1
|
Spasic J, Cavic M, Stanic N, Zaric B, Kovacevic T, Radosavljevic D, Nagorni-Obradovic L. Low-Cost Genetic and Clinical Predictors of Response and Toxicity of Platinum-Based Chemotherapy in Advanced Non-Small Cell Lung Cancer. Dose Response 2022; 20:15593258221111666. [PMID: 35783235 PMCID: PMC9247378 DOI: 10.1177/15593258221111666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Background This study aimed to evaluate for the first time whether certain genetic and
clinical factors could serve as minimally invasive predictors of survival
and toxicity to platinum-based chemotherapy in advanced lung
adenocarcinoma. Methods The study included 121 advanced lung adenocarcinoma patients treated with
platinum-based dublets until progression or unacceptable toxicity. Response
was evaluated using standard radiological methods and toxicity graded
according to the Common Terminology Criteria for Adverse Events (CTCAE)
v5.0. Genotyping was performed using PCR-RFLP. Statistical significance was
set at P < .05. Results No significant influence of the examined polymorphisms on the occurrence of
high-grade toxicity was detected. However, TP53 72Pro allele carriers were
more prone to nausea (P = .037) and thrombocytopenia (P = .051). Anemia and
neuropathy occurred more frequently in XRCC1 399Arg allele carriers (Pearson
χ2 test, P = .025 and P = .004 respectively). RAD51 135CC carriers were
significantly more prone to neutropenia (P = .027). Conclusions A set of easily determined genetic and clinical predictors of survival and
specific toxicity profiles of platinum-based chemotherapy in advanced lung
adenocarcinoma were determined in this study, which might be useful for the
construction of population-specific, time- and cost-efficient prognostic and
predictive algorithms.
Collapse
Affiliation(s)
- Jelena Spasic
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Serbia
| | - Milena Cavic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Serbia
| | - Nemanja Stanic
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Serbia
| | - Bojan Zaric
- Faculty of Medicine, University of Novi Sad, Serbia.,Institute for Pulmonary Diseases of Vojvodina, Serbia
| | - Tomi Kovacevic
- Faculty of Medicine, University of Novi Sad, Serbia.,Institute for Pulmonary Diseases of Vojvodina, Serbia
| | | | - Ljudmila Nagorni-Obradovic
- Faculty of Medicine, University of Belgrade, Serbia.,Clinic for Pulmonology, University Clinical Centre of Serbia, Serbia
| |
Collapse
|
2
|
Madhavan BK, Han Z, Singh B, Bordt N, Kaymak S, Bandapalli OR, Kihm L, Shahzad K, Isermann B, Herzig S, Nawroth P, Kumar V. Elevated Expression of the RAGE Variant- V in SCLC Mitigates the Effect of Chemotherapeutic Drugs. Cancers (Basel) 2021; 13:cancers13112843. [PMID: 34200336 PMCID: PMC8201239 DOI: 10.3390/cancers13112843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiomimetic drugs induce extensive genotoxic insults to their target cells. Irreparable DNA damage leaves cells with the choice between a program leading to cell death or senescence, but not DNA repair. Among the challenges of an advanced stage of small cell lung carcinoma (SCLC), the resistance to radiomimetic drugs is the most prominent one. In SCLC, the initial chemotherapeutic treatment primes cell to modify their DNA repair and cell cycle regulatory systems, using alternative but highly efficient forms of DNA repair and auxiliary factors. This modulated system now bypasses several regulatory controls. Thus, at this stage, cells become resistant to any beneficial effects of chemotherapeutic drugs. In the present study, we observed that variant-V of the receptor for advanced glycation end-products (RAGE) is abundantly expressed in advancing and metastasizing SCLC. Therefore, it may serve as a potential target for specific therapeutic interventions directed to SCLC. Abstract Small cell lung carcinoma (SCLC) is a highly aggressive malignancy with a very high mortality rate. A prominent part of this is because these carcinomas are refractory to chemotherapies, such as etoposide or cisplatin, making effective treatment almost impossible. Here, we report that elevated expression of the RAGE variant-V in SCLC promotes homology-directed DNA DSBs repair when challenged with anti-cancer drugs. This variant exclusively localizes to the nucleus, interacts with members of the double-strand break (DSB) repair machinery and thus promotes the recruitment of DSBs repair factors at the site of damage. Increased expression of this variant thus, promotes timely DNA repair. Congruently, the tumor cells expressing high levels of variant-V can tolerate chemotherapeutic drug treatment better than the RAGE depleted cells. Our findings reveal a yet undisclosed role of the RAGE variant-V in the homology-directed DNA repair. This variant thus can be a potential target to be considered for future therapeutic approaches in advanced SSLC.
Collapse
Affiliation(s)
- Bindhu K. Madhavan
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Zhe Han
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Bishal Singh
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Nico Bordt
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Serap Kaymak
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Obul Reddy Bandapalli
- Hopp Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany;
- Medical Faculty, Heidelberg University, 69117 Heidelberg, Germany
| | - Lars Kihm
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
| | - Khurrum Shahzad
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (K.S.); (B.I.)
| | - Berend Isermann
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (K.S.); (B.I.)
| | - Stephan Herzig
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Molecular Metabolic Control, Technical University Munich, 80333 Munich, Germany
- Helmholtz Center Munich, Institute for Diabetes and Cancer, D-85764 Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Programm, Helmholtz-Zentrum, 69120 Heidelberg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- Joint Heidelberg-IDC Translational Diabetes Programm, Helmholtz-Zentrum, 69120 Heidelberg, Germany
| | - Varun Kumar
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, INF 410, 69120 Heidelberg, Germany; (B.K.M.); (Z.H.); (B.S.); (N.B.); (S.K.); (L.K.); (P.N.)
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany;
- European Molecular Biology Laboratory, Advanced Light Microscopy Facility, 69117 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-56-6960
| |
Collapse
|
3
|
Gagno S, Bartoletti M, Romualdi C, Poletto E, Scalone S, Sorio R, Zanchetta M, De Mattia E, Roncato R, Cecchin E, Giorda G, Toffoli G. Pharmacogenetic score predicts overall survival, progression-free survival and platinum sensitivity in ovarian cancer. Pharmacogenomics 2020; 21:995-1010. [PMID: 32894980 DOI: 10.2217/pgs-2020-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To define the impact of polymorphisms in genes involved in platinum-taxane and estrogen activity in the outcome of platinum-based treated ovarian cancer patients (OCP). Patients & Methods: Two hundred and thirty OCP were analyzed for 124 germ-line polymorphisms to generate a prognostic score for overall survival (OS), progression-free survival (PFS) and platinum-free interval (PFI). Results: ABCG2 rs3219191D>I, UGT1A rs10929302G>A and UGT1A rs2741045T>C polymorphisms were significantly associated with all three parameters (OS, PFS and PFI) and were used to generate a score. Patients in high-risk group had a poorer OS (hazard ratio [HR]: 1.8; 95% CI: 1.3-2.7; p = 0.0019), PFS (HR: 2.0; 95% CI: 1.4-2.9; p < 0.0001) and PFI (HR: 1.9; 95% CI: 1.4-2.8; p = 0.0002) compared with those in low-risk group. Conclusion: The prognostic-score including polymorphisms involved in drug and estrogen pathways stratifies OCP according to OS, PFS and PFI.
Collapse
Affiliation(s)
- Sara Gagno
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Michele Bartoletti
- Department of Medicine (DAME), University of Udine, Via Palladio 8, 33100, Udine, Italy.,Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Chiara Romualdi
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35122, Padova, Italy
| | - Elena Poletto
- Department of Oncology, ASUI Udine University Hospital, Via Pozzuolo 330, 33100, Udine, Italy
| | - Simona Scalone
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Roberto Sorio
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Martina Zanchetta
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Elena De Mattia
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Rossana Roncato
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Erika Cecchin
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Giorgio Giorda
- Gynaecological Oncology Unit, Centro di Riferimento Oncologico (CRO) di Aviano, IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental & Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Via Franco Gallini 2, 33081, Aviano, Italy
| |
Collapse
|
4
|
Cavic M, Spasic J, Krivokuca A, Boljevic I, Kuburovic M, Radosavljevic D, Jankovic R. TP53 and DNA-repair gene polymorphisms genotyping as a low-cost lung adenocarcinoma screening tool. J Clin Pathol 2018; 72:75-80. [PMID: 30467244 DOI: 10.1136/jclinpath-2018-205553] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 12/16/2022]
Abstract
AIM TP53 and DNA repair polymorphisms have been proposed as cancer risk factors. This study evaluated the usability of TP53 Arg72Pro single-nucleotide polymorphism, X RCC1 Arg399Gln and RAD51 G135C as a low-cost lung adenocarcinoma screening tool. PATIENTS AND METHODS This case-control study included 78 atients with lung adenocarcinoma and 79 healthy matched controls. TP53, XRCC1 and RAD51 genotyping was done by PCR followed by restriction length polymorphism. Descriptive analyses included genotype and allelic frequencies and deviations of the frequencies from those expected under Hardy-Weinberg equilibrium were assessed using the χ2 test. The OR and 95% CIs were calculated as an estimate of relative risk, with significance set at p value <0.05. RESULTS The TP53 codon 72 Pro allele and the XRCC1 codon 399 Arg allele in a homozygous state were associated with lung adenocarcinoma (p=0.037; OR (95% CI) 2.42 (1.10 to 5.31)), that is, p=0.037; OR (95% CI) 2.16 (1.08 to 4.33), respectively. Also, carriers of the TP53 codon 72 Pro allele and the XRCC1 codon 399 ArgArg genotype older than 50 showed an even higher risk of developing lung adenocarcinoma (p=0.03 in both cases). CONCLUSIONS The TP53 codon 72 Arg allele and XRCC1 codon 399 Gln allele are likely to have a protective effect against lung adenocarcinoma, especially in individuals older than 50 years of age. XRCC1 and TP53 genotyping might be a useful low-cost tool for evaluating individual lung cancer risk, leading to earlier detection and management of this disease.
Collapse
Affiliation(s)
- Milena Cavic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jelena Spasic
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ana Krivokuca
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Ivana Boljevic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Mira Kuburovic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Davorin Radosavljevic
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Radmila Jankovic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| |
Collapse
|
5
|
Yang Q, Wu J, Luo Y, Huang N, Zhen N, Zhou Y, Sun F, Li Z, Pan Q, Li Y. (-)-Guaiol regulates RAD51 stability via autophagy to induce cell apoptosis in non-small cell lung cancer. Oncotarget 2018; 7:62585-62597. [PMID: 27566579 PMCID: PMC5308748 DOI: 10.18632/oncotarget.11540] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/11/2016] [Indexed: 12/21/2022] Open
Abstract
(-)-Guaiol, generally known as an antibacterial compound, has been found in many medicinal plants. Its roles in tumor suppression are still under investigation. In the study, we mainly focused on exploring its applications in dealing with non-small cell lung cancer (NSCLC) and the underlying mechanisms. Here, we show that (-)-Guaiol significantly inhibits cell growth of NSCLC cells both in vitro and in vivo. Further high throughput analysis reveals that RAD51, a pivotal factor in homologous recombination repair, is a potential target for it. The following mechanism studies show that (-)-Guaiol is involved in cell autophagy to regulate the expression of RAD51, leading to double-strand breaks triggered cell apoptosis. Moreover, targeting RAD51, which is highly overexpressed in the lung adenocarcinoma tissues, can significantly increase the chemosensitivity of NSCLC cells to (-)-Guaiol both in vitro and in vivo. All in all, our studies provide an attractive insight in applying (-)-Guaiol into NSCLC treatments and further suggest that knockdown of oncogenic RAD51 will greatly enhance the chemosensitivity of patients with NSCLC.
Collapse
Affiliation(s)
- Qingyuan Yang
- Department of Clinical Laboratory Medicine, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Jianchun Wu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yingbin Luo
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Nan Huang
- Department of Clinical Laboratory Medicine, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Ni Zhen
- Department of Clinical Laboratory Medicine, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Yun Zhou
- Department of Clinical Laboratory Medicine, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Zhi Li
- Department of Clinical Laboratory Medicine, Yangpu Hospital of Tongji University, Shanghai, 200090, China
| | - Qiuhui Pan
- Central Laboratory, Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Yan Li
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| |
Collapse
|
6
|
Assis J, Pereira C, Nogueira A, Pereira D, Carreira R, Medeiros R. Genetic variants as ovarian cancer first-line treatment hallmarks: A systematic review and meta-analysis. Cancer Treat Rev 2017; 61:35-52. [PMID: 29100168 DOI: 10.1016/j.ctrv.2017.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND The potential predictive value of genetic polymorphisms in ovarian cancer first-line treatment is inconsistently reported. We aimed to review ovarian cancer pharmacogenetic studies to update and summarize the available data and to provide directions for further research. METHODS A systematic review followed by a meta-analysis was conducted on cohort studies assessing the involvement of genetic polymorphisms in ovarian cancer first-line treatment response retrieved through a MEDLINE database search by November 2016. Studies were pooled and summary estimates and 95% confidence intervals (CI) were calculated using random or fixed-effects models as appropriate. RESULTS One hundred and forty-two studies gathering 106871 patients were included. Combined data suggested that GSTM1-null genotype patients have a lower risk of death compared to GSTM1-wt carriers, specifically in advanced stages (hazard ratio (HR), 0.68; 95% CI, 0.48-0.97) and when submitted to platinum-based chemotherapy (aHR, 0.61; 95% CI, 0.39-0.94). ERCC1 rs11615 and rs3212886 might have also a significant impact in treatment outcome (aHR, 0.67; 95% CI, 0.51-0.89; aHR, 1.28; 95% CI, 1.01-1.63, respectively). Moreover, ERCC2 rs13181 and rs1799793 showed a distinct ethnic behavior (Asians: aHR, 1.41; 95% CI, 0.80-2.49; aHR, 1.07; 95% CI, 0.62-1.86; Caucasians: aHR, 0.10; 95% CI, 0.01-0.96; aHR, 0.18; 95% CI, 0.05-0.68, respectively). CONCLUSION(S) The definition of integrative predictive models should encompass genetic information, especially regarding GSTM1 homozygous deletion. Justifying additional pharmacogenetic investigation are variants in ERCC1 and ERCC2, which highlight the DNA Repair ability to ovarian cancer prognosis. Further knowledge could aid to understand platinum-treatment failure and to tailor chemotherapy strategies.
Collapse
Affiliation(s)
- Joana Assis
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal; FMUP, Faculty of Medicine of Porto University, Porto, Portugal
| | - Carina Pereira
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal; CINTESIS, Center for Health Technology and Services Research, FMUP, Porto, Portugal
| | - Augusto Nogueira
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal; FMUP, Faculty of Medicine of Porto University, Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology, Porto, Portugal
| | - Rafael Carreira
- Centre of Biological Engineering, University of Minho, Braga, Portugal; SilicoLife, Lda, Braga, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal; Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal; CEBIMED, Faculty of Health Sciences of Fernando Pessoa University, Porto, Portugal.
| |
Collapse
|
7
|
Jane EP, Premkumar DR, Sutera PA, Cavaleri JM, Pollack IF. Survivin inhibitor YM155 induces mitochondrial dysfunction, autophagy, DNA damage and apoptosis in Bcl-xL silenced glioma cell lines. Mol Carcinog 2017; 56:1251-1265. [PMID: 27805285 PMCID: PMC6844150 DOI: 10.1002/mc.22587] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 10/20/2016] [Accepted: 10/28/2016] [Indexed: 01/07/2023]
Abstract
Because the anti-apoptotic protein Bcl-xL is overexpressed in glioma, one might expect that inhibiting or silencing this gene would promote tumor cell killing. However, our studies have shown that this approach has limited independent activity, but may tip the balance in favor of apoptosis induction in response to other therapeutic interventions. To address this issue, we performed a pharmacological screen using a panel of signaling inhibitors and chemotherapeutic agents in Bcl-xL silenced cells. Although limited apoptosis induction was observed with a series of inhibitors for receptor tyrosine kinases, PKC inhibitors, Src family members, JAK/STAT, histone deacetylase, the PI3K/Akt/mTOR pathway, MAP kinase, CDK, heat shock proteins, proteasomal processing, and various conventional chemotherapeutic agents, we observed a dramatic potentiation of apoptosis in Bcl-xL silenced cells with the survivin inhibitor, YM155. Treatment with YM155 increased the release of cytochrome c, smac/DIABLO and apoptosis inducing-factor, and promoted loss of mitochondrial membrane potential, activation of Bax, recruitment of LC3-II to the autophagosomes and apoptosis in Bcl-xL silenced cells. We also found an additional mechanism for the augmentation of apoptosis due to abrogation of DNA double-strand break repair mediated by Rad51 repression and enhanced accumulation of γH2AX. In summary, our observations may provide a new insight into the link between Bcl-xL and survivin inhibition for the development of novel therapies for glioma. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Esther P. Jane
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel R. Premkumar
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania,Correspondence to: Department of Neurosurgery, Children’s Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224
| | - Philip A. Sutera
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Ian F. Pollack
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania,Correspondence to: Department of Neurosurgery, Children’s Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224
| |
Collapse
|
8
|
An Z, Yu JR, Park WY. Rosiglitazone enhances radiosensitivity by inhibiting repair of DNA damage in cervical cancer cells. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2017; 56:89-98. [PMID: 28184999 DOI: 10.1007/s00411-016-0679-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 12/27/2016] [Indexed: 06/06/2023]
Abstract
Radiation therapy (RT) is one of the main treatment modalities for cervical cancer. Rosiglitazone (ROSI) has been reported to have antiproliferative effects against various types of cancer cells and also to induce antioxidant enzymes that can scavenge reactive oxygen species (ROS) and thereby modify radiosensitivity. Here, we explored the effect of ROSI on radiosensitivity and the underlying mechanisms in cervical cancer cells. Three cervical cancer cell lines (ME-180, HeLa, and SiHa) were used. The cells were pretreated with ROSI and then irradiated. Expression of proteins of interest was detected by western blot and immunofluorescence. Intracellular production of ROS was measured by H2DCFDA. Radiosensitivity was assessed by monitoring clonogenic survival. Expression of antioxidant enzymes (catalase, superoxide dismutases) was increased by ROSI in HeLa and SiHa cells, but not in ME-180 cells. With ROSI pre-treatment, cell survival after irradiation remained unchanged in HeLa and SiHa cells, but decreased in ME-180 cells. Radiation-induced expression of γ-H2AX was increased and that of RAD51 was decreased by ROSI pre-treatment in ME-180 cells, but not in HeLa cells. ROSI increases radiosensitivity by inhibiting RAD51-mediated repair of DNA damage in some cervical cancer cell lines; therefore, ROSI is a potential inhibitor of RAD51 that can be used to enhance the effect of RT in the treatment of some cervical cancers.
Collapse
Affiliation(s)
- Zhengzhe An
- Department of Environmental and Tropical Medicine, Konkuk University College of Medicine, Chungju, 380-701, Republic of Korea
| | - Jae-Ran Yu
- Department of Environmental and Tropical Medicine, Konkuk University College of Medicine, Chungju, 380-701, Republic of Korea
| | - Woo-Yoon Park
- Department of Radiation Oncology, Chungbuk National University College of Medicine, 52 Chungdae-ro 1, Seowon-gu, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
9
|
An Z, Yu JR, Park WY. T0070907 inhibits repair of radiation-induced DNA damage by targeting RAD51. Toxicol In Vitro 2016; 37:1-8. [DOI: 10.1016/j.tiv.2016.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/06/2016] [Accepted: 08/16/2016] [Indexed: 12/13/2022]
|
10
|
Mullane SA, Werner L, Guancial EA, Lis RT, Stack EC, Loda M, Kantoff PW, Choueiri TK, Rosenberg J, Bellmunt J. Expression Levels of DNA Damage Repair Proteins Are Associated With Overall Survival in Platinum-Treated Advanced Urothelial Carcinoma. Clin Genitourin Cancer 2016; 14:352-9. [PMID: 26778300 PMCID: PMC5508512 DOI: 10.1016/j.clgc.2015.12.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/17/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Combination platinum chemotherapy is standard first-line therapy for metastatic urothelial carcinoma (mUC). Defining the platinum response biomarkers for patients with mUC could establish personalize medicine and provide insights into mUC biology. Although DNA repair mechanisms have been hypothesized to mediate the platinum response, we sought to analyze whether increased expression of DNA damage genes would correlate with worse overall survival (OS) in patients with mUC. PATIENTS AND METHODS We retrospectively identified a clinically annotated cohort of patients with mUC, who had been treated with first-line platinum combination chemotherapy. A tissue microarray was constructed from formalin-fixed paraffin-embedded tissue from the primary tumor before treatment. Immunohistochemical analysis of the following DNA repair proteins was performed: ERCC1, RAD51, BRCA1/2, PAR, and PARP-1. Nuclear and cytoplasmic expression was analyzed using multispectral imaging. Nuclear staining was used for the survival analysis. Cox regression analysis was used to evaluate the associations between the percentage of positive nuclear staining and OS in multivariable analysis, controlling for known prognostic variables. RESULTS In a cohort of 104 patients with mUC, a greater percentage of nuclear staining of ERCC1 (hazard ratio [HR], 2.7; 95% confidence interval [CI], 1.5-4.9; P = .0007), RAD51 (HR, 5.6; 95% CI, 1.7-18.3; P = .005), and PAR (HR, 2.2; 95% CI, 1.1-4.4; P = .026) was associated with worse OS. BRCA1, BRCA2, and PARP-1 expression was not associated with OS (P = .76, P = .38, and P = .09, respectively). A greater percentage of combined ERCC1 and RAD51 nuclear staining was strongly associated with worse OS (P = .005). CONCLUSION A high percentage of nuclear staining of ERCC1, RAD51, and PAR, assessed by immunohistochemistry, correlated with worse OS for patients with mUC treated with first-line platinum combination chemotherapy, supporting the evidence of the DNA repair pathways' role in the prognosis of mUC. We also report new evidence that RAD51 and PAR might play a role in the platinum response. Additional prospective studies are required to determine the prognostic or predictive nature of these biomarkers in mUC.
Collapse
Affiliation(s)
- Stephanie A Mullane
- Bladder Cancer Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA
| | - Lillian Werner
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Elizabeth A Guancial
- Department of Medicine, Wilmot Cancer Institute, University of Rochester, Rochester, NY
| | - Rosina T Lis
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Edward C Stack
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Massimo Loda
- Department of Pathology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Philip W Kantoff
- Bladder Cancer Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Toni K Choueiri
- Bladder Cancer Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | | | - Joaquim Bellmunt
- Bladder Cancer Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| |
Collapse
|
11
|
Nogueira A, Assis J, Faustino I, Pereira D, Catarino R, Medeiros R. Base excision repair pathway: PARP1 genotypes as modulators of therapy response in cervical cancer patients. Biomarkers 2016; 22:70-76. [PMID: 27323894 DOI: 10.1080/1354750x.2016.1204006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
CONTEXT Genetic polymorphisms in genes of the base excision repair (BER) pathway appear to modulate the therapy response of cancer patients. PARP1 protein recognizes the DNA strand damage and facilitates the subsequent recruitment of BER proteins. Few studies have reported an association between PARP1 Val762Ala polymorphism (rs1136410) and cancer therapy response. OBJECTIVE The purpose of our study was to determine whether PARP1 Val762Ala polymorphism have prognostic value in patients with cervical cancer. MATERIALS AND METHODS Two hundred and sixty adult patients, with histologically confirmed cervical cancer, at FIGO-stages IB2-IVA, primarily treated with concurrent chemotherapy (cisplatin) and radiotherapy. Overall survival (OS) and disease-free survival (DFS) were the primary end points of the analysis. The PARP1 Val762Ala genetic variants were analyzed by allelic discrimination by real-time PCR. RESULTS We observed that peri- and postmenopausal women carrying the C-allele present a statistically significant lower OS and DFS (log-rank test, p = 0.008 and p = 0.006, respectively) among those with early stage cervical cancer. Cox regression analysis confirmed these results, after adjustment for other prognostic factors (for OS: HR, 3.70; 95%CI, 1.32-10.38; p = 0.013 and for DFS: HR, 3.97; 95%CI, 1.59-9.93; p = 0.003). CONCLUSIONS This is the first study evaluating the effect of PARP1 Val762Ala polymorphism in treatment response in cervical cancer patients. PARP1 genotypes may contribute as an independent prognostic factor in cervical cancer, being useful in predicting the clinical outcome.
Collapse
Affiliation(s)
- Augusto Nogueira
- a Molecular Oncology and Viral Pathology Group - Research Center , Portuguese Institute of Oncology , Porto , Portugal.,b FMUP, Faculty of Medicine of Porto University , Porto , Portugal.,c Research Department , Portuguese League against Cancer (NRNorte) , Porto , Portugal
| | - Joana Assis
- a Molecular Oncology and Viral Pathology Group - Research Center , Portuguese Institute of Oncology , Porto , Portugal.,b FMUP, Faculty of Medicine of Porto University , Porto , Portugal
| | - Ilda Faustino
- d Oncology Department , Portuguese Institute of Oncology , Porto , Portugal
| | - Deolinda Pereira
- a Molecular Oncology and Viral Pathology Group - Research Center , Portuguese Institute of Oncology , Porto , Portugal.,d Oncology Department , Portuguese Institute of Oncology , Porto , Portugal.,e ICBAS , Abel Salazar Institute for the Biomedical Sciences , Porto , Portugal
| | - Raquel Catarino
- a Molecular Oncology and Viral Pathology Group - Research Center , Portuguese Institute of Oncology , Porto , Portugal
| | - Rui Medeiros
- a Molecular Oncology and Viral Pathology Group - Research Center , Portuguese Institute of Oncology , Porto , Portugal.,c Research Department , Portuguese League against Cancer (NRNorte) , Porto , Portugal.,e ICBAS , Abel Salazar Institute for the Biomedical Sciences , Porto , Portugal.,f CEBIMED , Faculty of Health Sciences of Fernando Pessoa University , Porto , Portugal
| |
Collapse
|
12
|
Palmieri D, Scarpa M, Tessari A, Uka R, Amari F, Lee C, Richmond T, Foray C, Sheetz T, Braddom A, Burd CE, Parvin JD, Ludwig T, Croce CM, Coppola V. Ran Binding Protein 9 (RanBP9) is a novel mediator of cellular DNA damage response in lung cancer cells. Oncotarget 2016; 7:18371-83. [PMID: 26943034 PMCID: PMC4951294 DOI: 10.18632/oncotarget.7813] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/29/2016] [Indexed: 01/27/2023] Open
Abstract
Ran Binding Protein 9 (RanBP9, also known as RanBPM) is an evolutionary conserved scaffold protein present both in the nucleus and the cytoplasm of cells whose biological functions remain elusive. We show that active ATM phosphorylates RanBP9 on at least two different residues (S181 and S603). In response to IR, RanBP9 rapidly accumulates into the nucleus of lung cancer cells, but this nuclear accumulation is prevented by ATM inhibition. RanBP9 stable silencing in three different lung cancer cell lines significantly affects the DNA Damage Response (DDR), resulting in delayed activation of key components of the cellular response to IR such as ATM itself, Chk2, γH2AX, and p53. Accordingly, abrogation of RanBP9 expression reduces homologous recombination-dependent DNA repair efficiency, causing an abnormal activation of IR-induced senescence and apoptosis. In summary, here we report that RanBP9 is a novel mediator of the cellular DDR, whose accumulation into the nucleus upon IR is dependent on ATM kinase activity. RanBP9 absence hampers the molecular mechanisms leading to efficient repair of damaged DNA, resulting in enhanced sensitivity to genotoxic stress. These findings suggest that targeting RanBP9 might enhance lung cancer cell sensitivity to genotoxic anti-neoplastic treatment.
Collapse
Affiliation(s)
- Dario Palmieri
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Mario Scarpa
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Anna Tessari
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
| | - Rexhep Uka
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Foued Amari
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Cindy Lee
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Timothy Richmond
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
| | - Claudia Foray
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Tyler Sheetz
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
| | - Ashley Braddom
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
| | - Christin E. Burd
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Jeffrey D. Parvin
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Thomas Ludwig
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
| | - Vincenzo Coppola
- Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, 43210 Columbus, OH, USA
- Solid Tumor Biology Program, Comprehensive Cancer Center, The Ohio State University, 43210 Columbus, OH, USA
| |
Collapse
|
13
|
Phase I study of LY2603618, a CHK1 inhibitor, in combination with gemcitabine in Japanese patients with solid tumors. Anticancer Drugs 2015; 26:1043-53. [PMID: 26288133 DOI: 10.1097/cad.0000000000000278] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This phase I trial evaluated LY2603618, a selective inhibitor of the DNA damage checkpoint kinase 1, in combination with gemcitabine. Japanese patients with advanced solid tumors were enrolled. All patients received gemcitabine (1000 mg/m on days 1, 8, and 15 every 28 days) and either 170 mg (cohort 1) or 230 mg (cohort 2) of LY2603618. The primary objective was assessment of safety/tolerability. Pharmacokinetic/pharmacodynamic marker profiles were secondary objectives. Of the 17 patients enrolled, dose-limiting toxicities were observed in one patient in cohort 1 (n=7) and in two patients in cohort 2 (n=10). The most common grade 3 or more drug-related treatment-emergent adverse events were hematological. Three patients discontinued because of adverse events. Dose-dependent decreases in LY2603618 exposure were observed, but the LY2603618 pharmacokinetics at each dose were consistent within and between cycles and did not influence gemcitabine pharmacokinetics. Circulating plasma DNA decreased from baseline in all four patients who achieved a partial response. Administration of 170 or 230 mg of LY2603618 following a standard dose of gemcitabine showed acceptable safety and tolerability in Japanese patients with solid tumors.
Collapse
|
14
|
Acquisition of radioresistance in docetaxel-resistant human lung adenocarcinoma cells is linked with dysregulation of miR-451/c-Myc-survivin/rad-51 signaling. Oncotarget 2015; 5:6113-29. [PMID: 25026294 PMCID: PMC4171617 DOI: 10.18632/oncotarget.2176] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chemoresistant tumors usually fail to respond to radiotherapy. However, the mechanisms involved in chemo- and radiotherapy cross resistance are not fully understood. Previously, we have identified microRNA (miR)-451 as a tumor suppressor in lung adenocarcinoma (LAD). However, whether miR-451 plays critical roles in chemo- and radiotherapy cross resistance in LAD is unclear. Here, we established two docetaxel-resistant LAD cell models (SPC-A1/DTX and H1299/DTX), and showed that miR-451 was significantly downregulated in docetaxel-resistant LAD cells. Gain - and loss - of - function assays indicated that re-expression of miR-451 could reverse radioresistance of docetaxel-resistant LAD cells both in vitro and in vivo through promoting apoptosis and DNA double-strand breaks (DSBs). The proto-oncogene c-Myc was identified as a direct target of miR-451, and re-expression of miR-451 inhibited survivin and rad-51 expression by reducing the amount of c-Myc protein binding to their promoters. Silencing of c-Myc could phenocopy the effects of miR-451 upregulation, and restoration of c-Myc could partially rescue the effect of miR-451 upregulation on radiosensitivity of docetaxel-resistant LAD cells. Therefore, dysregulation of miR-451/c-Myc-survivin/rad-51 signaling is responsible for radioresistance of docetaxel-resistant LAD cells, and targeting it will be a potential strategy for reversing chemo- and radiotherapy cross resistance of LAD patients.
Collapse
|
15
|
Abstract
‘Pharmacogenetics or Pharmacogenomics’ (PG) is one of the most practiced cancer therapeutic strategies, tailored for individualized patients. Despite its popularity and rapid advancements in the field, many obstacles for cancer therapy PG still need to be overcome. By borrowing scientific systems from other disciplines such as cancer diagnosis, and therapeutic information from the diversity of tumor origins, categories and stages, cancer therapy PG may hopefully be improved. Furthermore, to quickly acquire genetic and pathologic information and seek therapeutic interventions, possible breakthroughs may come from beyond – changing the cancer therapeutic landscapes. The next generations of PG protocols and hospital routines for searching deadly cancer pathogenic pathways versus drug-targeting predictions are of great clinical significance for the future. Yet, progress of cancer therapy PG is entering into a bottleneck stage owing to simple model of relevant techniques and routines. Promoting or even innovating present PG modular is very necessary. This perspective highlights this issue by introducing new initiatives and ideas.
Collapse
|
16
|
Rosinha A, Assis J, Dias F, Nogueira A, Pereira D, Maurício J, Teixeira AL, Medeiros R. DNA repair system and renal cell carcinoma prognosis: under the influence of NBS1. Med Oncol 2015; 32:255. [PMID: 26493193 DOI: 10.1007/s12032-015-0701-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 10/09/2015] [Indexed: 01/06/2023]
Abstract
Nibrin (NBS1) is a protein involved in the maintenance of genomic stability and in DNA repair mechanisms. The NBS1 E185Q polymorphism (rs1805794) has been investigated in several studies, including its influence in the pathogenesis of renal cell carcinoma (RCC), although its prognostic value is still not determined for these patients. The purpose of the present work was to determine the role of NBS1 E185Q polymorphism as a prognostic factor/genetic marker of survival in patients with RCC. We conducted a hospital-based study analyzing 172 caucasian patients with histopathological diagnosis of RCC, for which polymorphism genotyping was performed by TaqMan(®) Allelic Discrimination methodology. In this study, we have found that male patients, non-metastatic at diagnosis and NBS1 C allele carriers (GC/CC) showed a lower 5-years survival when compared with GG genotype patients (P = 0.045). Furthermore, for carriers of low-activity NBS1 C allele, multivariate Cox regression analysis revealed almost a fourfold increase in risk of death at 5 years, after adjustment for age, histological type, Fuhrman's grade, tumor size and vascular permeation (HR 3.92; 95 % CI 1.33-11.57; P = 0.013). There were no statistically significant differences between the NBS1 E185Q genotypes and the assessed patients' clinical-pathological characteristics. Our results demonstrate for the first time the impact of NBS1 E185Q polymorphism in RCC prognosis suggesting that, for RCC male patients non-metastatic at diagnosis, this polymorphism might be a putative genetic marker in the clinical outcome.
Collapse
Affiliation(s)
- Alina Rosinha
- Oncology Department, Portuguese Institute of Oncology, Porto, Portugal.,Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal
| | - Joana Assis
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal.,FMUP, Faculty of Medicine of Porto University, Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal
| | - Augusto Nogueira
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal.,FMUP, Faculty of Medicine of Porto University, Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology, Porto, Portugal.,Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Porto, Portugal
| | - Joaquina Maurício
- Oncology Department, Portuguese Institute of Oncology, Porto, Portugal
| | - Ana Luísa Teixeira
- Oncology Department, Portuguese Institute of Oncology, Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group - Research Center, Portuguese Institute of Oncology, Porto, Portugal. .,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Porto, Portugal. .,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, Porto, Portugal. .,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal. .,IPO Porto, R Ant Bernardino de Almeida, 4200-072, Porto, Portugal.
| |
Collapse
|
17
|
Yu SS, Tu Y, Xu LL, Tao XQ, Xu S, Wang SS, Xiong YF, Mei JH. Effects of Rad51 on Survival of A549 Cells. Asian Pac J Cancer Prev 2015; 16:175-9. [DOI: 10.7314/apjcp.2015.16.1.175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
18
|
Oji Y, Tatsumi N, Kobayashi J, Fukuda M, Ueda T, Nakano E, Saito C, Shibata S, Sumikawa M, Fukushima H, Saito A, Hojo N, Suzuki M, Hoshikawa T, Shimura T, Morii E, Oka Y, Hosen N, Komatsu K, Sugiyama H. Wilms' tumor gene WT1 promotes homologous recombination-mediated DNA damage repair. Mol Carcinog 2014; 54:1758-71. [PMID: 25418835 DOI: 10.1002/mc.22248] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/05/2014] [Accepted: 10/10/2014] [Indexed: 01/16/2023]
Abstract
The Wilms' tumor gene WT1 is overexpressed in leukemia and various types of solid tumors and plays an oncogenic role in these malignancies. Alternative splicing at two sites yields four major isoforms, 17AA(+)KTS(+), 17AA(+)KTS(-), 17AA(-)KTS(+), and 17AA(-)KTS(-), and all the isoforms are expressed in the malignancies. However, among the four isoforms, function of WT1[17AA(-)KTS(+)] isoform still remains undetermined. In the present study, we showed that forced expression of WT1[17AA(-)KTS(+)] isoform significantly inhibited apoptosis by DNA-damaging agents such as Doxorubicin, Mitomycin, Camptothesisn, and Bleomycin in immortalized fibroblast MRC5SV and cervical cancer HeLa cells. Knockdown of Rad51, an essential factor for homologous recombination (HR)-mediated DNA repair canceled the resistance to Doxorubicin induced by WT1[17AA(-)KTS(+)] isoform. GFP recombination assay showed that WT1[17AA(-)KTS(+)] isoform alone promoted HR, but that three other WT1 isoforms did not. WT1[17AA(-)KTS(+)] isoform significantly upregulated the expression of HR genes, XRCC2, Rad51D, and Rad54. Knockdown of XRCC2, Rad51D, and Rad54 inhibited the HR activity and canceled resistance to Doxorubicin in MRC5SV cells with forced expression of WT1[17AA(-)KTS(+)] isoform. Furthermore, chromatin immunoprecipitation (ChIP) assay showed the binding of WT1[17AA(-)KTS(+)] isoform protein to promoters of XRCC2 and Rad51D. Immunohistochemical study showed that Rad54 and XRCC2 proteins were highly expressed in the majority of non-small-cell lung cancer (NSCLC) and gastric cancer, and that expression of these two proteins was significantly correlated with that of WT1 protein in NSCLCs. Our results presented here showed that WT1[17AA(-)KTS(+)] isoform had a function to promote HR-mediated DNA repair.
Collapse
Affiliation(s)
- Yusuke Oji
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoya Tatsumi
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Mari Fukuda
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tazu Ueda
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eri Nakano
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chisae Saito
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Syohei Shibata
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mihoko Sumikawa
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisashi Fukushima
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akari Saito
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nozomi Hojo
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Miyu Suzuki
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Hoshikawa
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tsutomu Shimura
- Department of Environmental Health, National Institute of Public Health, Saitama, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoki Hosen
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenshi Komatsu
- Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
19
|
The Role of Inflammation in Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 816:1-23. [DOI: 10.1007/978-3-0348-0837-8_1] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|