1
|
Gederaas OA, Sharma A, Mbarak S, Sporsheim B, Høgset A, Bogoeva V, Slupphaug G, Hagen L. Proteomic analysis reveals mechanisms underlying increased efficacy of bleomycin by photochemical internalization in bladder cancer cells. Mol Omics 2023; 19:585-597. [PMID: 37345535 DOI: 10.1039/d2mo00337f] [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: 06/23/2023]
Abstract
Photochemical internalization (PCI) is a promising new technology for site-specific drug delivery, developed from photodynamic therapy (PDT). In PCI, light-induced activation of a photosensitizer trapped inside endosomes together with e.g. chemotherapeutics, nucleic acids or immunotoxins, allows cytosolic delivery and enhanced local therapeutic effect. Here we have evaluated the photosensitizer meso-tetraphenyl chlorine disulphonate (TPCS2a/fimaporfin) in a proteome analysis of AY-27 rat bladder cancer cells in combination with the chemotherapeutic drug bleomycin (BML). We find that BLMPCI attenuates oxidative stress responses induced by BLM alone, while concomitantly increasing transcriptional repression and DNA damage responses. BLMPCI also mediates downregulation of bleomycin hydrolase (Blmh), which is responsible for cellular degradation of BLM, as well as several factors known to be involved in fibrotic responses. PCI-mediated delivery might thus allow reduced dosage of BLM and alleviate unwanted side effects from treatment, including pulmonary fibrosis.
Collapse
Affiliation(s)
- Odrun A Gederaas
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
- Department of Natural Sciences, UiA, University of Agder, N-4630, Kristiansand, Norway.
| | - Animesh Sharma
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
- Proteomics and Modomics Experimental Core, PROMEC, at NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Saide Mbarak
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
| | - Bjørnar Sporsheim
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
- CMIC Cellular & Molecular Imaging Core Facility, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, Trondheim, Norway
| | - Anders Høgset
- PCI Biotech AS, Ullernchaussen 64, 0379 Oslo, Norway
| | - Vanya Bogoeva
- Department of Molecular Biology and Cell Cycle, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Geir Slupphaug
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
- Proteomics and Modomics Experimental Core, PROMEC, at NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Lars Hagen
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, N-7489 Trondheim, Norway
- Proteomics and Modomics Experimental Core, PROMEC, at NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| |
Collapse
|
2
|
Wong OGW, Li J, Cheung ANY. Targeting DNA Damage Response Pathway in Ovarian Clear Cell Carcinoma. Front Oncol 2021; 11:666815. [PMID: 34737943 PMCID: PMC8560708 DOI: 10.3389/fonc.2021.666815] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is one of the major types of ovarian cancer and is of higher relative prevalence in Asians. It also shows higher possibility of resistance to cisplatin-based chemotherapy leading to poor prognosis. This may be attributed to the relative lack of mutations and aberrations in homologous recombination-associated genes, which are crucial in DNA damage response (DDR), such as BRCA1, BRCA2, p53, RAD51, and genes in the Fanconi anemia pathway. On the other hand, OCCC is characterized by a number of genetic defects rendering it vulnerable to DDR-targeting therapy, which is emerging as a potent treatment strategy for various cancer types. Mutations of ARID1A, PIK3CA, PTEN, and catenin beta 1 (CTNNB1), as well as overexpression of transcription factor hepatocyte nuclear factor-1β (HNF-1β), and microsatellite instability are common in OCCC. Of particular note is the loss-of-function mutations in ARID1A, which is found in approximately 50% of OCCC. ARID1A is crucial for processing of DNA double-strand break (DSB) and for sustaining DNA damage signaling, rendering ARID1A-deficient cells prone to impaired DNA damage checkpoint regulation and hence sensitive to poly ADP ribose polymerase (PARP) inhibitors. However, while preclinical studies have demonstrated the possibility to exploit DDR deficiency in OCCC for therapeutic purpose, progress in clinical application is lagging. In this review, we will recapitulate the preclinical studies supporting the potential of DDR targeting in OCCC treatment, with emphasis on the role of ARID1A in DDR. Companion diagnostic tests (CDx) for predicting susceptibility to PARP inhibitors are rapidly being developed for solid tumors including ovarian cancers and may readily be applicable on OCCC. The potential of various available DDR-targeting drugs for treating OCCC by drawing analogies with other solid tumors sharing similar genetic characteristics with OCCC will also be discussed.
Collapse
|
3
|
Acquired Evolution of Mitochondrial Metabolism Regulated by HNF1B in Ovarian Clear Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13102413. [PMID: 34067626 PMCID: PMC8157013 DOI: 10.3390/cancers13102413] [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: 03/02/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/11/2023] Open
Abstract
Clear cell carcinoma (CCC) of the ovary exhibits a unique morphology and clinically malignant behavior. The eosinophilic cytoplasm includes abundant glycogen. Although the growth is slow, the prognosis is poor owing to resistance to conventional chemotherapies. CCC often arises in endometriotic cysts and is accompanied by endometriosis. Based on these characteristics, three clinical questions are considered: why does ovarian cancer, especially CCC and endometrioid carcinoma, frequently occur in endometriotic cysts, why do distinct histological subtypes (CCC and endometrioid carcinoma) arise in the endometriotic cyst, and why does ovarian CCC possess unique characteristics? Mutations in AT-rich interacting domain-containing protein 1A and phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit alpha genes may contribute to the carcinogenesis of ovarian CCC, whereas hepatocyte nuclear factor-1-beta (HNF1B) plays crucial roles in sculpting the unique characteristics of ovarian CCC through metabolic alterations. HNF1B increases glutathione synthesis, activates anaerobic glycolysis called the Warburg effect, and suppresses mitochondria. These metabolic changes may be induced in stressful environments. Life has evolved to utilize and control energy; eukaryotes require mitochondria to transform oxygen reduction into useful energy. Because mitochondrial function is suppressed in ovarian CCC, these cancer cells probably acquired further metabolic evolution during the carcinogenic process in order to survive stressful environments.
Collapse
|
4
|
Kawahara N, Mizutani A, Matsubara S, Takeda Y, Kobayashi H. GSK-3β mediates the effects of HNF-1β overexpression in ovarian clear cell carcinoma. Exp Ther Med 2020; 20:122. [PMID: 33005248 PMCID: PMC7523276 DOI: 10.3892/etm.2020.9250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Deubiquitinase USP28 is a target gene of the transcription factor HNF1 homeobox β (HNF-1β), which promotes the survival of ovarian clear cell carcinoma (OCCC) cell lines. However, the pharmacological inhibition of HNF-1β can cause several adverse effects as it is abundantly expressed in numerous organ systems, including the kidney, liver, pancreas and digestive tract. Therefore, small interfering RNA (siRNA) screening was performed in the current study to identify other potential downstream targets of the HNF-1β-mediated pathway. The results revealed that glycogen synthase kinase-3β (GSK-3β) may be a potential downstream target affecting cell viability. To further clarify the effects of GSK-3β, two human OCCC cell lines, TOV-21G (HNF-1β overexpressing line) and ES2 (HNF-1β negative) were transfected with siRNA targeting GSK-3β or control vectors. Loss-of-function studies using RNAi-mediated gene silencing indicated that HNF-1β facilitated GSK-3β expression, resulting in the loss of phosphorylated nuclear factor-κB (p-NFκB) and the reduction of TOV-21G cell proliferation. The cell proliferation assay also revealed that GSK-3β inhibitors rescued the effects of HNF-1β silencing on cell viability in a dose-dependent manner. Furthermore, the GSK-3β inhibitor, AR-A014418, effectively inhibited tumor cell proliferation in a xenograft mouse model. In conclusion and to the best of our knowledge, the current study was the first to determine that GSK-3β is a target gene of HNF-1β. In addition, the results of the present study revealed the novel HNF-1β-GSK-3β-p-NFκB pathway, occurring in response to DNA damage. Targeting this pathway may therefore represent a putative, novel, anticancer strategy in patients with OCCC.
Collapse
Affiliation(s)
- Naoki Kawahara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Ayano Mizutani
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Yoshinori Takeda
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
5
|
Takenaka M, Köbel M, Garsed DW, Fereday S, Pandey A, Etemadmoghadam D, Hendley J, Kawabata A, Noguchi D, Yanaihara N, Takahashi H, Kiyokawa T, Ikegami M, Takano H, Isonishi S, Ochiai K, Traficante N, Gadipally S, Semple T, Vassiliadis D, Amarasinghe K, Li J, Mir Arnau G, Okamoto A, Friedlander M, Bowtell DDL. Survival Following Chemotherapy in Ovarian Clear Cell Carcinoma Is Not Associated with Pathological Misclassification of Tumor Histotype. Clin Cancer Res 2019; 25:3962-3973. [PMID: 30967419 DOI: 10.1158/1078-0432.ccr-18-3691] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/24/2019] [Accepted: 04/02/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Although ovarian clear cell carcinomas (OCCC) are commonly resistant to platinum-based chemotherapy, good clinical outcomes are observed in a subset of patients. The explanation for this is unknown but may be due to misclassification of high-grade serous ovarian cancer (HGSOC) as OCCC or mixed histology. EXPERIMENTAL DESIGN To discover potential biomarkers of survival benefit following platinum-based chemotherapy, we ascertained a cohort of 68 Japanese and Australian patients in whom progression-free survival (PFS) and overall survival (OS) could be assessed. We performed IHC reclassification of tumors, and targeted sequencing and immunohistochemistry of known driver genes. Exome sequencing was performed in 10 patients who had either unusually long survival (N = 5) or had a very short time to progression (N = 5). RESULTS The majority of mixed OCCC (N = 6, 85.7%) and a small proportion of pure OCCC (N = 3, 4.9%) were reclassified as likely HGSOC. However, the PFS and OS of patients with misclassified samples were similar to that of patients with pathologically validated OCCC. Absent HNF1B expression was significantly correlated with longer PFS and OS (P = 0.0194 and 0.0395, respectively). Mutations in ARID1A, PIK3CA, PPP2R1A, and TP53 were frequent, but did not explain length of PFS and OS. An exploratory exome analysis of patients with favorable and unfavorable outcomes did not identify novel outcome-associated driver mutations. CONCLUSIONS Survival benefit following chemotherapy in OCCC was not associated with pathological misclassification of tumor histotype. HNF1B loss may help identify the subset of patients with OCCC with a more favorable outcome.
Collapse
Affiliation(s)
- Masataka Takenaka
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Martin Köbel
- Department of Pathology and Laboratory Medicine, Foothill Medical Center, University of Calgary, Calgary, Canada
| | - Dale W Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - Sian Fereday
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ahwan Pandey
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Dariush Etemadmoghadam
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,Department of Pathology, University of Melbourne, Victoria, Australia
| | - Joy Hendley
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ayako Kawabata
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Daito Noguchi
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nozomu Yanaihara
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takako Kiyokawa
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masahiro Ikegami
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirokuni Takano
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Seiji Isonishi
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuhiko Ochiai
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | - Timothy Semple
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | | | - Jason Li
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Michael Friedlander
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia.
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia.,Department of Pathology, University of Melbourne, Victoria, Australia
| | | |
Collapse
|
6
|
Napsin A, Hepatocyte Nuclear Factor-1-Beta (HNF-1β), Estrogen and Progesterone Receptors Expression in Arias-Stella Reaction. Am J Surg Pathol 2019; 43:325-333. [DOI: 10.1097/pas.0000000000001212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Karam SD, Reddy K, Blatchford PJ, Waxweiler T, DeLouize AM, Oweida A, Somerset H, Marshall C, Young C, Davies KD, Kane M, Tan AC, Wang XJ, Jimeno A, Aisner DL, Bowles DW, Raben D. Final Report of a Phase I Trial of Olaparib with Cetuximab and Radiation for Heavy Smoker Patients with Locally Advanced Head and Neck Cancer. Clin Cancer Res 2018; 24:4949-4959. [PMID: 30084837 DOI: 10.1158/1078-0432.ccr-18-0467] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/16/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022]
Abstract
Purpose: Our goal was to evaluate the safety and toxicity of combining a PARP inhibitor, olaparib, with cetuximab and fractionated intensity-modulated radiotherapy for patients with locally advanced head and neck cancer and heavy smoking histories.Patients and Methods: Patients with ≥10 packs/year history of smoking were treated with olaparib at doses ranging from 25-200 mg orally twice daily beginning approximately 10 days prior to initiation of and with concurrent radiation (69.3 Gy in 33 fractions) using a time-to-event continual reassessment method model. Cetuximab was administered starting approximately 5 days prior to radiation per standard of care.Results: A total of 16 patients were entered onto the study, with 15 evaluable for acute toxicity. The most common treatment-related grade 3-4 side effects were radiation dermatitis and mucositis (38% and 69%, respectively). The MTD was determined to be 50 mg orally twice daily, but the recommended phase II dose was deemed to be 25 mg orally twice daily. At a median follow-up of 26 months, the actuarial median overall survival was 37 months, but was not reached for other endpoints. Two-year overall survival, progression-free survival, local control, and distant control rates were 72%, 63%, 72%, and 79%, respectively. Patients who continued to smoke during therapy experienced higher recurrence rates. MYC and KMT2A were identified as potential correlatives of response on gene amplification and mutational analysis.Conclusions: Olaparib at 25 mg orally twice daily with concurrent cetuximab and radiation was well tolerated with reduced dermatitis within the radiation field. Response rates were promising for this high-risk population. Clin Cancer Res; 24(20); 4949-59. ©2018 AACR.
Collapse
Affiliation(s)
- Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Krishna Reddy
- Department of Radiation Oncology, University of Toledo, Toledo, Ohio
| | - Patrick J Blatchford
- Department of Biostatistics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Tim Waxweiler
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Alicia M DeLouize
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Ayman Oweida
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Hilary Somerset
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Carrie Marshall
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Christian Young
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Kurtis D Davies
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Madeleine Kane
- Department of Medicine, Division of Medical Oncology, Anschutz Medical Campus, Aurora, Colorado
| | - Aik Choo Tan
- Department of Medicine, Division of Medical Oncology, Anschutz Medical Campus, Aurora, Colorado
| | - Xiao Jing Wang
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.,Denver Veterans Affairs Medical Center, Eastern Colorado Health Care System, Denver, Colorado
| | - Antonio Jimeno
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Dara L Aisner
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Daniel W Bowles
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.,Denver Veterans Affairs Medical Center, Eastern Colorado Health Care System, Denver, Colorado
| | - David Raben
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
8
|
Kobayashi H, Kawahara N, Ogawa K, Yamada Y, Iwai K, Niiro E, Morioka S. Conceptual frameworks of synthetic lethality in clear cell carcinoma of the ovary. Biomed Rep 2018; 9:112-118. [PMID: 30013776 DOI: 10.3892/br.2018.1114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022] Open
Abstract
Targeting non-oncogenes may result in the selective death of cancer cells. Clear cell carcinoma of the ovary (CCC) may exhibit resistance against conventional chemotherapy and is associated with poor prognosis. The aim of the present report was to review synthetic lethality-based therapies for CCC. Previous English-language studies were reviewed to accumulate preclinical and clinical data on targeting synthetic lethal partners. Synthetic lethal interactions have a variety of types, involving components of a backup or parallel pathway with overlapping functions, components encoded by paralogous pairs, subunit components that form heteromeric complexes and components that are arranged in a single linear pathway. A set of candidate gene targets potentially resulting in synthetic lethality have been previously identified. HNF class homeobox, AT-rich interaction domain 1A, ATR serine/threonine kinase, ATM serine/threonine kinase, checkpoint kinase 1 and phosphatase and tensin homolog may be the key partner genes. A variety of loss of function genes in CCC are driver or passenger events and may function as synthetic lethal pairs under replication stress conditions. Further clinical studies will be required to investigate the safety and therapeutic effect of synthetic lethality pairs in CCC tumor types with replication stress.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Naoki Kawahara
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Kenji Ogawa
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Yuki Yamada
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Kana Iwai
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Emiko Niiro
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Sachiko Morioka
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| |
Collapse
|
9
|
Ito F, Yoshimoto C, Yamada Y, Sudo T, Kobayashi H. The HNF-1β-USP28-Claspin pathway upregulates DNA damage-induced Chk1 activation in ovarian clear cell carcinoma. Oncotarget 2018; 9:17512-17522. [PMID: 29707125 PMCID: PMC5915133 DOI: 10.18632/oncotarget.24776] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/27/2018] [Indexed: 11/25/2022] Open
Abstract
Transcription factor hepatocyte nuclear factor 1-beta (HNF-1β) enhances checkpoint kinase 1 (Chk1) activation and promotes G2/M cell cycle progression in ovarian clear cell carcinoma (CCC) following exposure to diverse genotoxic agents including bleomycin. However, the underlying mechanism leading to checkpoint activation of HNF-1β still remains largely unknown. To clarify the effects of HNF-1β on cell cycle checkpoints, human CCC cell lines were transfected with siRNAs targeting HNF-1β, Claspin, USP28, or a control vector. Ubiquitination and stabilization of Claspin protein by HNF-1β was assessed by immunoprecipitation. Loss-of-function studies using RNAi-mediated gene silencing indicated that HNF-1β facilitated the Claspin expression after treatment with a genotoxic agent bleomycin, resulting in accumulation of phosphorylated Chk1 (p-Chk1) and promotion of survival in CCC cell lines. This study showed for the first time that USP28, a de-ubiquitinase crucial for Claspin expression, is one target gene of HNF-1β. Knockdown of endogenous USP28 suppressed the Claspin expression and p-Chk1 activation and cell viability. Our findings identify a novel pathway of the HNF-1β-USP28-Claspin-Chk1 axis in checkpoint signal amplification in response to DNA damage. Targeting this pathway may represent a putative, novel, anticancer strategy in ovarian CCC.
Collapse
Affiliation(s)
- Fuminori Ito
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Yuki Yamada
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Tamotsu Sudo
- Section of Translational Research, Hyogo Cancer Center, Akashi, Hyogo, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| |
Collapse
|
10
|
Bártů M, Dundr P, Němejcová K, Tichá I, Hojný H, Hájková N. The Role of HNF1B in Tumorigenesis of Solid Tumours: a Review of Current Knowledge. Folia Biol (Praha) 2018; 64:71-83. [PMID: 30394265 DOI: 10.14712/fb2018064030071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Hepatocyte nuclear factor 1-β is a transcription factor which plays a crucial role during ontogenesis in the differentiation of visceral endoderm from primitive endoderm, and is especially important for the normal development of the kidney, urogenital tract, gastrointestinal tract, liver, and pancreas. Despite the growing knowledge about the potential involvement of hepatocyte nuclear factor 1-β in the process of carcinogenesis, the exact underlying mechanism that would explain its rather varied effects in different tumours has not been sufficiently investigated. Most of the data regarding the significance of hepatocyte nuclear factor 1-β arise from genome- wide association studies and is concerned with the influence of single-nucleotide polymorphisms of hepatocyte nuclear factor 1-β on either the increased or decreased susceptibility to certain types of cancer. However, the influence of both the germinal and somatic mutations of this gene on the process of carcinogenesis is still poorly understood. According to current data, in some tumours hepatocyte nuclear factor 1-β acts as a protooncogene, while in others as a tumour suppressor gene, although the reasons for this are not clear. The exact incidence of hepatocyte nuclear factor 1-β mutations and the spectrum of tumours in which they may play a role in the process of carcinogenesis remain unknown. From the practical point of view, immunohistochemical expression of hepatocyte nuclear factor 1-β can be used in differential diagnostics of certain tumours, especially clear cell carcinoma. In our article we review the current knowledge regarding the significance of hepatocyte nuclear factor 1-β in carcinogenesis.
Collapse
Affiliation(s)
- M Bártů
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - P Dundr
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - K Němejcová
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - I Tichá
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - H Hojný
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - N Hájková
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
11
|
Kawahara N, Ogawa K, Nagayasu M, Kimura M, Sasaki Y, Kobayashi H. Candidate synthetic lethality partners to PARP inhibitors in the treatment of ovarian clear cell cancer. Biomed Rep 2017; 7:391-399. [PMID: 29109859 DOI: 10.3892/br.2017.990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023] Open
Abstract
Inhibitors of poly(ADP-ribose) polymerase (PARP) are new types of personalized treatment of relapsed platinum-sensitive ovarian cancer harboring BRCA1/2 mutations. Ovarian clear cell cancer (CCC), a subset of ovarian cancer, often appears as low-stage disease with a higher incidence among Japanese. Advanced CCC is highly aggressive with poor patient outcome. The aim of the present study was to determine the potential synthetic lethality gene pairs for PARP inhibitions in patients with CCC through virtual and biological screenings as well as clinical studies. We conducted a literature review for putative PARP sensitivity genes that are associated with the CCC pathophysiology. Previous studies identified a variety of putative target genes from several pathways associated with DNA damage repair, chromatin remodeling complex, PI3K-AKT-mTOR signaling, Notch signaling, cell cycle checkpoint signaling, BRCA-associated complex and Fanconi's anemia susceptibility genes that could be used as biomarkers or therapeutic targets for PARP inhibition. BRCA1/2, ATM, ATR, BARD1, CCNE1, CHEK1, CKS1B, DNMT1, ERBB2, FGFR2, MRE11A, MYC, NOTCH1 and PTEN were considered as candidate genes for synthetic lethality gene partners for PARP interactions. When considering the biological background underlying PARP inhibition, we hypothesized that PARP inhibitors would be a novel synthetic lethal therapeutic approach for CCC tumors harboring homologous recombination deficiency and activating oncogene mutations. The results showed that the majority of CCC tumors appear to have indicators of DNA repair dysfunction similar to those in BRCA-mutation carriers, suggesting the possible utility of PARP inhibitors in a subset of CCC.
Collapse
Affiliation(s)
- Naoki Kawahara
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Kenji Ogawa
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Mika Nagayasu
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Mai Kimura
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Yoshikazu Sasaki
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| |
Collapse
|
12
|
De Mattia E, Cecchin E, Roncato R, Toffoli G. Pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factors as emerging players in cancer precision medicine. Pharmacogenomics 2016; 17:1547-71. [DOI: 10.2217/pgs-2016-0095] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Great research effort has been focused on elucidating the contribution of host genetic variability on pharmacological outcomes in cancer. Nuclear receptors have emerged as mediators between environmental stimuli and drug pharmacokinetics and pharmacodynamics. The pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factors have been reported to regulate transcription of genes that encode drug metabolizing enzymes and transporters. Altered nuclear receptor expression has been shown to affect the metabolism and pharmacological profile of traditional chemotherapeutics and targeted agents. Accordingly, polymorphic variants in these genes have been studied as pharmacogenetic markers of outcome variability. This review summarizes the state of knowledge about the roles played by pregnane X receptor, constitutive androstane receptor and hepatocyte nuclear factor expression and genetics as predictive markers of anticancer drug toxicity and efficacy, which can improve cancer precision medicine.
Collapse
Affiliation(s)
- Elena De Mattia
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Rossana Roncato
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology, Centro di Riferimento Oncologico- National Cancer Institute, Aviano, Italy
| |
Collapse
|
13
|
Oxidative Stress and Antioxidant Defense in Endometriosis and Its Malignant Transformation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:848595. [PMID: 26185594 PMCID: PMC4491397 DOI: 10.1155/2015/848595] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/03/2015] [Accepted: 06/10/2015] [Indexed: 12/31/2022]
Abstract
The aim of this study was to investigate the role of redox status in endometriosis and its malignant transformation. A search was conducted between 1990 and 2014 through the English language literature (online MEDLINE PubMed database) using the keywords endometriosis combined with malignant transformation, oxidative stress, and antioxidant defense. In benign endometriosis, autoxidation and Fenton reaction of hemoglobin from the ferrous Fe2+ (oxyhemoglobin) state to the ferric Fe3+ (methemoglobin) state lead to production of excess reactive oxygen species (ROS) such as
O2− and ∙OH. Hemoglobin, heme, and iron derivatives in endometriotic cysts cause distortion in the homeostatic redox balance. Excess oxidative stress could trigger DNA damage and cell death. In contrast, endometriosis-associated ovarian cancer (EAOC) might be associated with an effective antioxidant defense, including heme oxygenases, cytochrome P450 family, and glutathione transferase family. The pattern of redox balance supports that enhanced antioxidants may be involved in the pathogenesis of malignant transformation. In conclusion, oxidant/antioxidant balance function is a double-edged sword, promoting cell death or carcinogenesis. Upregulation of antioxidant functions in endometriotic cyst may result in restoration of cell survival and subsequent malignant transformation.
Collapse
|
14
|
Kobayashi H, Sugimoto H, Onishi S, Nakano K. Novel biomarker candidates for the diagnosis of ovarian clear cell carcinoma. Oncol Lett 2015; 10:612-618. [PMID: 26622542 DOI: 10.3892/ol.2015.3367] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 05/27/2015] [Indexed: 02/07/2023] Open
Abstract
Ovarian clear cell carcinoma can arise from endometriosis; however, it is distinct from other types of epithelial ovarian carcinoma in terms of its clinicopathological and molecular features. Cancer antigen 125 lacks the sensitivity and specificity required for accurate clinical diagnosis of clear cell carcinoma. Therefore, the aim of the current review was to identify novel biomarker candidates for the immunohistochemical and serological diagnosis of clear cell carcinoma. A search of the relevant English language literature published between 1966 and 2014 was conducted using the PubMed MEDLINE online database. High-throughput tissue microarray technology and proteomic screening combined with mass spectrometry may provide additional information regarding diagnostic biomarker candidates for ovarian clear cell carcinoma. The present review summarizes the characteristics of potential genomic alterations that activate cancer signaling pathways and, thus, contribute to carcinogenesis. The major signaling pathways activated in clear cell carcinoma are associated with cell cycle regulation (hepatitis A virus cellular receptor 1 and tumor protein D52), growth factor signaling (insulin-like growth factor binding protein 1; KiSS-1 metastasis-suppressor; erb-b2 receptor tyrosine kinase 2; and fibroblast growth factor receptor 2), anti-apoptosis and survival pathways [sialidase 3 (membrane sialidase)], metabolism (γ-glutamyltransferase 1), chemoresistance (napsin A aspartic peptidase, glutathione peroxidase 3; and aldehyde dehydrogenase 1 family, member A1), coagulation [coagulation factor III (thromboplastin, tissue factor); and tissue factor pathway inhibitor 2], signaling (lectin, galactoside-binding and soluble, 3), and adhesion and the extracellular matrix [cadherin 1, type 1, E-cadherin (epithelial); versican; and laminin, α 5]. The present review of the relevant literature may provide a basis for additional clinical investigation of the ovarian clear cell carcinoma serum biomarker candidate proteins identified herein.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hitomi Sugimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Shunsuke Onishi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Kazutoshi Nakano
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
15
|
Kobayashi H, Shigetomi H, Yoshimoto C. Checkpoint kinase 1 inhibitors as targeted molecular agents for clear cell carcinoma of the ovary. Oncol Lett 2015; 10:571-576. [PMID: 26622535 DOI: 10.3892/ol.2015.3268] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 04/29/2015] [Indexed: 12/12/2022] Open
Abstract
In clear cell carcinoma of the ovary, chemoresistance frequently results in treatment failure. The present study aimed to review the potential association of transcription factor hepatocyte nuclear factor (HNF)-1β with cell cycle checkpoint machinery, as a mechanism for chemoresistance. The English-language literature on the subject was reviewed to identify genomic alterations and aberrant molecular pathways interacting with chemoresistance in clear cell carcinoma. Oxidative stress induced by repeated hemorrhage induces greater susceptibility of endometriotic cells to DNA damage, and subsequent malignant transformation results in endometriosis-associated ovarian cancer. Molecular changes, including those in HNF-1β and checkpoint kinase 1 (Chk1), may be a manifestation of essential alterations in cell cycle regulation, detoxification and chemoresistance in clear cell carcinoma. Chk1 is a critical signal transducer in the cell cycle checkpoint machinery. DNA damage, in turn, increases persistent phosphorylation of Chk1 and induction of G2/M phase cell cycle arrest in cells overexpressing HNF-1β. HNF-1β deletion induces apoptosis, suggesting that enhanced levels of HNF-1β may be associated with chemoresistance. Targeted therapy with Chk1 inhibitors may be explored as a potential treatment modality for patients with clear cell carcinoma. This provides a novel direction for combination therapy, including targeting of Chk1, which may overcome drug resistance and improve treatment efficacy.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
16
|
Mandato VD, Farnetti E, Torricelli F, Abrate M, Casali B, Ciarlini G, Pirillo D, Gelli MC, Nicoli D, Grassi M, LA Sala GB, Palomba S. HNF1B polymorphism influences the prognosis of endometrial cancer patients: a cohort study. BMC Cancer 2015; 15:229. [PMID: 25885815 PMCID: PMC4403886 DOI: 10.1186/s12885-015-1246-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 03/23/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND HNF1B (formerly known as TCF2) gene encodes for a transcription factor that regulates gene expression involved in normal mesodermal and endodermal developments. A close association between rs4430796 polymorphism of HNF1B gene and decreased endometrial cancer (EC) risk has been demonstrated. The aim of the current study was to test the hypothesis that rs4430796 polymorphism can influence the prognosis of EC patients. METHODS Retrospective cohort study. Clinical and pathological data were extrapolated and genotypes were assessed on formalin-fixed and paraffin-embedded non-tumour tissues. The influence of patients' genotype on overall survival and progression free survival were our main outcome measures. RESULTS A total of 191 EC patients were included in the final analysis. Overall survival differed significantly (P = 0.003) among genotypes. At multivariate analysis, a significant (P < 0.05) effect on overall survival was detected for FIGO stage, and rs4430796 polymorphism of HNF1B gene. After grouping EC patients according to adjuvant treatment, rs4430796 polymorphism resulted significantly (P < 0.001) related to overall survival only in subjects who received radiotherapy plus chemotherapy. A significant (P = 0.014) interaction between rs4430796 polymorphism and chemo-radiotherapy was also detected. Finally, only a trend (P = 0.090) towards significance was observed for rs4430796 polymorphism effect on progression free survival. CONCLUSIONS rs4430796 polymorphism of HNF1B gene influences independently the prognosis of EC patients with a potential effect on tumor chemo-sensitivity.
Collapse
Affiliation(s)
- Vincenzo Dario Mandato
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Enrico Farnetti
- Laboratory of Molecular Biology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Federica Torricelli
- Laboratory of Molecular Biology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Martino Abrate
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Bruno Casali
- Laboratory of Molecular Biology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Gino Ciarlini
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Debora Pirillo
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | | | - Davide Nicoli
- Laboratory of Molecular Biology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
| | - Mario Grassi
- Department of Brain and Behavioral Science, Medical and Genomics Statistics Unit, University of Pavia, Pavia, Italy.
| | - Giovanni Battista LA Sala
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy.
- University of Modena and Reggio Emilia, Modena, Italy.
| | - Stefano Palomba
- Unit of Gynecologic Oncology, IRCCS-CROB, Rionero in Vulture (Potenza), Potenza, Italy.
| |
Collapse
|