressing concern, aiming to enhance the clinical benefits of PARPi treatment and delay disease recurrence. Here, we summarize the mechanisms underlying PARPi resistance, methods for analyzing resistance, and strategies for overcoming it. Our goal is to inspire the development of more cost-effective and convenient methods for analyzing resistance mechanisms, as well as safer and more effective strategies to overcome resistance. These advancements can contribute to developing personalized approaches for treating ovarian cancer.
Collapse
Affiliation(s)
- Ruomeng Bi
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Li Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Mei Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhi Qiao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhen Li
- Clinical Research Unit, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| | - Yu Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
4
|
Saville KM, Al-Rahahleh RQ, Siddiqui AH, Andrews ME, Roos WP, Koczor CA, Andrews JF, Hayat F, Migaud ME, Sobol RW. Oncometabolite 2-hydroxyglutarate suppresses basal protein levels of DNA polymerase beta that enhances alkylating agent and PARG inhibition induced cytotoxicity. DNA Repair (Amst) 2024; 140:103700. [PMID: 38897003 PMCID: PMC11239280 DOI: 10.1016/j.dnarep.2024.103700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024]
Abstract
Mutations in isocitrate dehydrogenase isoform 1 (IDH1) are primarily found in secondary glioblastoma (GBM) and low-grade glioma but are rare in primary GBM. The standard treatment for GBM includes radiation combined with temozolomide, an alkylating agent. Fortunately, IDH1 mutant gliomas are sensitive to this treatment, resulting in a more favorable prognosis. However, it's estimated that up to 75 % of IDH1 mutant gliomas will progress to WHO grade IV over time and develop resistance to alkylating agents. Therefore, understanding the mechanism(s) by which IDH1 mutant gliomas confer sensitivity to alkylating agents is crucial for developing targeted chemotherapeutic approaches. The base excision repair (BER) pathway is responsible for repairing most base damage induced by alkylating agents. Defects in this pathway can lead to hypersensitivity to these agents due to unresolved DNA damage. The coordinated assembly and disassembly of BER protein complexes are essential for cell survival and for maintaining genomic integrity following alkylating agent exposure. These complexes rely on poly-ADP-ribose formation, an NAD+-dependent post-translational modification synthesized by PARP1 and PARP2 during the BER process. At the lesion site, poly-ADP-ribose facilitates the recruitment of XRCC1. This scaffold protein helps assemble BER proteins like DNA polymerase beta (Polβ), a bifunctional DNA polymerase containing both DNA synthesis and 5'-deoxyribose-phosphate lyase (5'dRP lyase) activity. Here, we confirm that IDH1 mutant glioma cells have defective NAD+ metabolism, but still produce sufficient nuclear NAD+ for robust PARP1 activation and BER complex formation in response to DNA damage. However, the overproduction of 2-hydroxyglutarate, an oncometabolite produced by the IDH1 R132H mutant protein, suppresses BER capacity by reducing Polβ protein levels. This defines a novel mechanism by which the IDH1 mutation in gliomas confers cellular sensitivity to alkylating agents and to inhibitors of the poly-ADP-ribose glycohydrolase, PARG.
Collapse
Affiliation(s)
- Kate M Saville
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Rasha Q Al-Rahahleh
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Aisha H Siddiqui
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Morgan E Andrews
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Wynand P Roos
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Christopher A Koczor
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Joel F Andrews
- Department Biochemistry and Molecular Biology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Faisal Hayat
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Marie E Migaud
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Robert W Sobol
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States.
| |
Collapse
|
5
|
Tai Y, Chow A, Han S, Coker C, Ma W, Gu Y, Estrada Navarro V, Kandpal M, Hibshoosh H, Kalinsky K, Manova-Todorova K, Safonov A, Walsh EM, Robson M, Norton L, Baer R, Merghoub T, Biswas AK, Acharyya S. FLT1 activation in cancer cells promotes PARP-inhibitor resistance in breast cancer. EMBO Mol Med 2024; 16:1957-1980. [PMID: 38956205 PMCID: PMC11319505 DOI: 10.1038/s44321-024-00094-2] [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: 09/06/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Acquired resistance to PARP inhibitors (PARPi) remains a treatment challenge for BRCA1/2-mutant breast cancer that drastically shortens patient survival. Although several resistance mechanisms have been identified, none have been successfully targeted in the clinic. Using new PARPi-resistance models of Brca1- and Bard1-mutant breast cancer generated in-vivo, we identified FLT1 (VEGFR1) as a driver of resistance. Unlike the known role of VEGF signaling in angiogenesis, we demonstrate a novel, non-canonical role for FLT1 signaling that protects cancer cells from PARPi in-vivo through a combination of cell-intrinsic and cell-extrinsic pathways. We demonstrate that FLT1 blockade suppresses AKT activation, increases tumor infiltration of CD8+ T cells, and causes dramatic regression of PARPi-resistant breast tumors in a T-cell-dependent manner. Moreover, PARPi-resistant tumor cells can be readily re-sensitized to PARPi by targeting Flt1 either genetically (Flt1-suppression) or pharmacologically (axitinib). Importantly, a retrospective series of breast cancer patients treated with PARPi demonstrated shorter progression-free survival in cases with FLT1 activation at pre-treatment. Our study therefore identifies FLT1 as a potential therapeutic target in PARPi-resistant, BRCA1/2-mutant breast cancer.
Collapse
Affiliation(s)
- Yifan Tai
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Biology, McGill University, Montreal, Quebec, QC, H3G0B1, Canada
| | - Angela Chow
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Seoyoung Han
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Jacobs School of Medicine, University of Buffalo, New York, NY, USA
| | - Courtney Coker
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Wanchao Ma
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Yifan Gu
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Valeria Estrada Navarro
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Manoj Kandpal
- Centre for Clinical and Translational Science, Rockefeller University Hospital, 1230 York Ave, New York, NY, 10065, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Kevin Kalinsky
- Winship Cancer Institute of Emory University, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Katia Manova-Todorova
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Anton Safonov
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Elaine M Walsh
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Department of Medicine, Georgetown Lombardi Comprehensive Cancer Center, 3800 Reservoir Rd, NW, Washington DC, 20007, USA
| | - Mark Robson
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Larry Norton
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Richard Baer
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Taha Merghoub
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Anup K Biswas
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Swarnali Acharyya
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Ave, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
6
|
Chen X, Yan Y, Liu Y, Yi Q, Xu Z. Tabersonine Enhances Olaparib Sensitivity through FHL1-Mediated Epithelial-Mesenchymal Transition in an Ovarian Tumor. JOURNAL OF NATURAL PRODUCTS 2024; 87:837-848. [PMID: 38417401 DOI: 10.1021/acs.jnatprod.3c01056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
Ovarian cancer (OVC) is one of the most aggressive gynecological malignancies worldwide. Although olaparib treatment has shown favorable outcomes against the treatment of OVC, its effectiveness remains limited in some OVC patients. Investigating new strategies to improve the therapeutic efficacy of olaparib against OVC is imperative. Our study identified tabersonine, a natural indole alkaloid, for its potential to increase the chemosensitivity of olaparib in OVC. The combined treatment of olaparib and tabersonine synergistically inhibited cell proliferation in OVC cells and suppressed tumor growth in A2780 xenografts. The combined treatment effectively suppressed epithelial-mesenchymal transition (EMT) by altering the expression of E-cadherin, N-cadherin, and vimentin and induced DNA damage responses. Integrating quantitative proteomics, FHL1 was identified as a potential regulator to modulate EMT after tabersonine treatment. Increased expression of FHL1 was induced by tabersonine treatment, while downregulation of FHL1 reversed the inhibitory effects of tabersonine on OVC cells by mediating EMT. In vivo findings further reflected that the combined treatment of tabersonine and olaparib significantly inhibited tumor growth and OVC metastasis through upregulation of FHL1. Our findings reveal the role of tabersonine in improving the sensitivity of olaparib in OVC through FHL1-mediated EMT, suggesting that tabersonine holds promise for future application in OVC treatment.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Qiaoli Yi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
7
|
Murphy KC, Ruscetti M. Advances in Making Cancer Mouse Models More Accessible and Informative through Non-Germline Genetic Engineering. Cold Spring Harb Perspect Med 2024; 14:a041348. [PMID: 37277206 PMCID: PMC10982712 DOI: 10.1101/cshperspect.a041348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Genetically engineered mouse models (GEMMs) allow for modeling of spontaneous tumorigenesis within its native microenvironment in mice and have provided invaluable insights into mechanisms of tumorigenesis and therapeutic strategies to treat human disease. However, as their generation requires germline manipulation and extensive animal breeding that is time-, labor-, and cost-intensive, traditional GEMMs are not accessible to most researchers, and fail to model the full breadth of cancer-associated genetic alterations and therapeutic targets. Recent advances in genome-editing technologies and their implementation in somatic tissues of mice have ushered in a new class of mouse models: non-germline GEMMs (nGEMMs). nGEMM approaches can be leveraged to generate somatic tumors de novo harboring virtually any individual or group of genetic alterations found in human cancer in a mouse through simple procedures that do not require breeding, greatly increasing the accessibility and speed and scale on which GEMMs can be produced. Here we describe the technologies and delivery systems used to create nGEMMs and highlight new biological insights derived from these models that have rapidly informed functional cancer genomics, precision medicine, and immune oncology.
Collapse
Affiliation(s)
- Katherine C Murphy
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA;
- Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
- Cancer Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
8
|
Schuhwerk H, Brabletz T. Mutual regulation of TGFβ-induced oncogenic EMT, cell cycle progression and the DDR. Semin Cancer Biol 2023; 97:86-103. [PMID: 38029866 DOI: 10.1016/j.semcancer.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/06/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
TGFβ signaling and the DNA damage response (DDR) are two cellular toolboxes with a strong impact on cancer biology. While TGFβ as a pleiotropic cytokine affects essentially all hallmarks of cancer, the multifunctional DDR mostly orchestrates cell cycle progression, DNA repair, chromatin remodeling and cell death. One oncogenic effect of TGFβ is the partial activation of epithelial-to-mesenchymal transition (EMT), conferring invasiveness, cellular plasticity and resistance to various noxae. Several reports show that both individual networks as well as their interface affect chemo-/radiotherapies. However, the underlying mechanisms remain poorly resolved. EMT often correlates with TGFβ-induced slowing of proliferation, yet numerous studies demonstrate that particularly the co-activated EMT transcription factors counteract anti-proliferative signaling in a partially non-redundant manner. Collectively, evidence piled up over decades underscore a multifaceted, reciprocal inter-connection of TGFβ signaling / EMT with the DDR / cell cycle progression, which we will discuss here. Altogether, we conclude that full cell cycle arrest is barely compatible with the propagation of oncogenic EMT traits and further propose that 'EMT-linked DDR plasticity' is a crucial, yet intricate facet of malignancy, decisively affecting metastasis formation and therapy resistance.
Collapse
Affiliation(s)
- Harald Schuhwerk
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
9
|
Kim H, Aliar K, Tharmapalan P, McCloskey CW, Kuttanamkuzhi A, Grünwald BT, Palomero L, Mahendralingam MJ, Waas M, Mer AS, Elliott MJ, Zhang B, Al-Zahrani KN, Langille ER, Parsons M, Narala S, Hofer S, Waterhouse PD, Hakem R, Haibe-Kains B, Kislinger T, Schramek D, Cescon DW, Pujana MA, Berman HK, Khokha R. Differential DNA damage repair and PARP inhibitor vulnerability of the mammary epithelial lineages. Cell Rep 2023; 42:113256. [PMID: 37847590 DOI: 10.1016/j.celrep.2023.113256] [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: 02/03/2023] [Revised: 09/02/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
It is widely assumed that all normal somatic cells can equally perform homologous recombination (HR) and non-homologous end joining in the DNA damage response (DDR). Here, we show that the DDR in normal mammary gland inherently depends on the epithelial cell lineage identity. Bioinformatics, post-irradiation DNA damage repair kinetics, and clonogenic assays demonstrated luminal lineage exhibiting a more pronounced DDR and HR repair compared to the basal lineage. Consequently, basal progenitors were far more sensitive to poly(ADP-ribose) polymerase inhibitors (PARPis) in both mouse and human mammary epithelium. Furthermore, PARPi sensitivity of murine and human breast cancer cell lines as well as patient-derived xenografts correlated with their molecular resemblance to the mammary progenitor lineages. Thus, mammary epithelial cells are intrinsically divergent in their DNA damage repair capacity and PARPi vulnerability, potentially influencing the clinical utility of this targeted therapy.
Collapse
Affiliation(s)
- Hyeyeon Kim
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Kazeera Aliar
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Pirashaanthy Tharmapalan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Curtis W McCloskey
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Barbara T Grünwald
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Luis Palomero
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Catalonia, Spain
| | - Mathepan J Mahendralingam
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Matthew Waas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Arvind S Mer
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mitchell J Elliott
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Bowen Zhang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Khalid N Al-Zahrani
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Ellen R Langille
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael Parsons
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Swami Narala
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stefan Hofer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Paul D Waterhouse
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2N2, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Miquel A Pujana
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Catalonia, Spain
| | - Hal K Berman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2N2, Canada.
| |
Collapse
|
10
|
Papatheodoridi A, Papamattheou E, Marinopoulos S, Ntanasis-Stathopoulos I, Dimitrakakis C, Giannos A, Kaparelou M, Liontos M, Dimopoulos MA, Zagouri F. Metaplastic Carcinoma of the Breast: Case Series of a Single Institute and Review of the Literature. Med Sci (Basel) 2023; 11:medsci11020035. [PMID: 37218987 DOI: 10.3390/medsci11020035] [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: 03/29/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/24/2023] Open
Abstract
Metaplastic carcinoma of the breast (MpBC) is a very rare and aggressive type of breast cancer. Data focusing on MpBC are limited. The aim of this study was to describe the clinicopathological features of MpBC and evaluate the prognosis of patients with MpBC. Eligible articles about MpBC were identified by searching CASES SERIES gov and the MEDLINE bibliographic database for the period of 1 January 2010 to 1 June 2021 with the keywords metaplastic breast cancer, mammary gland cancer, neoplasm, tumor, and metaplastic carcinoma. In this study, we also report 46 cases of MpBC stemming from our hospital. Survival rates, clinical behavior, and pathological characteristics were analyzed. Data from 205 patients were included for analysis. The mean age at diagnosis was 55 (14.7) years. The TNM stage at diagnosis was mostly stage II (58.5%) and most tumors were triple negative. The median overall survival was 66 (12-118) months, and the median disease-free survival was 56.8 (11-102) months. Multivariate Cox regression analysis revealed that surgical treatment was associated with decreased risk of death (hazard ratio 0.11, 95% confidence interval 0.02-0.54, p = 0.01) while advanced TNM stage was associated with increased risk of death (hazard ratio 1.5, 95% confidence interval 1.04-2.28, p = 0.03). Our results revealed that surgical treatment and TNM stage were the only independent risk factors related to patients' overall survival.
Collapse
Affiliation(s)
- Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
- Department of Physiology, Medical School of National and Kapodistrian University of Athens, 115 28 Athens, Greece
| | - Eleni Papamattheou
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
- 1st Department of Obstetrics & Gynecology, "Alexandra" Hospital, Medical School, University of Athens, 115 28 Athens, Greece
| | - Spyridon Marinopoulos
- 1st Department of Obstetrics & Gynecology, "Alexandra" Hospital, Medical School, University of Athens, 115 28 Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
| | - Constantine Dimitrakakis
- 1st Department of Obstetrics & Gynecology, "Alexandra" Hospital, Medical School, University of Athens, 115 28 Athens, Greece
| | - Aris Giannos
- 1st Department of Obstetrics & Gynecology, "Alexandra" Hospital, Medical School, University of Athens, 115 28 Athens, Greece
| | - Maria Kaparelou
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
| | - Michalis Liontos
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, 115 28 Athens, Greece
| |
Collapse
|
11
|
Taylor AK, Kosoff D, Emamekhoo H, Lang JM, Kyriakopoulos CE. PARP inhibitors in metastatic prostate cancer. Front Oncol 2023; 13:1159557. [PMID: 37168382 PMCID: PMC10165068 DOI: 10.3389/fonc.2023.1159557] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
Poly-ADP ribose polymerase inhibitors (PARPi) are an emerging therapeutic option for the treatment of prostate cancer. Their primary mechanism of action is via induction of synthetic lethality in cells with underlying deficiencies in homologous recombination repair (HRR). In men with metastatic castrate-resistant prostate cancer (mCRPC) and select HRR pathway alterations, PARPi treatment has been shown to induce objective tumor responses as well as improve progression free and overall survival. Presently, there are two PARPi, olaparib and rucaparib, that are FDA approved in the treatment of mCRPC. Ongoing research is focused on identifying which HRR alterations are best suited to predict response to PARPi so that these therapies can be most effectively utilized in the clinic. While resistance to PARPi remains a concern, combination therapies may represent a mechanism to overcome or delay resistance.
Collapse
Affiliation(s)
- Amy K. Taylor
- Department of Medicine, University of Wisconsin, Madison, WI, United States
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, WI, United States
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
| | - Hamid Emamekhoo
- Department of Medicine, University of Wisconsin, Madison, WI, United States
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin, Madison, WI, United States
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
| | - Christos E. Kyriakopoulos
- Department of Medicine, University of Wisconsin, Madison, WI, United States
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States
| |
Collapse
|
12
|
Dong S, Fang Y, Syed S. A rare case of advanced metaplastic breast carcinoma with response to treatment with Sacituzumab govitecan. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2023. [DOI: 10.1016/j.cpccr.2023.100239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
|
13
|
Rajawat J, Awasthi P, Banerjee M. PARP inhibitor olaparib induced differential protein expression in cervical cancer cells. J Proteomics 2023; 275:104823. [PMID: 36646275 DOI: 10.1016/j.jprot.2023.104823] [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: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
PARP inhibitors are a potential class of chemotherapeutic drugs but PARP inhibitor response has not been explored systematically. We lack a specific understanding of the subset of the proteome preferentially modified in various cancers by PARP inhibitors. Implications of PARP inhibitor and PARP1 in cervical cancer treatment and resistance are not fully elucidated. We conducted a mass spectrometry-based proteomic analysis of cervical cancer Hela cells treated with olaparib. We aimed to identify the alteration in the protein signaling pathway induced by PARP inhibitors beyond the DNA damage response pathway. Our data demonstrate a significant reduction in PARP activity and enhanced cell death after olaparib treatment. We further observed articulated proteomic changes with a significant enrichment of proteins in diverse cellular processes. The differentially expressed proteins were predominantly associated with RNA metabolism, mRNA splicing, processing, and RNA binding. Our data also identified proteins that could probably contribute to survival mechanisms resulting in resistance to PARP inhibitors. Hence, we put forth the overview of proteomic changes induced by PARP inhibitor olaparib in cervical cancer cells. This study highlights the significant proteins modified during PARP inhibition and thus could be a probable target for combination therapies with PARP inhibitors in cervical cancer. SIGNIFICANCE.
Collapse
Affiliation(s)
- Jyotika Rajawat
- Molecular & Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, U.P, India
| | - Poorwa Awasthi
- CSIR-Indian Institute of Toxicology Research, Lucknow 226001, U.P, India
| | - Monisha Banerjee
- Molecular & Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, U.P, India..
| |
Collapse
|
14
|
Kinnel B, Singh SK, Oprea-Ilies G, Singh R. Targeted Therapy and Mechanisms of Drug Resistance in Breast Cancer. Cancers (Basel) 2023; 15:1320. [PMID: 36831661 PMCID: PMC9954028 DOI: 10.3390/cancers15041320] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Breast cancer is the most common cause of cancer-related death in women worldwide. Multidrug resistance (MDR) has been a large hurdle in reducing BC death rates. The drug resistance mechanisms include increased drug efflux, enhanced DNA repair, senescence escape, epigenetic alterations, tumor heterogeneity, tumor microenvironment (TME), and the epithelial-to-mesenchymal transition (EMT), which make it challenging to overcome. This review aims to explain the mechanisms of resistance in BC further, identify viable drug targets, and elucidate how those targets relate to the progression of BC and drug resistance.
Collapse
Affiliation(s)
- Briana Kinnel
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Santosh Kumar Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Gabriela Oprea-Ilies
- Department of Pathology & Laboratory Medicine, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
15
|
Thomas HR, Hu B, Boyraz B, Johnson A, Bossuyt VI, Spring L, Jimenez RB. Metaplastic breast cancer: A review. Crit Rev Oncol Hematol 2023; 182:103924. [PMID: 36696934 DOI: 10.1016/j.critrevonc.2023.103924] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 12/19/2022] [Accepted: 01/20/2023] [Indexed: 01/23/2023] Open
Abstract
Metaplastic breast cancer (MpBC) is an uncommon aggressive malignancy that is associated with a poor prognosis. Due to its rarity, the relationships between the clinical and pathological features of MpBC, treatment approach, and clinical outcomes remain underexplored. In the following review article, we synthesize the existing data on the clinical, pathological and genomic features, management, and outcomes of MpBC. We also identify potential targets for future clinical trials.
Collapse
Affiliation(s)
- Horatio R Thomas
- Department of Radiation Oncology, University of California, San Francisco, United States.
| | - Bonnie Hu
- Department of Radiation Oncology, Massachusetts General Hospital, United States
| | - Baris Boyraz
- Department of Pathology, Massachusetts General Hospital, United States
| | - Andrew Johnson
- Department of Radiation Oncology, Massachusetts General Hospital, United States
| | - Veerle I Bossuyt
- Department of Pathology, Massachusetts General Hospital, United States
| | - Laura Spring
- Department of Medicine, Division of Medical Oncology, Massachusetts General Hospital, United States
| | - Rachel B Jimenez
- Department of Radiation Oncology, Massachusetts General Hospital, United States
| |
Collapse
|
16
|
Wen J, Wang S, Guo R, Liu D. CSF1R inhibitors are emerging immunotherapeutic drugs for cancer treatment. Eur J Med Chem 2023; 245:114884. [PMID: 36335744 DOI: 10.1016/j.ejmech.2022.114884] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/13/2022] [Accepted: 10/22/2022] [Indexed: 11/16/2022]
|
17
|
Pettitt SJ, Ryan CJ, Lord CJ. Exploiting Cancer Synthetic Lethality in Cancer-Lessons Learnt from PARP Inhibitors. Cancer Treat Res 2023; 186:13-23. [PMID: 37978128 DOI: 10.1007/978-3-031-30065-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
PARP inhibitors now have proven utility in the treatment of homologous recombination (HR) defective cancers. These drugs, and the synthetic lethality effect they exploit, have not only taught us how to approach the treatment of HR defective cancers but have also illuminated how resistance to a synthetic lethal approach can occur, how cancer-associated synthetic lethal effects are perhaps more complex than we imagine, how the better use of biomarkers could improve the success of treatment and even how drug resistance might be targeted. Here, we discuss some of the lessons learnt from the study of PARP inhibitor synthetic lethality and how these lessons might have wider application. Specifically, we discuss the concept of synthetic lethal penetrance, phenocopy effects in cancer such as BRCAness, synthetic lethal resistance, the polygenic and complex nature of synthetic lethal interactions, how evolutionary double binds could be exploited in treatment as well as future horizons for the field.
Collapse
Affiliation(s)
- Stephen J Pettitt
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Colm J Ryan
- School of Computer Science and Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
18
|
DNA Damage Response in Cancer Therapy and Resistance: Challenges and Opportunities. Int J Mol Sci 2022; 23:ijms232314672. [PMID: 36499000 PMCID: PMC9735783 DOI: 10.3390/ijms232314672] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Resistance to chemo- and radiotherapy is a common event among cancer patients and a reason why new cancer therapies and therapeutic strategies need to be in continuous investigation and development. DNA damage response (DDR) comprises several pathways that eliminate DNA damage to maintain genomic stability and integrity, but different types of cancers are associated with DDR machinery defects. Many improvements have been made in recent years, providing several drugs and therapeutic strategies for cancer patients, including those targeting the DDR pathways. Currently, poly (ADP-ribose) polymerase inhibitors (PARP inhibitors) are the DDR inhibitors (DDRi) approved for several cancers, including breast, ovarian, pancreatic, and prostate cancer. However, PARPi resistance is a growing issue in clinical settings that increases disease relapse and aggravate patients' prognosis. Additionally, resistance to other DDRi is also being found and investigated. The resistance mechanisms to DDRi include reversion mutations, epigenetic modification, stabilization of the replication fork, and increased drug efflux. This review highlights the DDR pathways in cancer therapy, its role in the resistance to conventional treatments, and its exploitation for anticancer treatment. Biomarkers of treatment response, combination strategies with other anticancer agents, resistance mechanisms, and liabilities of treatment with DDR inhibitors are also discussed.
Collapse
|
19
|
Zingg D, Bhin J, Yemelyanenko J, Kas SM, Rolfs F, Lutz C, Lee JK, Klarenbeek S, Silverman IM, Annunziato S, Chan CS, Piersma SR, Eijkman T, Badoux M, Gogola E, Siteur B, Sprengers J, de Klein B, de Goeij-de Haas RR, Riedlinger GM, Ke H, Madison R, Drenth AP, van der Burg E, Schut E, Henneman L, van Miltenburg MH, Proost N, Zhen H, Wientjens E, de Bruijn R, de Ruiter JR, Boon U, de Korte-Grimmerink R, van Gerwen B, Féliz L, Abou-Alfa GK, Ross JS, van de Ven M, Rottenberg S, Cuppen E, Chessex AV, Ali SM, Burn TC, Jimenez CR, Ganesan S, Wessels LFA, Jonkers J. Truncated FGFR2 is a clinically actionable oncogene in multiple cancers. Nature 2022; 608:609-617. [PMID: 35948633 PMCID: PMC9436779 DOI: 10.1038/s41586-022-05066-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/03/2022] [Indexed: 12/13/2022]
Abstract
Somatic hotspot mutations and structural amplifications and fusions that affect fibroblast growth factor receptor 2 (encoded by FGFR2) occur in multiple types of cancer1. However, clinical responses to FGFR inhibitors have remained variable1–9, emphasizing the need to better understand which FGFR2 alterations are oncogenic and therapeutically targetable. Here we apply transposon-based screening10,11 and tumour modelling in mice12,13, and find that the truncation of exon 18 (E18) of Fgfr2 is a potent driver mutation. Human oncogenomic datasets revealed a diverse set of FGFR2 alterations, including rearrangements, E1–E17 partial amplifications, and E18 nonsense and frameshift mutations, each causing the transcription of E18-truncated FGFR2 (FGFR2ΔE18). Functional in vitro and in vivo examination of a compendium of FGFR2ΔE18 and full-length variants pinpointed FGFR2-E18 truncation as single-driver alteration in cancer. By contrast, the oncogenic competence of FGFR2 full-length amplifications depended on a distinct landscape of cooperating driver genes. This suggests that genomic alterations that generate stable FGFR2ΔE18 variants are actionable therapeutic targets, which we confirmed in preclinical mouse and human tumour models, and in a clinical trial. We propose that cancers containing any FGFR2 variant with a truncated E18 should be considered for FGFR-targeted therapies. Truncation of exon 18 of FGFR2 (FGFR2ΔE18) is a potent driver mutation in mice and humans, and FGFR-targeted therapy should be considered for patients with cancer expressing stable FGFR2ΔE18 variants.
Collapse
Affiliation(s)
- Daniel Zingg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Jinhyuk Bhin
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Julia Yemelyanenko
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Sjors M Kas
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Frank Rolfs
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands.,OncoProteomics Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | | | - Sjoerd Klarenbeek
- Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Stefano Annunziato
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Chang S Chan
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine and Pharmacology, Rutgers University, Piscataway, NJ, USA
| | - Sander R Piersma
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Timo Eijkman
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Madelon Badoux
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Ewa Gogola
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Bjørn Siteur
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Justin Sprengers
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bim de Klein
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Richard R de Goeij-de Haas
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gregory M Riedlinger
- Department of Medicine and Pharmacology, Rutgers University, Piscataway, NJ, USA.,Department of Pathology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Hua Ke
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Medicine and Pharmacology, Rutgers University, Piscataway, NJ, USA
| | | | - Anne Paulien Drenth
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Eva Schut
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Linda Henneman
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands.,Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Martine H van Miltenburg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Natalie Proost
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Ellen Wientjens
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | - Roebi de Bruijn
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Julian R de Ruiter
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands.,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ute Boon
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Oncode Institute, Utrecht, The Netherlands
| | | | - Bastiaan van Gerwen
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Luis Féliz
- Incyte Biosciences International, Morges, Switzerland
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Medical College at Cornell University, New York, NY, USA
| | - Jeffrey S Ross
- Foundation Medicine, Cambridge, MA, USA.,Upstate University Hospital, Upstate Medical University, Syracuse, NY, USA
| | - Marieke van de Ven
- Mouse Clinic for Cancer and Aging, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sven Rottenberg
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Bern Center for Precision Medicine, University of Bern, Bern, Switzerland
| | - Edwin Cuppen
- Oncode Institute, Utrecht, The Netherlands.,Hartwig Medical Foundation, Amsterdam, The Netherlands.,Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | - Connie R Jimenez
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Shridar Ganesan
- Department of Medicine, Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA. .,Department of Medicine and Pharmacology, Rutgers University, Piscataway, NJ, USA.
| | - Lodewyk F A Wessels
- Oncode Institute, Utrecht, The Netherlands. .,Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands. .,Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
20
|
Margarido AS, Uceda-Castro R, Hahn K, de Bruijn R, Kester L, Hofland I, Lohuis J, Seinstra D, Broeks A, Jonkers J, Broekman MLD, Wesseling P, Vennin C, Vizoso M, van Rheenen J. Epithelial-to-Mesenchymal Transition Drives Invasiveness of Breast Cancer Brain Metastases. Cancers (Basel) 2022; 14:cancers14133115. [PMID: 35804890 PMCID: PMC9264851 DOI: 10.3390/cancers14133115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/30/2022] Open
Abstract
(1) Background: an increasing number of breast cancer patients develop lethal brain metastases (BM). The complete removal of these tumors by surgery becomes complicated when cells infiltrate into the brain parenchyma. However, little is known about the nature of these invading cells in breast cancer brain metastasis (BCBM). (2) Methods: we use intravital microscopy through a cranial window to study the behavior of invading cells in a mouse model of BCBM. (3) Results: we demonstrate that BCBM cells that escape from the metastatic mass and infiltrate into brain parenchyma undergo epithelial-to-mesenchymal transition (EMT). Moreover, cells undergoing EMT revert to an epithelial state when growing tumor masses in the brain. Lastly, through multiplex immunohistochemistry, we confirm the presence of these infiltrative cells in EMT in patient samples. (4) Conclusions: together, our data identify the critical role of EMT in the invasive behavior of BCBM, which warrants further consideration to target those cells when treating BCBM.
Collapse
Affiliation(s)
- Andreia S. Margarido
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Rebeca Uceda-Castro
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Kerstin Hahn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Roebi de Bruijn
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Lennart Kester
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (I.H.); (A.B.)
| | - Jeroen Lohuis
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Danielle Seinstra
- Department of Pathology, Amsterdam University Medical Centers/VUmc and Brain Tumor Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (D.S.); (P.W.)
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (I.H.); (A.B.)
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Marike L. D. Broekman
- Department of Neurosurgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Department of Neurosurgery, Haaglanden Medical Center, Lijnbaan, 2512 VA The Hague, The Netherlands
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers/VUmc and Brain Tumor Center Amsterdam, 1081 HV Amsterdam, The Netherlands; (D.S.); (P.W.)
- Laboratory for Childhood Cancer Pathology, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Claire Vennin
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
| | - Miguel Vizoso
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
- Correspondence: (M.V.); (J.v.R.)
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; (A.S.M.); (R.U.-C.); (K.H.); (R.d.B.); (L.K.); (J.L.); (J.J.); (C.V.)
- Correspondence: (M.V.); (J.v.R.)
| |
Collapse
|
21
|
Moyret-Lalle C, Prodhomme MK, Burlet D, Kashiwagi A, Petrilli V, Puisieux A, Seimiya H, Tissier A. Role of EMT in the DNA damage response, double-strand break repair pathway choice and its implications in cancer treatment. Cancer Sci 2022; 113:2214-2223. [PMID: 35534984 PMCID: PMC9277259 DOI: 10.1111/cas.15389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Numerous epithelial–mesenchymal transition (EMT) characteristics have now been demonstrated to participate in tumor development. Indeed, EMT is involved in invasion, acquisition of stem cell properties, and therapy‐associated resistance of cancer cells. Together, these mechanisms offer advantages in adapting to changes in the tumor microenvironment. However, recent findings have shown that EMT‐associated transcription factors (EMT‐TFs) may also be involved in DNA repair. A better understanding of the coordination between the DNA repair pathways and the role played by some EMT‐TFs in the DNA damage response (DDR) should pave the way for new treatments targeting tumor‐specific molecular vulnerabilities, which result in selective destruction of cancer cells. Here we review recent advances, providing novel insights into the role of EMT in the DDR and repair pathways, with a particular focus on the influence of EMT on cellular sensitivity to damage, as well as the implications of these relationships for improving the efficacy of cancer treatments.
Collapse
Affiliation(s)
- Caroline Moyret-Lalle
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Centre of Lyon, Lyon, France.,LabEx DEVweCAN, Université de Lyon, Lyon, France
| | - Mélanie K Prodhomme
- Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Delphine Burlet
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Centre of Lyon, Lyon, France
| | - Ayaka Kashiwagi
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Virginie Petrilli
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Centre of Lyon, Lyon, France
| | - Alain Puisieux
- Institut Curie, Versailles Saint-Quentin-en-Yvelines University, PSL Research University, Paris, France
| | - Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Agnès Tissier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Centre of Lyon, Lyon, France
| |
Collapse
|
22
|
Thirumal Kumar D, Udhaya Kumar S, Jain N, Sowmya B, Balsekar K, Siva R, Kamaraj B, Sidenna M, George Priya Doss C, Zayed H. Computational structural assessment of BReast CAncer type 1 susceptibility protein (BRCA1) and BRCA1-Associated Ring Domain protein 1 (BARD1) mutations on the protein-protein interface. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:375-397. [PMID: 35534113 DOI: 10.1016/bs.apcsb.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Breast cancer type 1 susceptibility protein (BRCA1) is closely related to the BRCA2 (breast cancer type 2 susceptibility protein) and BARD1 (BRCA1-associated RING domain-1) proteins. The homodimers were formed through their RING fingers; however they form more compact heterodimers preferentially, influencing BRCA1 residues 1-109 and BARD1 residues 26-119. We implemented an integrative computational pipeline to screen all the mutations in BRCA1 and identify the most significant mutations influencing the Protein-Protein Interactions (PPI) in the BRCA1-BARD1 protein complex. The amino acids involved in the PPI regions were identified from the PDBsum database with the PDB ID: 1JM7. We screened 2118 missense mutations in BRCA1 and none in BARD1 for pathogenicity and stability and analyzed the amino acid sequences for conserved residues. We identified the most significant mutations from these screenings as V11G, M18K, L22S, and T97R positioned in the PPI regions of the BRCA1-BARD1 protein complex. We further performed protein-protein docking using the ZDOCK server. The native protein-protein complex showed the highest binding score of 2118.613, and the V11G mutant protein complex showed the least binding score of 1992.949. The other three mutation protein complexes had binding scores between the native and V11G protein complexes. Finally, a molecular dynamics simulation study using GROMACS was performed to comprehend changes in the BRCA1-BARD1 complex's binding pattern due to the mutation. From the analysis, we observed the highest deviation with lowest compactness and a decrease in the intramolecular h-bonds in the BRCA1-BARD1 protein complex with the V11G mutation compared to the native complex or the complexes with other mutations.
Collapse
Affiliation(s)
- D Thirumal Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India; Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, India
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Nikita Jain
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Baviri Sowmya
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Kamakshi Balsekar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - R Siva
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Balu Kamaraj
- Department of Neuroscience Technology, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Jubail, Saudi Arabia
| | - Mariem Sidenna
- Department of Biomedical Sciences, College of Health and Sciences, QU Health, Qatar University, Doha, Qatar
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health and Sciences, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
23
|
Raikwar S, Jain A, Saraf S, Bidla PD, Panda PK, Tiwari A, Verma A, Jain SK. Opportunities in combinational chemo-immunotherapy for breast cancer using nanotechnology: an emerging landscape. Expert Opin Drug Deliv 2022; 19:247-268. [PMID: 35184620 DOI: 10.1080/17425247.2022.2044785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 02/17/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Breast carcinoma (BC) is one of the most frequent causes of cancer-related death among women, which is due to the poor response to conventional therapy. There are several complications associated with monotherapy for cancer, such as cytotoxicity to normal cells, multidrug resistance (MDR), side effects, and limited applications. To overcome these challenges, a combination of chemotherapy and immunotherapy (monoclonal antibodies, anticancer vaccines, checkpoint inhibitors, and cytokines) has been introduced. Drug delivery systems (DDSs) based on nanotechnology have more applications in BC treatment owing to their controlled and targeted drug release with lower toxicity and reduced adverse drug effects. Several nanocarriers, such as liposomes, nanoparticles, dendrimers, and micelles, have been used for the effective delivery of drugs. AREAS COVERED This article presents opportunities and challenges in BC treatment, the rationale for cancer immunotherapy, and several combinational approaches with their applications for BC treatment. EXPERT OPINION Nanotechnology can be used for the early prognosis and cure of BC. Several novel and targeted DDSs have been developed to enhance the efficacy of anticancer drugs. This article aims to understand new strategies for the treatment of BC and the appropriate design of nanocarriers used as a combinational DDS.
Collapse
Affiliation(s)
- Sarjana Raikwar
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Ankit Jain
- Department of Materials Engineering, Indian Institute of Science, Bangalore, India
| | - Shivani Saraf
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Pooja Das Bidla
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Pritish Kumar Panda
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Ankita Tiwari
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Amit Verma
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| |
Collapse
|
24
|
Vishwanath D, Girimanchanaika SS, Dukanya D, Rangappa S, Yang JR, Pandey V, Lobie PE, Basappa B. Design and Activity of Novel Oxadiazole Based Compounds That Target Poly(ADP-ribose) Polymerase. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030703. [PMID: 35163965 PMCID: PMC8839658 DOI: 10.3390/molecules27030703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Novel PARP inhibitors with selective mode-of-action have been approved for clinical use. Herein, oxadiazole based ligands that are predicted to target PARP-1 have been synthesized and screened for the loss of cell viability in mammary carcinoma cells, wherein seven compounds were observed to possess significant IC50 values in the range of 1.4 to 25 µM. Furthermore, compound 5u, inhibited the viability of MCF-7 cells with an IC50 value of 1.4µM, when compared to Olaparib (IC50 = 3.2 µM). Compound 5s also decreased cell viability in MCF-7 and MDA-MB-231 cells with IC50 values of 15.3 and 19.2 µM, respectively. Treatment of MCF-7 cells with compounds 5u and 5s produced PARP cleavage, H2AX phosphorylation and CASPASE-3 activation comparable to that observed with Olaparib. Compounds 5u and 5s also decreased foci-formation and 3D Matrigel growth of MCF-7 cells equivalent to or greater than that observed with Olaparib. Finally, in silico analysis demonstrated binding of compound 5s towardsthe catalytic site of PARP-1, indicating that these novel oxadiazoles synthesized herein may serve as exemplars for the development of new therapeutics in cancer.
Collapse
Affiliation(s)
- Divakar Vishwanath
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570006, India; (D.V.); (S.S.G.); (D.D.)
| | - Swamy S. Girimanchanaika
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570006, India; (D.V.); (S.S.G.); (D.D.)
| | - Dukanya Dukanya
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570006, India; (D.V.); (S.S.G.); (D.D.)
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Mandya 571448, India;
| | - Ji-Rui Yang
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.-R.Y.); (V.P.)
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.-R.Y.); (V.P.)
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Peter E. Lobie
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; (J.-R.Y.); (V.P.)
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
- Correspondence: (P.E.L.); (B.B.)
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570006, India; (D.V.); (S.S.G.); (D.D.)
- Correspondence: (P.E.L.); (B.B.)
| |
Collapse
|
25
|
Mavrommati I, Johnson F, Echeverria GV, Natrajan R. Subclonal heterogeneity and evolution in breast cancer. NPJ Breast Cancer 2021; 7:155. [PMID: 34934048 PMCID: PMC8692469 DOI: 10.1038/s41523-021-00363-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022] Open
Abstract
Subclonal heterogeneity and evolution are characteristics of breast cancer that play a fundamental role in tumour development, progression and resistance to current therapies. In this review, we focus on the recent advances in understanding the epigenetic and transcriptomic changes that occur within breast cancer and their importance in terms of cancer development, progression and therapy resistance with a particular focus on alterations at the single-cell level. Furthermore, we highlight the utility of using single-cell tracing and molecular barcoding methodologies in preclinical models to assess disease evolution and response to therapy. We discuss how the integration of single-cell profiling from patient samples can be used in conjunction with results from preclinical models to untangle the complexities of this disease and identify biomarkers of disease progression, including measures of intra-tumour heterogeneity themselves, and how enhancing this understanding has the potential to uncover new targetable vulnerabilities in breast cancer.
Collapse
Affiliation(s)
- Ioanna Mavrommati
- grid.18886.3fThe Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Flora Johnson
- grid.18886.3fThe Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Gloria V. Echeverria
- grid.39382.330000 0001 2160 926XLester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX USA ,grid.39382.330000 0001 2160 926XDepartment of Medicine, Baylor College of Medicine, Houston, TX USA ,grid.39382.330000 0001 2160 926XDan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX USA ,grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
| |
Collapse
|
26
|
PARP Inhibitors and Myeloid Neoplasms: A Double-Edged Sword. Cancers (Basel) 2021; 13:cancers13246385. [PMID: 34945003 PMCID: PMC8699275 DOI: 10.3390/cancers13246385] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Poly(ADP-ribose) polymerase (PARP) inhibitors, which are medications approved to treat various solid tumors, including breast, prostate, ovarian, and prostate cancers, are being examined in hematological malignancies. This review summarizes the potential role of PARP inhibitors in the treatment of myeloid diseases, particularly acute myeloid leukemia (AML). We review ongoing clinical studies investigating the safety and efficacy of PARP inhibitors in the treatment of AML, focusing on specific molecular and genetic AML subgroups that could be particularly sensitive to PARP inhibitor treatment. We also discuss reports describing an increased risk of treatment-related myeloid neoplasms in patients receiving PARP inhibitors for solid tumors. Abstract Despite recent discoveries and therapeutic advances in aggressive myeloid neoplasms, there remains a pressing need for improved therapies. For instance, in acute myeloid leukemia (AML), while most patients achieve a complete remission with conventional chemotherapy or the combination of a hypomethylating agent and venetoclax, de novo or acquired drug resistance often presents an insurmountable challenge, especially in older patients. Poly(ADP-ribose) polymerase (PARP) enzymes, PARP1 and PARP2, are involved in detecting DNA damage and repairing it through multiple pathways, including base excision repair, single-strand break repair, and double-strand break repair. In the context of AML, PARP inhibitors (PARPi) could potentially exploit the frequently dysfunctional DNA repair pathways that, similar to deficiencies in homologous recombination in BRCA-mutant disease, set the stage for cell killing. PARPi appear to be especially effective in AML with certain gene rearrangements and molecular characteristics (RUNX1-RUNX1T1 and PML-RARA fusions, FLT3- and IDH1-mutated). In addition, PARPi can enhance the efficacy of other agents, particularly alkylating agents, TOP1 poisons, and hypomethylating agents, that induce lesions ordinarily repaired via PARP1-dependent mechanisms. Conversely, emerging reports suggest that long-term treatment with PARPi for solid tumors is associated with an increased incidence of myelodysplastic syndrome (MDS) and AML. Here, we (i) review the pre-clinical and clinical data on the role of PARPi, specifically olaparib, talazoparib, and veliparib, in aggressive myeloid neoplasms and (ii) discuss the reported risk of MDS/AML with PARPi, especially as the indications for PARPi use expand to include patients with potentially curable cancer.
Collapse
|
27
|
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
|
28
|
Evolutionary dynamics of cancer multidrug resistance in response to olaparib and photodynamic therapy. Transl Oncol 2021; 14:101198. [PMID: 34418731 PMCID: PMC8387718 DOI: 10.1016/j.tranon.2021.101198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/15/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
P-glycoprotein (P-gp) is an adenosine triphosphate (ATP)-dependent drug efflux protein commonly associated with multidrug resistance in cancer chemotherapy. In this report, we used a dual-fluorescent co-culture model to study the population dynamics of the drug sensitive human ovarian cancer cell line (OVCAR-8-DsRed2) and its resistant subline that overexpresses P-gp (NCI/ADR-RES-EGFP) during the course of a photodynamic therapy (PDT)-olaparib combination regimen. Without treatment, OVCAR-8-DsRed2 cells grew more rapidly than the NCI/ADR-RES-EGFP cells. Olaparib treatment reduced the total number of cancer cells by 70±4% but selected for the resistant NCI/ADR-RES-EGFP population since olaparib is an efflux substrate for the P-gp pump. This study used the FDA-approved benzoporphyrin derivative (BPD) photosensitizer or its lipidated formulation ((16:0)LysoPC-BPD) to kill OVCAR-8 cells and reduce the likelihood that olaparib-resistant cells would have selective advantage. Three cycles of PDT effectively reduced the total cell number by 66±3%, while stabilizing the population ratio of sensitive and resistant cells at approximately 1:1. The combination of olaparib treatment and PDT enhanced PARP cleavage and deoxyribonucleic acid (DNA) damage, further decreasing the total cancer cell number down to 10±2%. We also showed that the combination of olaparib and (16:0)LysoPC-BPD-based PDT is up to 18-fold more effective in mitigating the selection of resistant NCI/ADR-RES-EGFP cells, compared to using olaparib and BPD-based PDT. These studies suggest that PDT may improve the effectiveness of olaparib, and the use of a lipidated photosensitizer formulation holds promise in overcoming cancer drug resistance.
Collapse
|
29
|
Xiong G, Wu Z, Yi J, Fu L, Yang Z, Hsieh C, Yin M, Zeng X, Wu C, Lu A, Chen X, Hou T, Cao D. ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res 2021; 49:W5-W14. [PMID: 33893803 PMCID: PMC8262709 DOI: 10.1093/nar/gkab255] [Citation(s) in RCA: 1196] [Impact Index Per Article: 299.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/20/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Because undesirable pharmacokinetics and toxicity of candidate compounds are the main reasons for the failure of drug development, it has been widely recognized that absorption, distribution, metabolism, excretion and toxicity (ADMET) should be evaluated as early as possible. In silico ADMET evaluation models have been developed as an additional tool to assist medicinal chemists in the design and optimization of leads. Here, we announced the release of ADMETlab 2.0, a completely redesigned version of the widely used AMDETlab web server for the predictions of pharmacokinetics and toxicity properties of chemicals, of which the supported ADMET-related endpoints are approximately twice the number of the endpoints in the previous version, including 17 physicochemical properties, 13 medicinal chemistry properties, 23 ADME properties, 27 toxicity endpoints and 8 toxicophore rules (751 substructures). A multi-task graph attention framework was employed to develop the robust and accurate models in ADMETlab 2.0. The batch computation module was provided in response to numerous requests from users, and the representation of the results was further optimized. The ADMETlab 2.0 server is freely available, without registration, at https://admetmesh.scbdd.com/.
Collapse
Affiliation(s)
- Guoli Xiong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Zhenxing Wu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jiacai Yi
- College of Computer, National University of Defense Technology, Changsha 410073, Hunan, China
| | - Li Fu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Zhijiang Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Changyu Hsieh
- Tencent Quantum Laboratory, Tencent, Shenzhen 518057, Guangdong, China
| | - Mingzhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiangxiang Zeng
- Deparment of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Chengkun Wu
- College of Computer, National University of Defense Technology, Changsha 410073, Hunan, China
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Tingjun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
30
|
Abstract
OBJECTIVE Activation of the constitutive nuclear and mitochondrial enzyme poly (ADP-ribose) polymerase (PARP) has been implicated in the pathogenesis of cell dysfunction, inflammation, and organ failure in various forms of critical illness. The objective of our study was to evaluate the efficacy and safety of the clinically approved PARP inhibitor olaparib in an experimental model of pancreatitis in vivo and in a pancreatic cell line subjected to oxidative stress in vitro. The preclinical studies were complemented with analysis of clinical samples to detect PARP activation in pancreatitis. METHODS Mice were subjected to cerulein-induced pancreatitis; circulating mediators and circulating organ injury markers; pancreatic myeloperoxidase and malondialdehyde levels were measured and histology of the pancreas was assessed. In human pancreatic duct epithelial cells (HPDE) subjected to oxidative stress, PARP activation was measured by PAR Western blotting and cell viability and DNA integrity were quantified. In clinical samples, PARP activation was assessed by PAR (the enzymatic product of PARP) immunohistochemistry. RESULTS In male mice subjected to pancreatitis, olaparib (3 mg/kg i.p.) improved pancreatic function: it reduced pancreatic myeloperoxidase and malondialdehyde levels, attenuated the plasma amylase levels, and improved the histological picture of the pancreas. It also attenuated the plasma levels of pro-inflammatory mediators (TNF-α, IL-1β, IL-2, IL-4, IL-6, IL-12, IP-10, KC) but not MCP-1, RANTES, or the anti-inflammatory cytokine IL-10. Finally, it prevented the slight, but significant increase in plasma blood urea nitrogen level, suggesting improved renal function. The protective effect of olaparib was also confirmed in female mice. In HPDE cells subjected to oxidative stress olaparib (1 μM) inhibited PARP activity, protected against the loss of cell viability, and prevented the loss of cellular NAD levels. Olaparib, at 1μM to 30 μM did not have any adverse effects on DNA integrity. In human pancreatic samples from patients who died of pancreatitis, increased accumulation of PAR was demonstrated. CONCLUSION Olaparib improves organ function and tempers the hyperinflammatory response in pancreatitis. It also protects against pancreatic cell injury in vitro without adversely affecting DNA integrity. Repurposing and eventual clinical introduction of this clinically approved PARP inhibitor may be warranted for the experimental therapy of pancreatitis.
Collapse
|
31
|
PARP inhibitor olaparib has a potential to increase the effectiveness of electrochemotherapy in BRCA1 mutated breast cancer in mice. Bioelectrochemistry 2021; 140:107832. [PMID: 33984694 DOI: 10.1016/j.bioelechem.2021.107832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
Electrochemotherapy (ECT), a local therapy, has different effectiveness among tumor types. In breast cancer, its effectiveness is low; therefore, combined therapies are needed. The aim of our study was to combine ECT with PARP inhibitor olaparib, which could inhibit the repair of bleomycin or cisplatin induced DNA damage and potentiate the effectiveness of ECT. The effects of combined therapy were studied in BRCA1 mutated (HCC1937) and non-mutated (HCC1143) triple negative breast cancer cell lines. Therapeutic effectiveness was studied in 2D and 3D cell cultures and in vivo on subcutaneous HCC1937 tumor model in mice. The underlying mechanism of combined therapy was determined with the evaluation of γH2AX foci. Combined therapy of ECT with bleomycin and olaparib potentiated the effectiveness of ECT in BRCA1 mutated HCC1937, but not in non-mutated HCC1143 cells. The combined therapy had a synergistic effect, which was due to the increased number of DNA double strand breaks. Addition of olaparib to ECT with bleomycin in vivo in HCC1937 tumor model had only minimal effect, indicating repetitive olaparib treatment would be needed. This study demonstrates that DNA repair inhibiting drugs, like olaparib, have the potential to increase the effectiveness of ECT with bleomycin.
Collapse
|
32
|
Lo CSY, van Toorn M, Gaggioli V, Paes Dias M, Zhu Y, Manolika EM, Zhao W, van der Does M, Mukherjee C, G S C Souto Gonçalves J, van Royen ME, French PJ, Demmers J, Smal I, Lans H, Wheeler D, Jonkers J, Chaudhuri AR, Marteijn JA, Taneja N. SMARCAD1-mediated active replication fork stability maintains genome integrity. SCIENCE ADVANCES 2021; 7:7/19/eabe7804. [PMID: 33952518 PMCID: PMC8099181 DOI: 10.1126/sciadv.abe7804] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/16/2021] [Indexed: 05/17/2023]
Abstract
The stalled fork protection pathway mediated by breast cancer 1/2 (BRCA1/2) proteins is critical for replication fork stability. However, it is unclear whether additional mechanisms are required to maintain replication fork stability. We describe a hitherto unknown mechanism, by which the SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily-A containing DEAD/H box-1 (SMARCAD1) stabilizes active replication forks, that is essential to maintaining resistance towards replication poisons. We find that SMARCAD1 prevents accumulation of 53BP1-associated nucleosomes to preclude toxic enrichment of 53BP1 at the forks. In the absence of SMARCAD1, 53BP1 mediates untimely dissociation of PCNA via the PCNA-unloader ATAD5, causing frequent fork stalling, inefficient fork restart, and accumulation of single-stranded DNA. Although loss of 53BP1 in SMARCAD1 mutants rescues these defects and restores genome stability, this rescued stabilization also requires BRCA1-mediated fork protection. Notably, fork protection-challenged BRCA1-deficient naïve- or chemoresistant tumors require SMARCAD1-mediated active fork stabilization to maintain unperturbed fork progression and cellular proliferation.
Collapse
Affiliation(s)
- Calvin Shun Yu Lo
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Marvin van Toorn
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
- Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Vincent Gaggioli
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Mariana Paes Dias
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Yifan Zhu
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Eleni Maria Manolika
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Wei Zhao
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Marit van der Does
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Chirantani Mukherjee
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - João G S C Souto Gonçalves
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Martin E van Royen
- Department of Pathology, Cancer Treatment Screening Facility (CTSF), Erasmus Optical Imaging Centre (OIC), Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, Netherlands
| | - Pim J French
- Department of Neurology and Cancer Treatment Screening Facility (CTSF), Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Jeroen Demmers
- Proteomics Center and Department of Biochemistry, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, Netherlands
| | - Ihor Smal
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Hannes Lans
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - David Wheeler
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Arnab Ray Chaudhuri
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Jurgen A Marteijn
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
- Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Nitika Taneja
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands.
| |
Collapse
|
33
|
Yamaguchi H, Kitami K, Wu X, He L, Wang J, Wang B, Komatsu Y. Alteration of DNA Damage Response Causes Cleft Palate. Front Physiol 2021; 12:649492. [PMID: 33854442 PMCID: PMC8039291 DOI: 10.3389/fphys.2021.649492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Cleft palate is one of the most common craniofacial birth defects, however, little is known about how changes in the DNA damage response (DDR) cause cleft palate. To determine the role of DDR during palatogenesis, the DDR process was altered using a pharmacological intervention approach. A compromised DDR caused by a poly (ADP-ribose) polymerase (PARP) enzyme inhibitor resulted in cleft palate in wild-type mouse embryos, with increased DNA damage and apoptosis. In addition, a mouse genetic approach was employed to disrupt breast cancer 1 (BRCA1) and breast cancer 2 (BRCA2), known as key players in DDR. An ectomesenchymal-specific deletion of Brca1 or Brca2 resulted in cleft palate due to attenuation of cell survival. This was supported by the phenotypes of the ectomesenchymal-specific Brca1/Brca2 double-knockout mice. The cleft palate phenotype was rescued by superimposing p53 null alleles, demonstrating that the BRCA1/2-p53 DDR pathway is critical for palatogenesis. Our study highlights the importance of DDR in palatogenesis.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Kohei Kitami
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Xiao Wu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Li He
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Jianbo Wang
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States
| | - Bin Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Yoshihiro Komatsu
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, United States.,Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
34
|
Pan Y, Han H, Labbe KE, Zhang H, Wong KK. Recent advances in preclinical models for lung squamous cell carcinoma. Oncogene 2021; 40:2817-2829. [PMID: 33707749 DOI: 10.1038/s41388-021-01723-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 12/25/2022]
Abstract
Lung squamous cell carcinoma (LUSC) represents a major subtype of non-small cell lung cancer with limited treatment options. Previous studies have elucidated the complex genetic landscape of LUSC and revealed multiple altered genes and pathways. However, in stark contrast to lung adenocarcinoma, few targetable driver mutations have been established so far and targeted therapies for LUSC remain unsuccessful. Immunotherapy has revolutionized LUSC treatment and is currently approved as the new standard of care. To gain a better understanding of the LUSC biology, improved modeling systems are urgently needed. Preclinical models, particularly those mimicking human disease with an intact tumor immune microenvironment, are an invaluable tool to study cancer development and evaluate new therapeutic targets. Here, we discuss recent advances in LUSC preclinical models, with a focus on genetically engineered mouse models (GEMMs) and organoids, in the context of evolving precision medicine and immunotherapy.
Collapse
Affiliation(s)
- Yuanwang Pan
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Han Han
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Kristen E Labbe
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Hua Zhang
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA.
| | - Kwok-Kin Wong
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA.
| |
Collapse
|
35
|
McCart Reed AE, Kalaw EM, Lakhani SR. An Update on the Molecular Pathology of Metaplastic Breast Cancer. BREAST CANCER-TARGETS AND THERAPY 2021; 13:161-170. [PMID: 33664587 PMCID: PMC7924111 DOI: 10.2147/bctt.s296784] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
Metaplastic breast cancer (MpBC) is a fascinating morphologic sub-type of breast cancer, characterised by intra-tumoural heterogeneity. By definition, these tumors show regions of metaplasia that can present as spindle, squamous, chondroid or even osseous differentiation. MpBC are typically triple-negative, and are therefore not targetable with hormone therapy or anti-HER2 therapies, leaving only chemotherapeutics for management. MpBC are known for their aggressive course and poor response to chemotherapy. We review herein the pathology and molecular landscape of MpBC and discuss opportunities for targetted therapies as well as immunotherapies.
Collapse
Affiliation(s)
- Amy E McCart Reed
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Emarene M Kalaw
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia.,Pathology Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
36
|
Li LY, Guan YD, Chen XS, Yang JM, Cheng Y. DNA Repair Pathways in Cancer Therapy and Resistance. Front Pharmacol 2021; 11:629266. [PMID: 33628188 PMCID: PMC7898236 DOI: 10.3389/fphar.2020.629266] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/31/2020] [Indexed: 12/21/2022] Open
Abstract
DNA repair pathways are triggered to maintain genetic stability and integrity when mammalian cells are exposed to endogenous or exogenous DNA-damaging agents. The deregulation of DNA repair pathways is associated with the initiation and progression of cancer. As the primary anti-cancer therapies, ionizing radiation and chemotherapeutic agents induce cell death by directly or indirectly causing DNA damage, dysregulation of the DNA damage response may contribute to hypersensitivity or resistance of cancer cells to genotoxic agents and targeting DNA repair pathway can increase the tumor sensitivity to cancer therapies. Therefore, targeting DNA repair pathways may be a potential therapeutic approach for cancer treatment. A better understanding of the biology and the regulatory mechanisms of DNA repair pathways has the potential to facilitate the development of inhibitors of nuclear and mitochondria DNA repair pathways for enhancing anticancer effect of DNA damage-based therapy.
Collapse
Affiliation(s)
- Lan-Ya Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yi-di Guan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi-Sha Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-Ming Yang
- Department of Cancer Biology and Toxicology, Department of Pharmacology, College of Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Gadaleta-Caldarola G, Nenna R, Lanotte L, Doronzo A, Gadaleta-Caldarola A, Roma ID, Lombardi L, Infusino S. Metaplastic breast cancer: an old histotype but a current therapeutic problem. Future Oncol 2021; 17:955-963. [PMID: 33538176 DOI: 10.2217/fon-2020-0490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Metaplastic breast cancer (MPBC) is a rare and aggressive tumor type in great need of satisfactory therapies. Although most cases of MPBC are 'triple negative', they are nonetheless related to worse outcomes compared with other triple-negative invasive tumors. MPBC presents high levels of genetic and molecular heterogeneity, suggesting that novel targeted therapies can be exploited. Overexpression of PD-L1 and high levels of tumor-infiltrating lymphocytes have also been observed in these tumors, suggesting a role for immunotherapy. We present an updated literature revision on clinical, histopathological and molecular features of MPBC and their significance to prognosis and therapy options. We discuss emerging efforts to improve and personalize prognostic and therapeutic approaches, exploiting the molecular signature of MPBC with targeted therapies and immunotherapies.
Collapse
Affiliation(s)
| | - Rosanna Nenna
- Anatomic Patology Unit, "L. Bonomo" Hospital, Andria (BT), ASL BT, 76123, Italy
| | - Laura Lanotte
- Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), ASL BT, 76121, Italy
| | - Antonio Doronzo
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria "Ospedali Riuniti", Foggia, 71100, Italy
| | | | - Ileana de Roma
- Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), ASL BT, 76121, Italy
| | - Lucia Lombardi
- Medical Oncology Unit, "Mons. R. Dimiccoli" Hospital, Barletta (BT), ASL BT, 76121, Italy
| | - Stefania Infusino
- Medical Oncology Unit, "SS. Annunziata" Hospital, Cosenza (CS), Azienda Ospedaliera di Cosenza, 87100, Italy
| |
Collapse
|
38
|
Abstract
Owing to increased awareness of the importance of mammogram and advances in surgical technology, survival rate of patients with primary breast cancer has dramatically increased. Despite all these advances in breast cancer treatment, there are no currently available treatments for this disease once it metastasizes to distant organs including bones, lungs, brain, and liver. This is mainly attributed to the complexity of metastatic process. Recent advances in technology enabled cancer biologists to dissect each step of the metastatic process, and this led to discovery of major players and molecules in this process. In this section, we will discuss recent discovery and advances in the field of breast cancer metastasis research.
Collapse
|
39
|
Chang TC, Matossian MD, Elliott S, Burks HE, Sabol RA, Ucar DA, Wathieu H, Zabaleta J, Valle LD, Gill S, Martin E, Riker AI, Miele L, Bunnell BA, Burow ME, Collins-Burow BM. Evaluation of deacetylase inhibition in metaplastic breast carcinoma using multiple derivations of preclinical models of a new patient-derived tumor. PLoS One 2020; 15:e0226464. [PMID: 33035223 PMCID: PMC7546483 DOI: 10.1371/journal.pone.0226464] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Metaplastic breast carcinoma (MBC) is a clinically aggressive and rare subtype of breast cancer, with similar features to basal-like breast cancers. Due to rapid growth rates and characteristic heterogeneity, MBC is often unresponsive to standard chemotherapies; and novel targeted therapeutic discovery is urgently needed. Histone deacetylase inhibitors (DACi) suppress tumor growth and metastasis through regulation of the epithelial-to-mesenchymal transition axis in various cancers, including basal-like breast cancers. We utilized a new MBC patient-derived xenograft (PDX) to examine the effect of DACi therapy on MBC. Cell morphology, cell cycle-associated gene expressions, transwell migration, and metastasis were evaluated in patient-derived cells and tumors after treatment with romidepsin and panobinostat. Derivations of our PDX model, including cells, spheres, organoids, explants, and in vivo implanted tumors were treated. Finally, we tested the effects of combining DACi with approved chemotherapeutics on relative cell biomass. DACi significantly suppressed the total number of lung metastasis in vivo using our PDX model, suggesting a role for DACi in preventing circulating tumor cells from seeding distal tissue sites. These data were supported by our findings that DACi reduced cell migration, populations, and expression of mesenchymal-associated genes. While DACi treatment did affect cell cycle-regulating genes in vitro, tumor growth was not affected compared to controls. Importantly, gene expression results varied depending on the cellular or tumor system used, emphasizing the importance of using multiple derivations of cancer models in preclinical therapeutic discovery research. Furthermore, DACi sensitized and produced a synergistic effect with approved oncology therapeutics on inherently resistant MBC. This study introduced a role for DACi in suppressing the migratory and mesenchymal phenotype of MBC cells through regulation of the epithelial-mesenchymal transition axis and suppression of the CTC population. Preliminary evidence that DACi treatment in combination with MEK1/2 inhibitors exerts a synergistic effect on MBC cells was also demonstrated.
Collapse
Affiliation(s)
- Tiffany C. Chang
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail: (TCC); (BMCB)
| | - Margarite D. Matossian
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Steven Elliott
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Hope E. Burks
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Rachel A. Sabol
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Deniz A. Ucar
- Department of Genetics, Louisiana State University School of Medicine, New Orleans, Louisiana, United States of America
| | - Henri Wathieu
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Jovanny Zabaleta
- Department of Pediatrics, Louisiana State University School of Medicine, New Orleans, Louisiana, United States of America
| | - Luis De Valle
- Department of Pathology, Louisiana State University School of Medicine, New Orleans, Louisiana, United States of America
| | - Sukhmani Gill
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Elizabeth Martin
- Department of Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Adam I. Riker
- Department of Surgery, Louisiana State University School of Medicine, New Orleans, Louisiana, United States of America
| | - Lucio Miele
- Department of Genetics, Louisiana State University School of Medicine, New Orleans, Louisiana, United States of America
| | - Bruce A. Bunnell
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Matthew E. Burow
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Bridgette M. Collins-Burow
- Department of Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail: (TCC); (BMCB)
| |
Collapse
|
40
|
Wang Y, Zuo M, Jin H, Lai M, Luo J, Cheng Z. Inhibition of ELF3 confers synthetic lethality of PARP inhibitor in non-small cell lung cancer. J Recept Signal Transduct Res 2020; 41:304-311. [PMID: 32814472 DOI: 10.1080/10799893.2020.1808676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND E74 Like ETS Transcription Factor 3 (ELF3) functions as a transcriptional factor to regulate non-small cell lung cancer (NSCLC) differentiation and progression. Poly(ADP-ribose) polymerase (PARP) inhibitors demonstrate anti-tumor effect in NSCLC. This study aimed to investigate whether ELF3 confers synthetic lethal with PARP inhibitor in NSCLC. MATERIALS AND METHODS The sensitivity of PARP inhibitor, Olaparib, to different NSCLC cell lines was determined by half maximal inhibitory concentration (IC50). Expression of ELF3 in NSCLC cell lines was evaluated by western blot. The effects of ELF3 on cytotoxicity of Olaparib to NSCLC were investigated by MTT (3-(4,5- di methyl thiazol -2-yl)-2,5-di phenyl tetrazolium bromide) and colony formation assays. The underlying mechanism involved in synthetic lethality with ELF3 and PARP inhibitors in NSCLC were detected by immunofluorescence and Western blot. RESULTS ELF3 was up-regulated in NSCLC cell lines exhibiting resistance to PARP inhibitor, Olaparib. Knock down of ELF3 decreased the sensitivity and enhanced cytotoxicity of Olaparib to NSCLC cells. Moreover, knock down of ELF3 increased S139 phosphorylated histone H2AX (γH2AX), and inhibited homologous recombination activity via down-regulation of DNA repair protein RAD51 homolog 1 (RAD51), thus showing deficiency in DNA damage repair. Over-expression of ELF3 could up-regulate phosphorylation of AKT (Protein kinase B), while knock down of ELF3 regulated homologous recombination-mediated DNA repair via down-regulation of phosphorylation of AKT. CONCLUSION Knock down of ELF3 revealed homologous recombination deficiency via AKT signaling pathway, and synthetic lethality with ELF3 inhibition and PARP inhibitor indicated the clinical significance of PARP inhibitor in ELF3-deficient NSCLC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Min Zuo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Hongtao Jin
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Meina Lai
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jinfeng Luo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Zhiqiang Cheng
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
41
|
Xiong Y, Guo Y, Liu Y, Wang H, Gong W, Liu Y, Wang X, Gao Y, Yu F, Su D, Wang F, Zhu Y, Zhao Y, Wu Y, Qin Z, Sun X, Ren B, Jiang B, Jin W, Shen Z, Tang Z, Song X, Wang L, Liu X, Zhou C, Jiang B. Pamiparib is a potent and selective PARP inhibitor with unique potential for the treatment of brain tumor. Neoplasia 2020; 22:431-440. [PMID: 32652442 PMCID: PMC7350150 DOI: 10.1016/j.neo.2020.06.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/01/2023] Open
Abstract
Pamiparib, an investigational Poly (ADP-ribose) polymerase (PARP) inhibitor in clinical development, demonstrates excellent selectivity for both PARP1 and PARP2, and superb anti-proliferation activities in tumor cell lines with BRCA1/2 mutations or HR pathway deficiency (HRD). Pamiparib has good bioavailability and is 16-fold more potent than olaparib in an efficacy study using BRCA1 mutated MDA-MB-436 breast cancer xenograft model. Pamiparib also shows strong anti-tumor synergy with temozolomide (TMZ), a DNA alkylating agent used to treat brain tumors. Compared to other PARP inhibitors, pamiparib demonstrated improved penetration across the blood brain barrier (BBB) in mice. Oral administration of pamiparib at a dose as low as 3 mg/kg is sufficient to abrogate PARylation in brain tumor tissues. In SCLC-derived, TMZ-resistant H209 intracranial xenograft model, combination of pamiparib with TMZ overcomes its resistance and shows significant tumor inhibitory effects and prolonged life span. Our data suggests that combination of pamiparib with TMZ has unique potential for treatment of brain tumors. Currently, the combination therapy of pamiparib with TMZ is evaluated in clinical trial [NCT03150862].
Collapse
Affiliation(s)
- Yao Xiong
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Yin Guo
- Department of Discovery Biology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Ye Liu
- Department of Biochemistry, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Hexiang Wang
- Department of Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Wenfeng Gong
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Yong Liu
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Xing Wang
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Yajuan Gao
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Fenglong Yu
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Dan Su
- Department of DMPK, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Fan Wang
- Department of DMPK, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Yutong Zhu
- Department of Discovery Biology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Yuan Zhao
- Department of Discovery Biology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Yiyuan Wu
- Department of Discovery Biology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Zhen Qin
- Department of Discovery Biology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Xuebing Sun
- Department of Biochemistry, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Bo Ren
- Department of Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Bin Jiang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Wei Jin
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Zhirong Shen
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Zhiyu Tang
- Department of Clinic Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Xiaomin Song
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Lai Wang
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Xuesong Liu
- Department of Discovery Biology, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Changyou Zhou
- Department of Chemistry, BeiGene (Beijing) Co., Ltd., Beijing, PR China
| | - Beibei Jiang
- Department of In Vivo Pharmacology, BeiGene (Beijing) Co., Ltd., Beijing, PR China.
| |
Collapse
|
42
|
Zebrafish Xenografts Unveil Sensitivity to Olaparib beyond BRCA Status. Cancers (Basel) 2020; 12:cancers12071769. [PMID: 32630796 PMCID: PMC7408583 DOI: 10.3390/cancers12071769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/11/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibition in BRCA-mutated cells results in an incapacity to repair DNA damage, leading to cell death caused by synthetic lethality. Within the treatment options for advanced triple negative breast cancer, the PARP inhibitor olaparib is only given to patients with BRCA1/2 mutations. However, these patients may show resistance to this drug and BRCA1/2 wild-type tumors can show a striking sensitivity, making BRCA status a poor biomarker for treatment choice. Aiming to investigate if the zebrafish model can discriminate sensitivities to olaparib, we developed zebrafish xenografts with different BRCA status and measured tumor response to treatment, as well as its impact on angiogenesis and metastasis. When challenged with olaparib, xenografts revealed sensitivity phenotypes independent of BRCA. Moreover, its combination with ionizing radiation increased the cytotoxic effects, showing potential as a combinatorial regimen. In conclusion, we show that the zebrafish xenograft model may be used as a sensitivity profiling platform for olaparib in monotherapy or in combinatorial regimens. Hence, this model presents as a promising option for the future establishment of patient-derived xenografts for personalized medicine approaches beyond BRCA status.
Collapse
|
43
|
Cleary JM, Aguirre AJ, Shapiro GI, D'Andrea AD. Biomarker-Guided Development of DNA Repair Inhibitors. Mol Cell 2020; 78:1070-1085. [PMID: 32459988 PMCID: PMC7316088 DOI: 10.1016/j.molcel.2020.04.035] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/02/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Anti-cancer drugs targeting the DNA damage response (DDR) exploit genetic or functional defects in this pathway through synthetic lethal mechanisms. For example, defects in homologous recombination (HR) repair arise in cancer cells through inherited or acquired mutations in BRCA1, BRCA2, or other genes in the Fanconi anemia/BRCA pathway, and these tumors have been shown to be particularly sensitive to inhibitors of the base excision repair (BER) protein poly (ADP-ribose) polymerase (PARP). Recent work has identified additional genomic and functional assays of DNA repair that provide new predictive and pharmacodynamic biomarkers for these targeted therapies. Here, we examine the development of selective agents targeting DNA repair, including PARP inhibitors; inhibitors of the DNA damage kinases ataxia-telangiectasia and Rad3 related (ATR), CHK1, WEE1, and ataxia-telangiectasia mutated (ATM); and inhibitors of classical non-homologous end joining (cNHEJ) and alternative end joining (Alt EJ). We also review the biomarkers that guide the use of these agents and current clinical trials with these therapies.
Collapse
Affiliation(s)
- James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alan D D'Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Liang Y, Yu L, Zhang D, Zhao X, Gao H, Slagle BL, Goss JA, Wang X, Li K, Lin SY. BRIT1 dysfunction confers synergistic inhibition of hepatocellular carcinoma by targeting poly (ADP-ribose) polymerases and PI3K. Am J Cancer Res 2020; 10:1900-1918. [PMID: 32642299 PMCID: PMC7339272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 05/01/2020] [Indexed: 06/11/2023] Open
Abstract
BRIT1 has emerged as a novel key player in homologous recombination (HR). It is located in 8p23, a locus frequently deleted in hepatocellular carcinoma (HCC). Previously, we found that BRIT1-deficiency triggered genomic instability and tumor formation in our mouse model. Here we aim to determine whether BRIT1 aberrations are implicated in HCC and, if so, whether they can be used for targeted therapy with PARP inhibitors and other agents. We analyzed HCC samples for BRIT1 alterations at DNA, RNA and protein levels. BRIT1 was found deleted and/or downregulated in ~30% of HCC samples; BRIT1 mutant K659fsX10 identified in HCC abolished DNA repair function. Notably, BRIT1 deletion was correlated with poor survival and high recurrence of HCC. To determine the role of BRIT1 deficiency in potentiating the drug response, we subsequently generated BRIT1-deficient HCC cells, determined their HR defects, and assessed their response to the PARPi olaparib and PI3K inhibitor in vitro and in mice. BRIT1-deficient HCC cells were HR defective and hypersensitive to olaparib alone or in combination with PI3K inhibitor BEZ235, both in vitro and in vivo. The cytotoxicity of olaparib alone or in combination with BEZ235 was largely alleviated by ectopic BRIT1. We also found that BEZ235 markedly enhanced the production of poly (ADP-ribose) and the level of double-strand breaks (DSB) and single-strand breaks (SSB) in BRIT1-deficient cells. In summary, our results identify BRIT1 deficiency as a potential driver for HCC development, and BRIT1 status is critical to sensitivity to treatment with olaparib and/or BEZ235. PI3K inhibition induces substantial DNA damage and makes cells more dependent on PARP activity in the context of BRIT1 deficiency, thus, BRIT1 depletion facilitates enhancing synthetic lethality of PARP inhibitors and PI3K inhibitors in HCC. This study provides a new mechanistic foundation for significantly expanding the application of PARPi in HCC therapy.
Collapse
Affiliation(s)
- Yulong Liang
- The Michael E. DeBakey Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - Lihou Yu
- The Michael E. DeBakey Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - Dongxiao Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical UniversityBeijing, China
| | - Xuemei Zhao
- School of Pharmaceutical Science, Shandong First Medical UniversityTai’an, Shandong, China
| | - Hong Gao
- The Michael E. DeBakey Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - Betty L Slagle
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX, USA
| | - John A Goss
- The Michael E. DeBakey Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - Xiaosong Wang
- UPMC Hillman Cancer Center, Department of Pathology, University of PittsburghPittsburgh, PA, USA
| | - Kaiyi Li
- The Michael E. DeBakey Department of Surgery, Baylor College of MedicineHouston, TX, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, UT MD Anderson Cancer CenterHouston, TX, USA
| |
Collapse
|
45
|
A multifunctional magnetic nanosystem based on "two strikes" effect for synergistic anticancer therapy in triple-negative breast cancer. J Control Release 2020; 322:401-415. [PMID: 32246976 DOI: 10.1016/j.jconrel.2020.03.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Multifunctional magnetic nanoparticles (MNPs) were widely used for ablation of cancer cells because of their potential on physical treatment. Herein, we developed the "cell targeting destructive" multifunctional polymeric nanoparticles (named as HA-Olb-PPMNPs) based on PEI-PLGA co-loaded with the anticancer drug Olaparib (Olb) and superparamagnetic iron oxide nanoparticles (Fe3O4 NPs), and further coated with a low molecular weight hyaluronic acid (HA) on its surface. Due to the high affinity between HA and CD44-receptor on cell surface of triple negative breast cancer (TNBC), an active targeting can be achieved. Under a rotating magnetic field (RMF), HA-Olb-PPMNPs produced a physical transfer of mechanical force by incomplete rotation. This mechanical force could cause the "two strikes" effect on the cells, in which "First-strike" was to damage the cell membrane structure (magneto-cell-lysis), another "Second-strike" could activate the lysosome-mitochondrial pathway by injuring lysosomes to induce cell apoptosis (magneto-cell-apoptosis). Therefore, the mechanical force and Olb exert dual anti-tumor effect to achieve synergistic therapeutic in the presence of RMF. This study proposes a novel multi-therapeutic concept for TNBC, as well as provided evidences of new anti-tumor therapeutic effects induced by the magnetic nanoparticles drug system.
Collapse
|
46
|
Annunziato S, Lutz C, Henneman L, Bhin J, Wong K, Siteur B, van Gerwen B, de Korte‐Grimmerink R, Zafra MP, Schatoff EM, Drenth AP, van der Burg E, Eijkman T, Mukherjee S, Boroviak K, Wessels LFA, van de Ven M, Huijbers IJ, Adams DJ, Dow LE, Jonkers J. In situ CRISPR-Cas9 base editing for the development of genetically engineered mouse models of breast cancer. EMBO J 2020; 39:e102169. [PMID: 31930530 PMCID: PMC7049816 DOI: 10.15252/embj.2019102169] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/26/2022] Open
Abstract
Genetically engineered mouse models (GEMMs) of cancer have proven to be of great value for basic and translational research. Although CRISPR-based gene disruption offers a fast-track approach for perturbing gene function and circumvents certain limitations of standard GEMM development, it does not provide a flexible platform for recapitulating clinically relevant missense mutations in vivo. To this end, we generated knock-in mice with Cre-conditional expression of a cytidine base editor and tested their utility for precise somatic engineering of missense mutations in key cancer drivers. Upon intraductal delivery of sgRNA-encoding vectors, we could install point mutations with high efficiency in one or multiple endogenous genes in situ and assess the effect of defined allelic variants on mammary tumorigenesis. While the system also produces bystander insertions and deletions that can stochastically be selected for when targeting a tumor suppressor gene, we could effectively recapitulate oncogenic nonsense mutations. We successfully applied this system in a model of triple-negative breast cancer, providing the proof of concept for extending this flexible somatic base editing platform to other tissues and tumor types.
Collapse
Affiliation(s)
- Stefano Annunziato
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Catrin Lutz
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Linda Henneman
- Transgenic Core FacilityMouse Clinic for Cancer and Aging (MCCA)The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jinhyuk Bhin
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Molecular CarcinogenesisThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Kim Wong
- Wellcome Trust Sanger InstituteCambridgeUK
| | - Bjørn Siteur
- Preclinical Intervention UnitMouse Clinic for Cancer and Aging (MCCA)The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Bas van Gerwen
- Preclinical Intervention UnitMouse Clinic for Cancer and Aging (MCCA)The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Renske de Korte‐Grimmerink
- Preclinical Intervention UnitMouse Clinic for Cancer and Aging (MCCA)The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Maria Paz Zafra
- Division of Hematology and Medical OncologyDepartment of MedicineSandra and Edward Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Emma M Schatoff
- Division of Hematology and Medical OncologyDepartment of MedicineSandra and Edward Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
- Weill Cornell/Rockefeller/Sloan Kettering Tri‐I MD‐PhD ProgramNew YorkNYUSA
| | - Anne Paulien Drenth
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Eline van der Burg
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Timo Eijkman
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Siddhartha Mukherjee
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | | | - Lodewyk FA Wessels
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Molecular CarcinogenesisThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Marieke van de Ven
- Preclinical Intervention UnitMouse Clinic for Cancer and Aging (MCCA)The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Ivo J Huijbers
- Transgenic Core FacilityMouse Clinic for Cancer and Aging (MCCA)The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | | | - Lukas E Dow
- Division of Hematology and Medical OncologyDepartment of MedicineSandra and Edward Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
- Department of BiochemistrySandra and Edward Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Jos Jonkers
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Cancer Genomics NetherlandsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
47
|
Morosi L, Matteo C, Ceruti T, Giordano S, Ponzo M, Frapolli R, Zucchetti M, Davoli E, D'Incalci M, Ubezio P. Quantitative determination of niraparib and olaparib tumor distribution by mass spectrometry imaging. Int J Biol Sci 2020; 16:1363-1375. [PMID: 32210725 PMCID: PMC7085221 DOI: 10.7150/ijbs.41395] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/18/2020] [Indexed: 12/15/2022] Open
Abstract
Rationale: Optimal intratumor distribution of an anticancer drug is fundamental to reach an active concentration in neoplastic cells, ensuring the therapeutic effect. Determination of drug concentration in tumor homogenates by LC-MS/MS gives important information about this issue but the spatial information gets lost. Targeted mass spectrometry imaging (MSI) has great potential to visualize drug distribution in the different areas of tumor sections, with good spatial resolution and superior specificity. MSI is rapidly evolving as a quantitative technique to measure the absolute drug concentration in each single pixel. Methods: Different inorganic nanoparticles were tested as matrices to visualize the PARP inhibitors (PARPi) niraparib and olaparib. Normalization by deuterated internal standard and a custom preprocessing pipeline were applied to achieve a reliable single pixel quantification of the two drugs in human ovarian tumors from treated mice. Results: A quantitative method to visualize niraparib and olaparib in tumor tissue of treated mice was set up and validated regarding precision, accuracy, linearity, repeatability and limit of detection. The different tumor penetration of the two drugs was visualized by MSI and confirmed by LC-MS/MS, indicating the homogeneous distribution and higher tumor exposure reached by niraparib compared to olaparib. On the other hand, niraparib distribution was heterogeneous in an ovarian tumor model overexpressing the multidrug resistance protein P-gp, a possible cause of resistance to PARPi. Conclusions: The current work highlights for the first time quantitative distribution of PAPRi in tumor tissue. The different tumor distribution of niraparib and olaparib could have important clinical implications. These data confirm the validity of MSI for spatial quantitative measurement of drug distribution providing fundamental information for pharmacokinetic studies, drug discovery and the study of resistance mechanisms.
Collapse
Affiliation(s)
- Lavinia Morosi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Cristina Matteo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Tommaso Ceruti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Silvia Giordano
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Laboratory of Mass Spectrometry
| | - Marianna Ponzo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Roberta Frapolli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Massimo Zucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Enrico Davoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Laboratory of Mass Spectrometry
| | - Maurizio D'Incalci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| | - Paolo Ubezio
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology
| |
Collapse
|
48
|
Schipper K, Drenth AP, van der Burg E, Cornelissen S, Klarenbeek S, Nethe M, Jonkers J. Truncated ASPP2 Drives Initiation and Progression of Invasive Lobular Carcinoma via Distinct Mechanisms. Cancer Res 2020; 80:1486-1497. [PMID: 32060147 DOI: 10.1158/0008-5472.can-19-3607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/21/2019] [Accepted: 02/03/2020] [Indexed: 12/14/2022]
Abstract
Invasive lobular carcinoma (ILC) accounts for 8%-14% of all breast cancer cases. The main hallmark of ILCs is the functional loss of the cell-cell adhesion protein E-cadherin. Nonetheless, loss of E-cadherin alone does not predispose mice to mammary tumor development, indicating that additional perturbations are required for ILC formation. Previously, we identified an N-terminal truncation variant of ASPP2 (t-ASPP2) as a driver of ILC in mice with mammary-specific loss of E-cadherin. Here we showed that expression of t-ASPP2 induced actomyosin relaxation, enabling adhesion and survival of E-cadherin-deficient murine mammary epithelial cells on stiff matrices like fibrillar collagen. The induction of actomyosin relaxation by t-ASPP2 was dependent on its interaction with protein phosphatase 1, but not on t-ASPP2-induced YAP activation. Truncated ASPP2 collaborated with both E-cadherin loss and PI3K pathway activation via PTEN loss in ILC development. t-ASPP2-induced actomyosin relaxation was required for ILC initiation, but not progression. Conversely, YAP activation induced by t-ASPP2 contributed to tumor growth and progression while being dispensable for tumor initiation. Together, these findings highlight two distinct mechanisms through which t-ASPP2 promotes ILC initiation and progression. SIGNIFICANCE: Truncated ASPP2 cooperates with E-cadherin and PTEN loss to drive breast cancer initiation and progression via two distinct mechanisms. ASPP2-induced actomyosin relaxation drives tumor initiation, while ASPP2-mediated YAP activation enhances tumor progression.
Collapse
Affiliation(s)
- Koen Schipper
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anne Paulien Drenth
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Samuel Cornelissen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Micha Nethe
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
49
|
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
|
50
|
Abstract
In this review, Slade provides an overview of the molecular mechanisms and cellular consequences of PARP and PARG inhibition. The author also highlights the clinical performance of four PARP inhibitors used in cancer therapy (olaparib, rucaparib, niraparib, and talazoparib) and discusses the predictive biomarkers of inhibitor sensitivity and mechanisms of resistance as well as the means of overcoming them through combination therapy. Oxidative and replication stress underlie genomic instability of cancer cells. Amplifying genomic instability through radiotherapy and chemotherapy has been a powerful but nonselective means of killing cancer cells. Precision medicine has revolutionized cancer therapy by putting forth the concept of selective targeting of cancer cells. Poly(ADP-ribose) polymerase (PARP) inhibitors represent a successful example of precision medicine as the first drugs targeting DNA damage response to have entered the clinic. PARP inhibitors act through synthetic lethality with mutations in DNA repair genes and were approved for the treatment of BRCA mutated ovarian and breast cancer. PARP inhibitors destabilize replication forks through PARP DNA entrapment and induce cell death through replication stress-induced mitotic catastrophe. Inhibitors of poly(ADP-ribose) glycohydrolase (PARG) exploit and exacerbate replication deficiencies of cancer cells and may complement PARP inhibitors in targeting a broad range of cancer types with different sources of genomic instability. Here I provide an overview of the molecular mechanisms and cellular consequences of PARP and PARG inhibition. I highlight clinical performance of four PARP inhibitors used in cancer therapy (olaparib, rucaparib, niraparib, and talazoparib) and discuss the predictive biomarkers of inhibitor sensitivity, mechanisms of resistance as well as the means of overcoming them through combination therapy.
Collapse
Affiliation(s)
- Dea Slade
- Department of Biochemistry, Max Perutz Labs, Vienna Biocenter (VBC), University of Vienna, 1030 Vienna, Austria
| |
Collapse
|