1
|
Demuth P, Thibol L, Lemsch A, Potlitz F, Schulig L, Grathwol C, Manolikakes G, Schade D, Roukos V, Link A, Fahrer J. Targeting PARP-1 and DNA Damage Response Defects in Colorectal Cancer Chemotherapy with Established and Novel PARP Inhibitors. Cancers (Basel) 2024; 16:3441. [PMID: 39456536 PMCID: PMC11506018 DOI: 10.3390/cancers16203441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
The DNA repair protein PARP-1 emerged as a valuable target in the treatment of tumor entities with deficiencies of BRCA1/2, such as breast cancer. More recently, the application of PARP inhibitors (PARPi) such as olaparib has been expanded to other cancer entities including colorectal cancer (CRC). We previously demonstrated that PARP-1 is overexpressed in human CRC and promotes CRC progression in a mouse model. However, acquired resistance to PARPi and cytotoxicity-mediated adverse effects limit their clinical applicability. Here, we detailed the role of PARP-1 as a therapeutic target in CRC and studied the efficacy of novel PARPi compounds in wildtype (WT) and DNA repair-deficient CRC cell lines together with the chemotherapeutics irinotecan (IT), 5-fluorouracil (5-FU), and oxaliplatin (OXA). Based on the ComPlat molecule archive, we identified novel PARPi candidates by molecular docking experiments in silico, which were then confirmed by in vitro PARP activity measurements. Two promising candidates (X17613 and X17618) also showed potent PARP-1 inhibition in a CRC cell-based assay. In contrast to olaparib, the PARPi candidates caused no PARP-1 trapping and, consistently, were not or only weakly cytotoxic in WT CRC cells and their BRCA2- or ATR-deficient counterparts. Importantly, both PARPi candidates did not affect the viability of nonmalignant human colonic epithelial cells. While both olaparib and veliparib increased the sensitivity of WT CRC cells towards IT, no synergism was observed for X17613 and X17618. Finally, we provided evidence that all PARPi (olaparib > veliparib > X17613 > X17618) synergize with chemotherapeutic drugs (IT > OXA) in a BRCA2-dependent manner in CRC cells, whereas ATR deficiency had only a minor impact. Collectively, our study identified novel lead structures with potent PARP-1 inhibitory activity in CRC cells but low cytotoxicity due to the lack of PARP-1 trapping, which synergized with IT in homologous recombination deficiency.
Collapse
Affiliation(s)
- Philipp Demuth
- Department of Chemistry, Division of Food Chemistry and Toxicology, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany; (P.D.); (L.T.); (A.L.)
| | - Lea Thibol
- Department of Chemistry, Division of Food Chemistry and Toxicology, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany; (P.D.); (L.T.); (A.L.)
| | - Anna Lemsch
- Department of Chemistry, Division of Food Chemistry and Toxicology, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany; (P.D.); (L.T.); (A.L.)
| | - Felix Potlitz
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (F.P.); (L.S.); (A.L.)
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (F.P.); (L.S.); (A.L.)
| | - Christoph Grathwol
- Institute of Biological and Chemical Systems—Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), 76344 Eggenstein-Leopoldshafen, Germany;
| | - Georg Manolikakes
- Department of Chemistry, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany;
| | - Dennis Schade
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany;
| | | | - Andreas Link
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (F.P.); (L.S.); (A.L.)
| | - Jörg Fahrer
- Department of Chemistry, Division of Food Chemistry and Toxicology, RPTU Kaiserslautern-Landau, 67663 Kaiserslautern, Germany; (P.D.); (L.T.); (A.L.)
| |
Collapse
|
2
|
Madaan V, Kollara A, Spaner D, Brown TJ. ISGylation enhances dsRNA-induced interferon response and NFκB signaling in fallopian tube epithelial cells. J Biol Chem 2024; 300:107686. [PMID: 39159817 PMCID: PMC11418117 DOI: 10.1016/j.jbc.2024.107686] [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: 02/22/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
Heritable mutations in BRCA1 associate with increased risk of high-grade serous tubo-ovarian cancer. Nongenetic risk factors associated with this cancer, which arises from fallopian tube epithelial (FTE) cells, suggests a role for repetitive ovulation wherein FTE cells are exposed to inflammatory signaling molecules within follicular fluid. We previously reported increased NFκB and EGFR signaling in BRCA1-deficient primary FTE cells, with follicular fluid exposure further increasing abundance of interferon-stimulated gene (ISG) transcripts, including the ubiquitin-like protein ISG15 and other ISGylation pathway members. Both NFκB and type I interferon signaling are upregulated by stimulation of cGAS-STING or MDA5 and RIGI pattern recognition receptors. Since some pattern recognition receptors and their signal transduction pathway members are ISGylated, we tested the impact of ISG15 and ISGylation on interferon regulatory factor 3 (IRF3) and NFκB signaling through cGAS-STING or RIGI and MDA5 activation. Expression of ISG15 or UBA7, the E1-like ISG15-activating enzyme, in immortalized FTE cells was disrupted by CRISPR gene editing. Activation of IRF3 by RIGI or MDA5 but not cGAS-STING was attenuated by loss of either ISG15 or UBA7 and this was reflected by a similar effect on NFκB activation and downstream targets. Loss of ISGylation decreased levels of both MDA5 and RIGI, with knockdown of RIGI but not MDA5, decreasing IRF3 and NFκB activation in parental cells. These finding indicate that ISGylation enhances the ability of dsRNA to activate cytokine release and proinflammatory signaling. Further work to explore ISGylation as a target for prevention of high-grade serous tubo-ovarian cancer in BRCA1 mutation carriers is warranted.
Collapse
Affiliation(s)
- Vidushi Madaan
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Kollara
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada
| | - David Spaner
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Theodore J Brown
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Wang T, Kyle AH, Baker JHE, Liu NA, Banáth JP, Teymori S, Minchinton AI. DNA-PK inhibitor AZD7648 is a more portent radiosensitizer than PARP inhibitor Olaparib in BRCA1/2 deficient tumors. DNA Repair (Amst) 2024; 139:103689. [PMID: 38749239 DOI: 10.1016/j.dnarep.2024.103689] [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: 01/25/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024]
Abstract
The effectiveness of radiotherapy depends on the sensitivities of 'normal' and cancer cells to the administered radiation dose. Increasing the radiosensitivity of cancers by inhibiting DNA damage repair is a goal of much current research, however success depends on avoiding concomitant sensitization of normal tissues inevitably irradiated during therapy. In this study we investigated the mechanisms of radiosensitization for DNA-PK and PARP inhibitors by examining the impacts on proliferating vs quiescent cell populations. Experiments were performed in BRCA1/2null and wild-type parental cancer models in vitro and in vivo. Overall AZD7648 has greater radiosensitizing activity relative to Olaparib, with BRCA2-deficient models showing the greatest sensitivity. However, DNA-PK inhibitor AZD7648 also produced greater toxicity in all irradiated mice. While both DNA-PK and PARP inhibition sensitizes wild type tumor cells to radiation, in BRCA1/2 deficient cells PARP inhibition by Olaparib had limited radiosensitization capacity. Quiescent cells are more radioresistant than proliferating cells, and these were also effectively sensitized by AZD7648 while Olaparib was unable to increase radiation-induced cell kill, even in BRCA1/2null cells. These findings underscore the distinct mechanisms of radiosensitization for DNA-PK and PARP inhibitors. While DNA-PK inhibitors are able to target both proliferating and non-proliferating tumor cells for greater overall anti-cancer benefit, their application is limited by exacerbation of normal tissue toxicities. Conversely, PARP inhibitors exhibit selective activity for proliferating cells, providing a mechanism for targeting activity to cancers, but due to poor activity in non-proliferating cells they have an overall reduced impact on tumor growth control. This study highlights the importance of creating a therapeutic ratio with DNA damage repair inhibition radiation sensitizing strategies.
Collapse
Affiliation(s)
- Taixiang Wang
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Alastair H Kyle
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Jennifer H E Baker
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Nannan A Liu
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Judit P Banáth
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Sevin Teymori
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Andrew I Minchinton
- BC Cancer Research Institute, Department of Integrative Oncology, 675W 10th Ave, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
4
|
Najafabadi MG, Gray GK, Kong LR, Gupta K, Perera D, Naylor H, Brugge JS, Venkitaraman AR, Shehata M. A transcriptional response to replication stress selectively expands a subset of Brca2-mutant mammary epithelial cells. Nat Commun 2023; 14:5206. [PMID: 37626143 PMCID: PMC10457340 DOI: 10.1038/s41467-023-40956-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Germline BRCA2 mutation carriers frequently develop luminal-like breast cancers, but it remains unclear how BRCA2 mutations affect mammary epithelial subpopulations. Here, we report that monoallelic Brca2mut/WT mammary organoids subjected to replication stress activate a transcriptional response that selectively expands Brca2mut/WT luminal cells lacking hormone receptor expression (HR-). While CyTOF analyses reveal comparable epithelial compositions among wildtype and Brca2mut/WT mammary glands, Brca2mut/WT HR- luminal cells exhibit greater organoid formation and preferentially survive and expand under replication stress. ScRNA-seq analysis corroborates the expansion of HR- luminal cells which express elevated transcript levels of Tetraspanin-8 (Tspan8) and Thrsp, plus pathways implicated in replication stress survival including Type I interferon responses. Notably, CRISPR/Cas9-mediated deletion of Tspan8 or Thrsp prevents Brca2mut/WT HR- luminal cell expansion. Our findings indicate that Brca2mut/WT cells activate a transcriptional response after replication stress that preferentially favours outgrowth of HR- luminal cells through the expression of interferon-responsive and mammary alveolar genes.
Collapse
Affiliation(s)
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA, USA
| | - Li Ren Kong
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, NUS School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, National University of Singapore, Singapore, Singapore
| | - Komal Gupta
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, National University of Singapore, Singapore, Singapore
| | - David Perera
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | - Huw Naylor
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA, USA
| | - Ashok R Venkitaraman
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Institute of Molecular & Cellular Biology Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
| | - Mona Shehata
- Department of Oncology, University of Cambridge, Cambridge, UK.
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
5
|
Wang T, Kyle AH, Baker JHE, Liu NA, Banáth JP, Minchinton AI. DNA-PK inhibition extends the therapeutic effects of Top2 poisoning to non-proliferating cells, increasing activity at a cost. Sci Rep 2023; 13:12429. [PMID: 37528151 PMCID: PMC10394067 DOI: 10.1038/s41598-023-39649-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023] Open
Abstract
Type II topoisomerase (Top2) poisoning therapy is used to treat a broad range of cancers via induction of double strand breaks (DSBs) in cells undergoing replication and transcription. Preventing the repair of DSBs via inhibition of DNA-PK, an inhibitor of non-homologous end-joining (NHEJ), increases cell kill with Top2 poisons and has led to the initiation of several clinical trials. To elucidate the cellular mechanisms leading to synergistic activity of dual DNA-PK/Top2 inhibition we looked at their effects in cycling versus non-cycling cells, in 3D spheroids and in xenograft models. Combined DNA-PK/Top2 inhibition was found to not only increase the cell kill in proliferating cells, the cell population that is typically most vulnerable to Top2 poisoning, but also in non-proliferative but transcriptionally active cells. This effect was observed in both cancer and normal tissue models, killing more cells than high concentrations of etoposide alone. The combination treatment delayed tumor growth in mice compared to Top2 poisoning alone, but also led to increased toxicity. These findings demonstrate sensitization of Top2β-expressing, non-cycling cells to Top2 poisoning by DNA-PK inhibition. Expansion of the target cell population of Top2 poison treatment to include non-proliferating cells via combination with DNA damage repair inhibitors has implications for efficacy and toxicity of these combinations, including for inhibitors of DNA-PK currently in clinical trial.
Collapse
Affiliation(s)
- Taixiang Wang
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Alastair H Kyle
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Jennifer H E Baker
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Nannan A Liu
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Judit P Banáth
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Andrew I Minchinton
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
6
|
Jiang Y, Zhang H, Wang J, Chen J, Guo Z, Liu Y, Hua H. Exploiting RIG-I-like receptor pathway for cancer immunotherapy. J Hematol Oncol 2023; 16:8. [PMID: 36755342 PMCID: PMC9906624 DOI: 10.1186/s13045-023-01405-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
RIG-I-like receptors (RLRs) are intracellular pattern recognition receptors that detect viral or bacterial infection and induce host innate immune responses. The RLRs family comprises retinoic acid-inducible gene 1 (RIG-I), melanoma differentiation-associated gene 5 (MDA5) and laboratory of genetics and physiology 2 (LGP2) that have distinctive features. These receptors not only recognize RNA intermediates from viruses and bacteria, but also interact with endogenous RNA such as the mislocalized mitochondrial RNA, the aberrantly reactivated repetitive or transposable elements in the human genome. Evasion of RLRs-mediated immune response may lead to sustained infection, defective host immunity and carcinogenesis. Therapeutic targeting RLRs may not only provoke anti-infection effects, but also induce anticancer immunity or sensitize "immune-cold" tumors to immune checkpoint blockade. In this review, we summarize the current knowledge of RLRs signaling and discuss the rationale for therapeutic targeting RLRs in cancer. We describe how RLRs can be activated by synthetic RNA, oncolytic viruses, viral mimicry and radio-chemotherapy, and how the RNA agonists of RLRs can be systemically delivered in vivo. The integration of RLRs agonism with RNA interference or CAR-T cells provides new dimensions that complement cancer immunotherapy. Moreover, we update the progress of recent clinical trials for cancer therapy involving RLRs activation and immune modulation. Further studies of the mechanisms underlying RLRs signaling will shed new light on the development of cancer therapeutics. Manipulation of RLRs signaling represents an opportunity for clinically relevant cancer therapy. Addressing the challenges in this field will help develop future generations of cancer immunotherapy.
Collapse
Affiliation(s)
- Yangfu Jiang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Hongying Zhang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Jinzhu Chen
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zeyu Guo
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongliang Liu
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Groelly FJ, Porru M, Zimmer J, Benainous H, De Visser Y, Kosova AA, Di Vito S, Serra V, Ryan A, Leonetti C, Bruna A, Biroccio A, Tarsounas M. Anti-tumoural activity of the G-quadruplex ligand pyridostatin against BRCA1/2-deficient tumours. EMBO Mol Med 2022; 14:e14501. [PMID: 35107878 PMCID: PMC8899905 DOI: 10.15252/emmm.202114501] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
The cells with compromised BRCA1 or BRCA2 (BRCA1/2) function accumulate stalled replication forks, which leads to replication‐associated DNA damage and genomic instability, a signature of BRCA1/2‐mutated tumours. Targeted therapies against BRCA1/2‐mutated tumours exploit this vulnerability by introducing additional DNA lesions. Because homologous recombination (HR) repair is abrogated in the absence of BRCA1 or BRCA2, these lesions are specifically lethal to tumour cells, but not to the healthy tissue. Ligands that bind and stabilise G‐quadruplexes (G4s) have recently emerged as a class of compounds that selectively eliminate the cells and tumours lacking BRCA1 or BRCA2. Pyridostatin is a small molecule that binds G4s and is specifically toxic to BRCA1/2‐deficient cells in vitro. However, its in vivo potential has not yet been evaluated. Here, we demonstrate that pyridostatin exhibits a high specific activity against BRCA1/2‐deficient tumours, including patient‐derived xenograft tumours that have acquired PARP inhibitor (PARPi) resistance. Mechanistically, we demonstrate that pyridostatin disrupts replication leading to DNA double‐stranded breaks (DSBs) that can be repaired in the absence of BRCA1/2 by canonical non‐homologous end joining (C‐NHEJ). Consistent with this, chemical inhibitors of DNA‐PKcs, a core component of C‐NHEJ kinase activity, act synergistically with pyridostatin in eliminating BRCA1/2‐deficient cells and tumours. Furthermore, we demonstrate that pyridostatin triggers cGAS/STING‐dependent innate immune responses when BRCA1 or BRCA2 is abrogated. Paclitaxel, a drug routinely used in cancer chemotherapy, potentiates the in vivo toxicity of pyridostatin. Overall, our results demonstrate that pyridostatin is a compound suitable for further therapeutic development, alone or in combination with paclitaxel and DNA‐PKcs inhibitors, for the benefit of cancer patients carrying BRCA1/2 mutations.
Collapse
Affiliation(s)
- Florian J Groelly
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Manuela Porru
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Jutta Zimmer
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Hugo Benainous
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yanti De Visser
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Anastasiya A Kosova
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Serena Di Vito
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy.,Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Anderson Ryan
- Lung Cancer Translational Science Research Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Carlo Leonetti
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alejandra Bruna
- Molecular Pathology Division, Centre for Cancer Evolution, The Institute of Cancer Research, London, UK
| | - Annamaria Biroccio
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Madalena Tarsounas
- Genome Stability and Tumourigenesis Group, The MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Akay M, Funingana IG, Patel G, Mustapha R, Gjafa E, Ng T, Ng K, Flynn MJ. An In-Depth Review of Niraparib in Ovarian Cancer: Mechanism of Action, Clinical Efficacy and Future Directions. Oncol Ther 2021; 9:347-364. [PMID: 34363200 PMCID: PMC8593085 DOI: 10.1007/s40487-021-00167-z] [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: 06/21/2021] [Accepted: 07/22/2021] [Indexed: 01/24/2023] Open
Abstract
Niraparib is an oral, potent, highly selective poly-ADP ribose polymerase 1 (PARP1) and PARP2 inhibitor. In most developed countries, it is approved as a maintenance treatment for epithelial ovarian, fallopian tube, or primary peritoneal cancer in patients with complete or partial response to platinum-based therapy. These approvals are based on results of randomised, double-blind, placebo-controlled trials, particularly the NOVA trial and more recently the PRIMA trial. In this comprehensive review, we delve into the scientific basis of PARP inhibition, discussing both preclinical and clinical data which have led to the current approval status of niraparib. We also discuss ongoing trials and biological rationale of combination treatments involving niraparib, with particular focus on antiangiogenic drugs, immune checkpoint inhibitors and cyclic GMP-AMP synthase stimulator of interferon genes (cGAS/STING) pathway. In addition, we reflect on potential strategies and challenges of utilising current biomarkers for treatment selection of patients to ensure maximal benefit.
Collapse
Affiliation(s)
- Melek Akay
- Department of Medical Oncology, St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Grisma Patel
- Department of Medical Oncology, University College London Hospitals, London, UK
| | - Rami Mustapha
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
- Cancer Research UK King's Health Partners Centre, London, UK
| | - Ernese Gjafa
- Department of Medical Oncology, Barts Health NHS Trust, London, UK
| | - Tony Ng
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
- Cancer Research UK King's Health Partners Centre, London, UK
- Cancer Institute, University College London, 72 Huntley Street, London, UK
| | - Kenrick Ng
- Department of Medical Oncology, University College London Hospitals, London, UK.
- Cancer Institute, University College London, 72 Huntley Street, London, UK.
| | - Michael J Flynn
- Department of Medical Oncology, University College London Hospitals, London, UK
| |
Collapse
|
9
|
Funingana IG, Reinius MAV, Petrillo A, Ang JE, Brenton JD. Can integrative biomarker approaches improve prediction of platinum and PARP inhibitor response in ovarian cancer? Semin Cancer Biol 2021; 77:67-82. [PMID: 33607245 DOI: 10.1016/j.semcancer.2021.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 12/28/2022]
Abstract
Epithelial ovarian carcinoma (EOC) encompasses distinct histological, molecular and genomic entities that determine intrinsic sensitivity to platinum-based chemotherapy. Current management of each subtype is determined by factors including tumour grade and stage, but only a small number of biomarkers can predict treatment response. The recent incorporation of PARP inhibitors into routine clinical practice has underscored the need to personalise ovarian cancer treatment based on tumour biology. In this article, we review the strengths and limitations of predictive biomarkers in current clinical practice and highlight integrative strategies that may inform the development of future personalised medicine programs and composite biomarkers.
Collapse
Affiliation(s)
- Ionut-Gabriel Funingana
- Department of Oncology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Marika A V Reinius
- Department of Oncology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Angelica Petrillo
- Medical Oncology Unit, Ospedale del Mare, Naples, Italy; University of Study of Campania "L.Vanvitelli", Naples, Italy.
| | - Joo Ern Ang
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James D Brenton
- Department of Oncology, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK; Department of Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Identifying Biomarkers of Alzheimer's Disease via a Novel Structured Sparse Canonical Correlation Analysis Approach. J Mol Neurosci 2021; 72:323-335. [PMID: 34570360 DOI: 10.1007/s12031-021-01915-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/09/2021] [Indexed: 02/05/2023]
Abstract
Using correlation analysis to study the potential connection between brain genetics and imaging has become an effective method to understand neurodegenerative diseases. Sparse canonical correlation analysis (SCCA) makes it possible to study high-dimensional genetic information. The traditional SCCA methods can only process single-modal genetic and image data, which to some extent weaken the close connection of the brain's biological network. In some recently proposed multimodal SCCA methods, due to the limitations of penalty items, the pre-processed data needs to be further filtered to make the dimensions uniform, which may destroy the potential association of data in the same modal. In this research, in order to combine data between different modalities and to ensure that the chain relationship or graph network relationship within the same modality will not be destroyed, the original generalized fused lasso penalty was replaced with the fused pairwise group lasso (FGL) and the graph-guided pairwise group lasso (GGL) based on the method of joint sparse canonical correlation analysis (JSCCA). We used prior knowledge to construct a supervised bivariate learning model and use linear regression to select quantitative traits (QTs) of images that are strongly correlated with the Mini-mental State Examination (MMSE) scores. Compared with FGL-SCCA, the model we constructed obtained a higher gene-ROI correlation coefficient and identified more significant biomarkers, providing a theoretical basis for further understanding the complex pathology of neurodegenerative diseases.
Collapse
|
11
|
Lee JM, Botesteanu DA, Tomita Y, Yuno A, Lee MJ, Kohn EC, Annunziata CM, Matulonis U, MacDonald LA, Nair JR, Macneill KM, Trepel JB. Patients with BRCA mutated ovarian cancer may have fewer circulating MDSC and more peripheral CD8 + T cells compared with women with BRCA wild-type disease during the early disease course. Oncol Lett 2019; 18:3914-3924. [PMID: 31516602 DOI: 10.3892/ol.2019.10731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022] Open
Abstract
Immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) are associated with immunologic tolerance and poor prognosis in ovarian cancer (OvCa). We hypothesized that women with germline BRCA1 and BRCA2 mutation-associated (gBRCAm) OvCa would have fewer circulating immunosuppressive immune cells compared to those with BRCA wild-type (BRCAwt) disease during their early disease course (<5 years post-diagnosis) where gBRCAm is a favorable prognostic factor. We collected and viably froze peripheral blood mononuclear cells (PBMCs) from patients with recurrent OvCa olaparib clinical trials (NCT01445418/NCT01237067). Immune subset analyses were performed using flow cytometry for Tregs, exhausted CD8+ T cells, monocytes and MDSCs. Functional marker expression, including cytotoxic T lymphocyte-associated protein 4 (CTLA-4), T cell immunoglobulin and mucin domain 3 (TIM-3) and programmed cell death protein 1 (PD-1) was evaluated. Data were analyzed using FlowJo. Pretreatment PBMCs were collected from 41 patients (16 gBRCAm/25 BRCAwt). The percentage of MDSCs among viable CD45+ PBMC was lower in gBRCAm OvCa compared with BRCAwt OvCa (median 0.565 vs. 0.93%, P=0.0086) but this difference was not seen in those women >5 years post-diagnosis. CD8+ T cells among viable CD45+ PBMCs and CTLA-4+/CD8+ T cells were higher in gBRCAm carriers than patients with BRCAwt, in particular for those <5 years post-diagnosis (median 20.4 vs. 9.78%, P=0.031 and median MFI 0.19 vs. 0.22, P=0.0074, respectively). TIM-3 expression on Tregs was associated with poor progression-free survival, independent of gBRCAm status (P<0.001). Our pilot data suggested that patients with gBRCAm OvCa may have fewer circulating MDSCs but higher CD8+ T cells in PBMCs during their early disease course. This may contribute to the observed survival benefit for these women in their first post-diagnosis decade.
Collapse
Affiliation(s)
- Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Dana-Adriana Botesteanu
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yusuke Tomita
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Akira Yuno
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Elise C Kohn
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ursula Matulonis
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Lauren A MacDonald
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Jayakumar R Nair
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Kimberley M Macneill
- Division of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Jane B Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Reisländer T, Lombardi EP, Groelly FJ, Miar A, Porru M, Di Vito S, Wright B, Lockstone H, Biroccio A, Harris A, Londoño-Vallejo A, Tarsounas M. BRCA2 abrogation triggers innate immune responses potentiated by treatment with PARP inhibitors. Nat Commun 2019; 10:3143. [PMID: 31316060 PMCID: PMC6637138 DOI: 10.1038/s41467-019-11048-5] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/18/2019] [Indexed: 01/21/2023] Open
Abstract
Heterozygous germline mutations in BRCA2 predispose to breast and ovarian cancer. Contrary to non-cancerous cells, where BRCA2 deletion causes cell cycle arrest or cell death, tumors carrying BRCA2 inactivation continue to proliferate. Here we set out to investigate adaptation to loss of BRCA2 focusing on genome-wide transcriptome alterations. Human cells in which BRCA2 expression is inhibited for 4 or 28 days are subjected to RNA-seq analyses revealing a biphasic response to BRCA2 abrogation. The early, acute response consists of downregulation of genes involved in cell cycle progression, DNA replication and repair and is associated with cell cycle arrest in G1. Surprisingly, the late, chronic response consists predominantly of upregulation of interferon-stimulated genes (ISGs). Activation of the cGAS-STING-STAT pathway detected in these cells further substantiates the concept that BRCA2 abrogation triggers cell-intrinsic immune signaling. Importantly, we find that treatment with PARP inhibitors stimulates the interferon response in cells and tumors lacking BRCA2.
Collapse
Affiliation(s)
- Timo Reisländer
- Genome Stability and Tumourigenesis Group, The CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Emilia Puig Lombardi
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR3244, F-75005, Paris, France
| | - Florian J Groelly
- Genome Stability and Tumourigenesis Group, The CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Ana Miar
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
| | - Manuela Porru
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Serena Di Vito
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Benjamin Wright
- Bioinformatics and Statistical Genetics Core, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Helen Lockstone
- Bioinformatics and Statistical Genetics Core, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Annamaria Biroccio
- Area of Translational Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Adrian Harris
- Hypoxia and Angiogenesis Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, Oxford, OX3 9DS, UK
| | - Arturo Londoño-Vallejo
- Institut Curie, PSL Research University, CNRS, UMR3244, F-75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR3244, F-75005, Paris, France
| | - Madalena Tarsounas
- Genome Stability and Tumourigenesis Group, The CR-UK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| |
Collapse
|
13
|
Tacconi EMC, Badie S, De Gregoriis G, Reisländer T, Lai X, Porru M, Folio C, Moore J, Kopp A, Baguña Torres J, Sneddon D, Green M, Dedic S, Lee JW, Batra AS, Rueda OM, Bruna A, Leonetti C, Caldas C, Cornelissen B, Brino L, Ryan A, Biroccio A, Tarsounas M. Chlorambucil targets BRCA1/2-deficient tumours and counteracts PARP inhibitor resistance. EMBO Mol Med 2019; 11:e9982. [PMID: 31273933 PMCID: PMC6609913 DOI: 10.15252/emmm.201809982] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 01/03/2023] Open
Abstract
Due to compromised homologous recombination (HR) repair, BRCA1- and BRCA2-mutated tumours accumulate DNA damage and genomic rearrangements conducive of tumour progression. To identify drugs that target specifically BRCA2-deficient cells, we screened a chemical library containing compounds in clinical use. The top hit was chlorambucil, a bifunctional alkylating agent used for the treatment of chronic lymphocytic leukaemia (CLL). We establish that chlorambucil is specifically toxic to BRCA1/2-deficient cells, including olaparib-resistant and cisplatin-resistant ones, suggesting the potential clinical use of chlorambucil against disease which has become resistant to these drugs. Additionally, chlorambucil eradicates BRCA2-deficient xenografts and inhibits growth of olaparib-resistant patient-derived tumour xenografts (PDTXs). We demonstrate that chlorambucil inflicts replication-associated DNA double-strand breaks (DSBs), similarly to cisplatin, and we identify ATR, FANCD2 and the SNM1A nuclease as determinants of sensitivity to both drugs. Importantly, chlorambucil is substantially less toxic to normal cells and tissues in vitro and in vivo relative to cisplatin. Because chlorambucil and cisplatin are equally effective inhibitors of BRCA2-compromised tumours, our results indicate that chlorambucil has a higher therapeutic index than cisplatin in targeting BRCA-deficient tumours.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/deficiency
- BRCA2 Protein/deficiency
- Cell Line, Tumor
- Chlorambucil/pharmacology
- Cricetinae
- Drug Delivery Systems
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Male
- Mice
- Mice, SCID
- Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors
- Peroxisome Proliferator-Activated Receptors/metabolism
- Phthalazines/pharmacology
- Piperazines/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Eliana MC Tacconi
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Sophie Badie
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Giuliana De Gregoriis
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Timo Reisländer
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Xianning Lai
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Manuela Porru
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Cecilia Folio
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - John Moore
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Arnaud Kopp
- Institut de Génétique et de Biologie Cellulaire et Moléculaire (IGBMC)Inserm U1258, CNRS (UMR 7104)Université de StrasbourgIllkirchFrance
| | - Júlia Baguña Torres
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Deborah Sneddon
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Marcus Green
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Simon Dedic
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Jonathan W Lee
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Ankita Sati Batra
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Oscar M Rueda
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Alejandra Bruna
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Carlo Leonetti
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Carlos Caldas
- Department of OncologyCancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Bart Cornelissen
- Radiopharmaceuticals and Molecular Imaging GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Laurent Brino
- Institut de Génétique et de Biologie Cellulaire et Moléculaire (IGBMC)Inserm U1258, CNRS (UMR 7104)Université de StrasbourgIllkirchFrance
| | - Anderson Ryan
- Lung Cancer Translational Science Research GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| | - Annamaria Biroccio
- Area of Translational ResearchIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Madalena Tarsounas
- Genome Stability and Tumorigenesis GroupDepartment of OncologyThe CR‐UK/MRC Oxford Institute for Radiation OncologyUniversity of OxfordOxfordUK
| |
Collapse
|
14
|
DNA Repair Deficiency in Breast Cancer: Opportunities for Immunotherapy. JOURNAL OF ONCOLOGY 2019; 2019:4325105. [PMID: 31320901 PMCID: PMC6607732 DOI: 10.1155/2019/4325105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/04/2019] [Accepted: 05/29/2019] [Indexed: 12/19/2022]
Abstract
Historically the development of anticancer treatments has been focused on their effect on tumor cells alone. However, newer treatments have shifted attention to targets on immune cells, resulting in dramatic responses. The effect of DNA repair deficiency on the microenvironment remains an area of key interest. Moreover, established therapies such as DNA damaging treatments such as chemotherapy and PARP inhibitors further modify the tumor microenvironment. Here we describe DNA repair pathways in breast cancer and activation of innate immune pathways in DNA repair deficiency, in particular, the STING (STimulator of INterferon Genes) pathway. Breast tumors with DNA repair deficiency are associated with upregulation of immune checkpoints including PD-L1 (Programmed Death Ligand-1) and may represent a target population for single agent or combination immunotherapy treatment.
Collapse
|
15
|
Olney KC, Nyer DB, Vargas DA, Wilson Sayres MA, Haynes KA. The synthetic histone-binding regulator protein PcTF activates interferon genes in breast cancer cells. BMC SYSTEMS BIOLOGY 2018; 12:83. [PMID: 30253781 PMCID: PMC6156859 DOI: 10.1186/s12918-018-0608-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
Background Mounting evidence from genome-wide studies of cancer shows that chromatin-mediated epigenetic silencing at large cohorts of genes is strongly linked to a poor prognosis. This mechanism is thought to prevent cell differentiation and enable evasion of the immune system. Drugging the cancer epigenome with small molecule inhibitors to release silenced genes from the repressed state has emerged as a powerful approach for cancer research and drug development. Targets of these inhibitors include chromatin-modifying enzymes that can acquire drug-resistant mutations. In order to directly target a generally conserved feature, elevated trimethyl-lysine 27 on histone H3 (H3K27me3), we developed the Polycomb-based Transcription Factor (PcTF), a fusion activator that targets methyl-histone marks via its N-terminal H3K27me3-binding motif, and co-regulates sets of silenced genes. Results Here, we report transcriptome profiling analyses of PcTF-treated breast cancer model cell lines. We identified a set of 19 PcTF-upregulated genes, or PUGs, that were consistent across three distinct breast cancer cell lines. These genes are associated with the interferon response pathway. Conclusions Our results demonstrate for the first time a chromatin-mediated interferon-related transcriptional response driven by an engineered fusion protein that physically links repressive histone marks with active transcription. Electronic supplementary material The online version of this article (10.1186/s12918-018-0608-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kimberly C Olney
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, 85287-4501, AZ, USA
| | - David B Nyer
- School of Biological and Health Systems Engineering, Arizona State University, 501 E Tyler Mall, Tempe, AZ, 85287-9709, USA
| | - Daniel A Vargas
- School of Biological and Health Systems Engineering, Arizona State University, 501 E Tyler Mall, Tempe, AZ, 85287-9709, USA
| | - Melissa A Wilson Sayres
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, 85287-4501, AZ, USA.,Center for Evolution and Medicine, Arizona State University, 427 E Tyler Mall, Tempe, 85287-1701, AZ, USA
| | - Karmella A Haynes
- School of Biological and Health Systems Engineering, Arizona State University, 501 E Tyler Mall, Tempe, AZ, 85287-9709, USA.
| |
Collapse
|
16
|
Hollingsworth J, Lau A, Tone A, Kollara A, Allen L, Colgan TJ, Dube V, Rosen B, Murphy KJ, Greenblatt EM, Feigenberg T, Virtanen C, Brown TJ. BRCA1 Mutation Status and Follicular Fluid Exposure Alters NFκB Signaling and ISGylation in Human Fallopian Tube Epithelial Cells. Neoplasia 2018; 20:697-709. [PMID: 29852322 PMCID: PMC6030391 DOI: 10.1016/j.neo.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/07/2018] [Accepted: 05/11/2018] [Indexed: 12/31/2022] Open
Abstract
Germline BRCA1 or BRCA2 mutations (mtBRCA1 and mtBRCA2) increase risk for high-grade serous ovarian cancer (HGSOC), the most commonly diagnosed epithelial ovarian cancer histotype. Other identified risk factors for this cancer, which originates primarily in the distal fallopian tube epithelium (FTE), implicate ovulation, during which the FTE cells become transiently exposed to follicular fluid (FF). To test whether mtBRCA1 or mtBRCA2 nonmalignant FTE cells respond differently to periovulatory FF exposure than control patient FTE cells, gene expression profiles from primary FTE cultures derived from BRCA1 or BRCA2 mutation carriers or control patients were compared at baseline, 24 hours after FF exposure, and 24 hours after FF replacement with culture medium. Hierarchical clustering revealed both FF exposure and BRCA mutation status affect gene expression, with BRCA1 mutation having the greatest impact. Gene set enrichment analysis revealed increased NFκB and EGFR signaling at baseline in mtBRCA1 samples, with increased interferon target gene expression, including members of the ISGylation pathway, observed after recovery from FF exposure. Gene set enrichment analysis did not identify altered pathway signaling in mtBRCA2 samples. An inverse relationship between EGFR signaling and ISGylation with BRCA1 protein levels was verified in an immortalized FTE cell line, OE-E6/E7, stably transfected with BRCA1 cDNA. Suppression of ISG15 and ISGylated protein levels by increased BRCA1 expression was found to be mediated by decreased NFκB signaling. These studies indicate that increased NFκB signaling associated with decreased BRCA1 expression results in increased ISG15 and protein ISGylation following FF exposure, which may be involved in predisposition to HGSOC.
Collapse
Affiliation(s)
- Julia Hollingsworth
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Institute of Medical Sciences, University of Toronto, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Angela Lau
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Department of Physiology, University of Toronto, Toronto, ON
| | - Alicia Tone
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - Alexandra Kollara
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Lisa Allen
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Terence J Colgan
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON
| | - Valerie Dube
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON; Department of Pathology, Women's College Hospital, Toronto, ON
| | - Barry Rosen
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - K Joan Murphy
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | - Ellen M Greenblatt
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON
| | - Tomer Feigenberg
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Division of Gynecologic Oncology, Princess Margaret Cancer Centre, Toronto, ON
| | | | - Theodore J Brown
- The Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON; Institute of Medical Sciences, University of Toronto, Toronto, ON; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON; Department of Physiology, University of Toronto, Toronto, ON.
| |
Collapse
|
17
|
Clifford C, Vitkin N, Nersesian S, Reid-Schachter G, Francis JA, Koti M. Multi-omics in high-grade serous ovarian cancer: Biomarkers from genome to the immunome. Am J Reprod Immunol 2018; 80:e12975. [DOI: 10.1111/aji.12975] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/16/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Cole Clifford
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
| | - Natasha Vitkin
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
- Cancer Biology and Genetics; Queen's Cancer Research Institute; Queen's University; Kingston ON Canada
| | - Sarah Nersesian
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
- Cancer Biology and Genetics; Queen's Cancer Research Institute; Queen's University; Kingston ON Canada
| | | | - Julie-Ann Francis
- Department of Obstetrics and Gynecology; Kingston Health Sciences Center; Queen's University; Kingston ON Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences; Queen's University; Kingston ON Canada
- Cancer Biology and Genetics; Queen's Cancer Research Institute; Queen's University; Kingston ON Canada
- Department of Obstetrics and Gynecology; Kingston Health Sciences Center; Queen's University; Kingston ON Canada
| |
Collapse
|
18
|
Prosser KE, Leung AWY, Harrypersad S, Lewis AR, Bally MB, Walsby CJ. Transition Metal Ions Promote the Bioavailability of Hydrophobic Therapeutics: Cu and Zn Interactions with RNA Polymerase I Inhibitor CX5461. Chemistry 2018; 24:6334-6338. [PMID: 29490115 DOI: 10.1002/chem.201800289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Indexed: 01/27/2023]
Abstract
Low aqueous solubility is a major barrier to the clinical application of otherwise promising drug candidates. We demonstrate that this issue can be resolved in medicinal molecules containing potential ligating groups, through the addition of labile transition-metal ions. Incubation of the chemotherapeutic CX5461 with Cu2+ or Zn2+ enables solubilization at neutral pH but does not affect intrinsic cytotoxicity. Spectroscopic and computational studies demonstrate that this arises from coordination to the pyrazine functionality of CX5461 and may involve bidentate coordination at physiological pH.
Collapse
Affiliation(s)
- Kathleen E Prosser
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada
| | - Ada W Y Leung
- Department of Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Ave, Vancouver, BC, V5Z 4E6, Canada
| | - Shane Harrypersad
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada
| | - Andrew R Lewis
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada
| | - Marcel B Bally
- Department of Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Ave, Vancouver, BC, V5Z 4E6, Canada
| | - Charles J Walsby
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada
| |
Collapse
|
19
|
Ghosh R, Roy S, Franco S. PARP1 depletion induces RIG-I-dependent signaling in human cancer cells. PLoS One 2018; 13:e0194611. [PMID: 29590171 PMCID: PMC5874037 DOI: 10.1371/journal.pone.0194611] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/06/2018] [Indexed: 12/11/2022] Open
Abstract
DNA Damage Response (DDR) and DNA repair pathways are emerging as potent, ubiquitous suppressors of innate immune signaling in human cells. Here, we show that human cells surviving depletion of the Single Strand Break (SSB) repair protein PARP1 undergo p21-dependent senescence or cell cycle checkpoint activation in the context of activation of innate immune signaling, or viral mimicry. Specifically, we observe induction of a large number of interferon-stimulated genes (ISGs) and multiple pattern recognition receptors (PRRs; including RIG-I, MDA-5, MAVS, TLR3 and STING) and increased nuclear IRF3 staining. Mechanistically, depletion of the double-stranded RNA (dsRNA) helicase RIG-I or its downstream effector MAVS specifically rescues ISG induction in PARP1-depleted cells, suggesting that the RIG-I/MAVS pathway is required for sustained ISG expression in this context. Experiments with conditioned media or a neutralizing antibody to the α/β-IFN receptor revealed that persistent ISG expression additionally requires an autocrine/paracrine loop. Finally, loss of PARP1 and radiation-induced DNA damage strongly synergize in the induction of p21 and ISGs. Overall, these findings increase our understanding of how PARP1 may suppress deleterious phenotypes associated to aging, inflammation and cancer in humans.
Collapse
Affiliation(s)
- Rajib Ghosh
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sanchita Roy
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Sonia Franco
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
20
|
Li N, Yang Y, Liang C, Qiu Q, Pan C, Li M, Yang S, Chen L, Zhu X, Hu Y. Tmem30a Plays Critical Roles in Ensuring the Survival of Hematopoietic Cells and Leukemia Cells in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1457-1468. [PMID: 29574182 DOI: 10.1016/j.ajpath.2018.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/03/2018] [Accepted: 02/27/2018] [Indexed: 02/05/2023]
Abstract
The fundamental structure of eukaryotic cell plasma membrane is the phospholipid bilayer, which contains four major phospholipids. These phospholipids are asymmetrically distributed between the outer and inner leaflets. P4-ATPase flippase complexes play essential roles in ensuring this asymmetry. We found that conditional deletion of Tmem30a, the β subunit of P4-ATPase flippase complex, caused pancytopenia in mice. Tmem30a deficiency resulted in depletion of lineage-committed blood cells in the peripheral blood, spleen, and bone marrow. Ablation of Tmem30a also caused the depletion of hematopoietic stem cells (HSCs). HSC RNA sequencing results revealed that multiple biological processes and signal pathways were involved in the event, including mammalian target of rapamycin signaling, genes for HSC stemness, and genes responding to interferons. Our results also revealed that targeting Tmem30a signaling had therapeutic utility in BCR/ABL1-induced chronic myeloid leukemia.
Collapse
Affiliation(s)
- Ning Li
- Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Cailing Liang
- Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiang Qiu
- Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cong Pan
- Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mengyuan Li
- Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; Chengdu Institute of Biology, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China.
| | - Yiguo Hu
- Department of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
21
|
Xu H, Di Antonio M, McKinney S, Mathew V, Ho B, O'Neil NJ, Santos ND, Silvester J, Wei V, Garcia J, Kabeer F, Lai D, Soriano P, Banáth J, Chiu DS, Yap D, Le DD, Ye FB, Zhang A, Thu K, Soong J, Lin SC, Tsai AHC, Osako T, Algara T, Saunders DN, Wong J, Xian J, Bally MB, Brenton JD, Brown GW, Shah SP, Cescon D, Mak TW, Caldas C, Stirling PC, Hieter P, Balasubramanian S, Aparicio S. CX-5461 is a DNA G-quadruplex stabilizer with selective lethality in BRCA1/2 deficient tumours. Nat Commun 2017; 8:14432. [PMID: 28211448 PMCID: PMC5321743 DOI: 10.1038/ncomms14432] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/28/2016] [Indexed: 12/29/2022] Open
Abstract
G-quadruplex DNAs form four-stranded helical structures and are proposed to play key roles in different cellular processes. Targeting G-quadruplex DNAs for cancer treatment is a very promising prospect. Here, we show that CX-5461 is a G-quadruplex stabilizer, with specific toxicity against BRCA deficiencies in cancer cells and polyclonal patient-derived xenograft models, including tumours resistant to PARP inhibition. Exposure to CX-5461, and its related drug CX-3543, blocks replication forks and induces ssDNA gaps or breaks. The BRCA and NHEJ pathways are required for the repair of CX-5461 and CX-3543-induced DNA damage and failure to do so leads to lethality. These data strengthen the concept of G4 targeting as a therapeutic approach, specifically for targeting HR and NHEJ deficient cancers and other tumours deficient for DNA damage repair. CX-5461 is now in advanced phase I clinical trial for patients with BRCA1/2 deficient tumours (Canadian trial, NCT02719977, opened May 2016).
Collapse
Affiliation(s)
- Hong Xu
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Marco Di Antonio
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Steven McKinney
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Veena Mathew
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Brandon Ho
- Department of Biochemistry and Donnelly Centre, University of Toronto, 160 College Street, Toronto, Ontario, Canada M5S 3E1
| | - Nigel J. O'Neil
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada V6T 1Z4
| | - Nancy Dos Santos
- Advanced Therapeutics, BC Cancer Agency and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Jennifer Silvester
- Campbell Family Institute for Breast Cancer Research, Princess Margret Cancer Centre, 610 University Avenue, Toronto, Canada M5G 2M9
| | - Vivien Wei
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Jessica Garcia
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Farhia Kabeer
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Daniel Lai
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Priscilla Soriano
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Judit Banáth
- Department of Integrative Oncology, BC Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Derek S. Chiu
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Damian Yap
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Daniel D. Le
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Frank B. Ye
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada V6T 1Z4
| | - Anni Zhang
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Kelsie Thu
- Campbell Family Institute for Breast Cancer Research, Princess Margret Cancer Centre, 610 University Avenue, Toronto, Canada M5G 2M9
| | - John Soong
- Senhwa Biosciences, Inc., 9 F, No.205-1, Section 3, Peihsin Road, Hsintien District, New Taipei City 23143, Taiwan R.O.C
| | - Shu-chuan Lin
- Senhwa Biosciences, Inc., 9 F, No.205-1, Section 3, Peihsin Road, Hsintien District, New Taipei City 23143, Taiwan R.O.C
| | - Angela Hsin Chin Tsai
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Tomo Osako
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Teresa Algara
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Darren N. Saunders
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Jason Wong
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Jian Xian
- Cancer Research UK Cambridge Research Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Marcel B. Bally
- Advanced Therapeutics, BC Cancer Agency and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - James D. Brenton
- Cancer Research UK Cambridge Research Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Grant W. Brown
- Department of Biochemistry and Donnelly Centre, University of Toronto, 160 College Street, Toronto, Ontario, Canada M5S 3E1
| | - Sohrab P. Shah
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - David Cescon
- Campbell Family Institute for Breast Cancer Research, Princess Margret Cancer Centre, 610 University Avenue, Toronto, Canada M5G 2M9
- Division of Medical Oncology and Hematology, Department of Medicine, University of Toronto, Toronto, Canada M5S 1A8
| | - Tak W. Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margret Cancer Centre, 610 University Avenue, Toronto, Canada M5G 2M9
| | - Carlos Caldas
- Cancer Research UK Cambridge Research Institute and Department of Oncology, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Peter C. Stirling
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| | - Phil Hieter
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada V6T 1Z4
| | - Shankar Balasubramanian
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Samuel Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, and Department of Pathology and Laboratory Medicine, University of British Columbia, 675 West 10th Avenue, Vancouver, British Columbia, Canada V5Z 1L3
| |
Collapse
|
22
|
Genomic amplification of Fanconi anemia complementation group A (FancA) in head and neck squamous cell carcinoma (HNSCC): Cellular mechanisms of radioresistance and clinical relevance. Cancer Lett 2016; 386:87-99. [PMID: 27867017 DOI: 10.1016/j.canlet.2016.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/25/2016] [Accepted: 11/10/2016] [Indexed: 01/06/2023]
Abstract
Radio (chemo) therapy is a crucial treatment modality for head and neck squamous cell carcinoma (HNSCC), but relapse is frequent, and the underlying mechanisms remain largely elusive. Therefore, novel biomarkers are urgently needed. Previously, we identified gains on 16q23-24 to be associated with amplification of the Fanconi anemia A (FancA) gene and to correlate with reduced progression-free survival after radiotherapy. Here, we analyzed the effects of FancA on radiation sensitivity in vitro, characterized the underlying mechanisms, and evaluated their clinical relevance. Silencing of FancA expression in HNSCC cell lines with genomic gains on 16q23-24 resulted in significantly impaired clonogenic survival upon irradiation. Conversely, overexpression of FancA in immortalized keratinocytes conferred increased survival accompanied by improved DNA repair, reduced accumulation of chromosomal translocations, but no hyperactivation of the FA/BRCA-pathway. Downregulation of interferon signaling as identified by microarray analyses, enforced irradiation-induced senescence, and elevated production of the senescence-associated secretory phenotype (SASP) appeared to be candidate mechanisms contributing to FancA-mediated radioresistance. Data of the TCGA HNSCC cohort confirmed the association of gains on 16q24.3 with FancA overexpression and impaired overall survival. Importantly, transcriptomic alterations similar to those observed upon FancA overexpression in vitro strengthened the clinical relevance. Overall, FancA amplification and overexpression appear to be crucial for radiotherapeutic failure in HNSCC.
Collapse
|
23
|
Legrier ME, Bièche I, Gaston J, Beurdeley A, Yvonnet V, Déas O, Thuleau A, Château-Joubert S, Servely JL, Vacher S, Lassalle M, Depil S, Tucker GC, Fontaine JJ, Poupon MF, Roman-Roman S, Judde JG, Decaudin D, Cairo S, Marangoni E. Activation of IFN/STAT1 signalling predicts response to chemotherapy in oestrogen receptor-negative breast cancer. Br J Cancer 2015; 114:177-87. [PMID: 26695443 PMCID: PMC4815803 DOI: 10.1038/bjc.2015.398] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/07/2015] [Accepted: 10/15/2015] [Indexed: 12/13/2022] Open
Abstract
Background: Oestrogen receptor-negative (ER−) breast cancer is intrinsically sensitive to chemotherapy. However, tumour response is often incomplete, and relapse occurs with high frequency. The aim of this work was to analyse the molecular characteristics of residual tumours and early response to chemotherapy in patient-derived xenografts (PDXs) of breast cancer. Methods: Gene and protein expression profiles were analysed in a panel of ER− breast cancer PDXs before and after chemotherapy treatment. Tumour and stromal interferon-gamma expression was measured in xenografts lysates by human and mouse cytokine arrays, respectively. Results: The analysis of residual tumour cells in chemo-responder PDX revealed a strong overexpression of IFN-inducible genes, induced early after AC treatment and associated with increased STAT1 phosphorylation, DNA-damage and apoptosis. No increase in IFN-inducible gene expression was observed in chemo-resistant PDXs upon chemotherapy. Overexpression of IFN-related genes was associated with human IFN-γ secretion by tumour cells. Conclusions: Treatment-induced activation of the IFN/STAT1 pathway in tumour cells is associated with chemotherapy response in ER− breast cancer. Further validations in prospective clinical trials will aim to evaluate the usefulness of this signature to assist therapeutic strategies in the clinical setting.
Collapse
Affiliation(s)
| | - Ivan Bièche
- Genetics Department, Hospital, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| | - Julie Gaston
- XenTech, 4 rue Pierre Fontaine, Evry 91000, France
| | | | | | - Olivier Déas
- XenTech, 4 rue Pierre Fontaine, Evry 91000, France
| | - Aurélie Thuleau
- Translational Research Department, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| | | | - Jean-Luc Servely
- Department of Pathology, Veterinary School of Alfort, Maisons-Alfort 94704, France.,INRA, Phase Department, Nouzilly, France
| | - Sophie Vacher
- Genetics Department, Hospital, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| | | | - Stéphane Depil
- Institut de Recherches Servier, PIT Oncology, Croissy-sur-Seine 78290, France
| | - Gordon C Tucker
- Institut de Recherches Servier, PIT Oncology, Croissy-sur-Seine 78290, France
| | | | | | - Sergio Roman-Roman
- Translational Research Department, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| | | | - Didier Decaudin
- Translational Research Department, Institut Curie, 26 rue d'Ulm, Paris 75005, France.,Medical Oncology Department, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| | | | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, 26 rue d'Ulm, Paris 75005, France
| |
Collapse
|
24
|
O'Malley P, Mulla Z, Nesic O. Multiple sclerosis and breast cancer. J Neurol Sci 2015; 356:137-41. [DOI: 10.1016/j.jns.2015.06.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 05/30/2015] [Accepted: 06/13/2015] [Indexed: 02/09/2023]
|