1
|
Nakamura T, Hatanaka KC, Kawamoto Y, Kaneko S, Ishida K, Harada K, Yuki S, Komatsu Y, Hatanaka Y, Sakamoto N. Lack of association between SLFN11 expression and treatment efficacy or survival outcomes in patients with pancreatic ductal adenocarcinoma. J Cancer Res Clin Oncol 2025; 151:159. [PMID: 40346357 PMCID: PMC12064574 DOI: 10.1007/s00432-025-06216-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 04/27/2025] [Indexed: 05/11/2025]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Despite the use of aggressive combination chemotherapy regimens, outcomes remain unsatisfactory. Schlafen family member 11 (SLFN11) has been reported to regulate the DNA damage response and influence tumor sensitivity to certain chemotherapeutic agents. This study aimed to investigate the expression of SLFN11 in PDAC and its potential as a biomarker for predicting treatment efficacy and survival outcomes. METHODS This retrospective observational cohort study included 158 patients with unresectable or borderline resectable PDAC who received palliative chemotherapy. Patients were classified into three groups: metastatic, locally advanced, and borderline resectable PDAC. Immunohistochemical staining for SLFN11 was performed on biopsy specimens, and expression levels were quantified using the histo-score (H-score). Associations between SLFN11 expression and clinical outcomes, including progression-free survival and overall survival, were analyzed using Kaplan-Meier methods and Cox regression models. RESULTS SLFN11 expression was observed in 54.4% of PDAC tissues. The median H-score for SLFN11 expression was higher in metastatic cases than in locally advanced and borderline resectable cases. However, no significant association was found between SLFN11 expression and the efficacy of chemotherapy or clinical outcomes. CONCLUSION Despite the hypothesized role of SLFN11 as a predictive biomarker for chemotherapy efficacy, no significant association was found between SLFN11 expression and clinical outcomes in PDAC. Further studies with larger cohorts and more detailed staging are needed to clarify the potential utility of SLFN11 as a therapeutic biomarker in PDAC.
Collapse
Affiliation(s)
- Takeaki Nakamura
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan.
| | - Shiho Kaneko
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Koichi Ishida
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Kazuaki Harada
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
2
|
Ogawa A, Izumikawa K, Tate S, Isoyama S, Mori M, Fujiwara K, Watanabe S, Ohga T, Jo U, Taniyama D, Kitajima S, Tanaka S, Onji H, Kageyama SI, Yamamoto G, Saito H, Morita TY, Okada M, Natsumeda M, Nagahama M, Kobayashi J, Ohashi A, Sasanuma H, Higashiyama S, Dan S, Pommier Y, Murai J. SLFN11-mediated ribosome biogenesis impairment induces TP53-independent apoptosis. Mol Cell 2025; 85:894-912.e10. [PMID: 39909041 PMCID: PMC11890970 DOI: 10.1016/j.molcel.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/11/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
Impairment of ribosome biogenesis (RiBi) triggered by inhibition of ribosomal RNA (rRNA) synthesis and processing leads to various biological effects. We report that Schlafen 11 (SLFN11) induces TP53-independent apoptosis through RiBi impairment. Upon replication stress, SLFN11 inhibits rRNA synthesis with RNA polymerase I accumulation and increased chromatin accessibility in the ribosomal DNA (rDNA) genes. SLFN11-dependent RiBi impairment preferentially depletes short-lived proteins, particularly MCL1, leading to apoptosis in response to replication stress. SLFN11's Walker B motif (E669), DNA-binding site (K652), dephosphorylation site for single-strand DNA binding (S753), and RNase sites (E209/E214) are all required for the SLFN11-mediated RiBi impairment. Comparable effects were obtained with direct RNA polymerase I inhibitors and other RiBi inhibitory conditions regardless of SLFN11. These findings were extended across 34 diverse human cancer cell lines. Thus, we demonstrate that RiBi impairment is a robust inactivator of MCL1 and an additional proapoptotic mechanism by which SLFN11 sensitizes cancer cells to chemotherapeutic agents.
Collapse
Affiliation(s)
- Akane Ogawa
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Keiichi Izumikawa
- Laboratory of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Sota Tate
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Sho Isoyama
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Masaru Mori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Kohei Fujiwara
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Soyoka Watanabe
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Takayuki Ohga
- Laboratory of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Daiki Taniyama
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Shojiro Kitajima
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Soichiro Tanaka
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Hiroshi Onji
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Shun-Ichiro Kageyama
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Gaku Yamamoto
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Hitoshi Saito
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Tomoko Yamamori Morita
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masami Nagahama
- Laboratory of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Junya Kobayashi
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Department of Radiological Sciences, School of Health Sciences at Narita, International University of Health and Welfare, Narita, Tokyo 286-0048, Japan
| | - Akihiro Ohashi
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Hiroyuki Sasanuma
- Department of Genome Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-0057, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan; Department of Oncogenesis and Tumor Regulation, Osaka International Cancer Institute, Osaka 103-0027, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814, USA.
| | - Junko Murai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan; Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan; Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
3
|
Karim NA, Miao J, Reckamp KL, Gay CM, Byers LA, Zhao YQ, Redman MW, Carrizosa DR, Wang WL, Petty WJ, Mehta K, Faller BA, Agamah ES, Kasbari SS, Malisetti RK, Kumar A, Schallenkamp J, Alluri KC, Gray JE, Kelly K. Phase II Randomized Study of Maintenance Atezolizumab Versus Atezolizumab Plus Talazoparib in Patients With SLFN11 Positive Extensive-Stage SCLC: S1929. J Thorac Oncol 2025; 20:383-394. [PMID: 39505259 DOI: 10.1016/j.jtho.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/03/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE To evaluate whether the addition of a poly (adenosine diphosphate-ribose) polymerase inhibitor talazoparib to maintenance immune checkpoint inhibitor atezolizumab after frontline chemoimmunotherapy improved outcomes in patients with Schlafen 11 (SLFN11)-positive extensive-stage SCLC (ES-SCLC). METHODS Patients with newly diagnosed SLFN11 expressing (H-score ≥ 1, evaluated centrally) ES-SCLC were randomized to maintenance atezolizumab (A) versus atezolizumab plus talazoparib (AT) after frontline chemotherapy plus atezolizumab. The primary objective was to compare progression-free survival (PFS) using a one-sided 10% level stratified log-rank test. Secondary endpoints included objective response rate, overall survival, and toxicity. The target sample size was 84 eligible patients. RESULTS From June 15, 2020, to December 15, 2022, 106 eligible patients were randomized (54 to AT and 52 to A). Progression-free survival was improved with AT versus A (hazard ratio = 0.66, 80% confidence interval: 0.50-0.86, one-sided p = 0.019) with a median PFS of 2.9 and 2.4 months; overall survival was not different between groups (hazard ratio = 0.98, 80% confidence interval: 0.71-1.36, one-sided p = 0.47). Grade 3 and higher non-hematologic treatment-related adverse events occurred in 17% of patients with AT and 14% of patients with A. Grade 3 and higher hematological treatment-related adverse events were more common in AT (50%) than in A (4%) (p < 0.001). CONCLUSION Maintenance AT improved PFS in patients with SLFN11-positive ES-SCLC that did not progress after initial chemo-immunotherapy. Hematologic toxicity, primarily grade 3 anemia, was increased with AT, as expected. Prospective biomarker selection was demonstrated, paving the way for future evaluation of novel therapies in molecularly defined SCLC populations.
Collapse
Affiliation(s)
- Nagla Abdel Karim
- Inova Schar Cancer Institute, Fairfax, Virginia; University of Virginia, Fairfax, Virginia.
| | - Jieling Miao
- SWOG Statistical Center and Data Management Center, Seattle, Washington; Fred Hutchinson Cancer Center, Seattle, Washington
| | - Karen L Reckamp
- Cedars-Sinai Medical Center, Los Angeles, California; Department of Thoracic Head and Neck Medical Oncology, The University of Texas Maryland
| | | | | | - Ying-Qi Zhao
- SWOG Statistical Center and Data Management Center, Seattle, Washington; Fred Hutchinson Cancer Center, Seattle, Washington
| | - Mary W Redman
- SWOG Statistical Center and Data Management Center, Seattle, Washington; Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Wei-Lien Wang
- Department of Pathology, MD Anderson Cancer Center, The University of Texas Houston, Texas
| | | | - Kathan Mehta
- Medstar Georgetown Cancer Institute, Washington, District of Columbia
| | | | | | - Samer S Kasbari
- Southeastern Medical Oncology Center, Goldsboro, North Carolina
| | - Rajini K Malisetti
- Minnesota Oncology Hematology PA - Coon Rapids, Minneapolis, Minneapolis
| | - Atul Kumar
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | | | | | | | - Karen Kelly
- UC Davis Comprehensive Cancer Center, Sacramento, California
| |
Collapse
|
4
|
Kaczorowski M, Chłopek M, Daum O, Ylaya K, Vaněček T, Szczepaniak M, Krawczyk K, Kowalik A, Michal M, Lasota J, Miettinen M. MMR deficiency is frequent in colorectal carcinomas with diffuse SLFN11 immunostaining: clinicopathologic and molecular study of 31 cases identified among 3,300 tumors. J Pathol Clin Res 2025; 11:e70025. [PMID: 40105034 PMCID: PMC11920882 DOI: 10.1002/2056-4538.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
Schlafen 11 (SLFN11), a regulator of cell fate following DNA injury, sensitizes tumor cells to DNA-damaging agents. Patients with SLFN11-positive tumors may benefit from DNA-damaging chemotherapies. SLFN11 has been studied in different types of cancer including colorectal carcinomas. However, colorectal carcinomas with diffuse positivity (expression in ≥80% of tumor cells) have not been meticulously characterized. SLFN11 immunostaining of tumor microarrays (TMAs) with 3,300 primary CRCs identified 65 (~2.0%) tumors with focal staining (<10% of tumor nuclei positive), 83 (~2.5%) with patchy (≥10% and <80%) and 51 (~1.5%) with diffuse (≥80%) SLFN11 positivity. The latter was confirmed on full sections from donor blocks in 31 (~1%) cases, which were further studied including evaluation of additional immunohistochemical markers, genotyping with targeted DNA sequencing, and assessment of microsatellite instability. SLFN11-positive carcinomas were mostly (21/31, 68%) right-sided tumors with a female predominance (21/31, 68%) and median age of 67 years. Eighteen of 31 (58%) contained areas of mucinous differentiation. Deficiency of mismatch repair proteins was detected in 65% (20/31) of SLFN11-positive carcinomas. Moreover, MLH1 (n = 2), MSH2, MSH6, and PMS2 germline mutations were identified in 25% (5/20) of patients with mismatch repair deficient tumors. BRAF p.V600E mutation was found in 45% (9/20) of mismatch repair deficient, but only 1 of 11 proficient tumors. Colorectal carcinomas with diffuse SLFN11 positivity were often mismatch repair deficient tumors with their typical clinical, morphological, and molecular characteristics.
Collapse
Affiliation(s)
- Maciej Kaczorowski
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
- Department of Clinical and Experimental Pathology, Wrocław Medical University, Wrocław, Poland
| | | | - Ondřej Daum
- Sikl's Institute of Pathology, Faculty of Medicine and Teaching Hospital in Plzen, Charles University, Plzen, Czech Republic
- Bioptical Laboratory, Ltd., Plzen, Czech Republic
| | - Kris Ylaya
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Tomáš Vaněček
- Sikl's Institute of Pathology, Faculty of Medicine and Teaching Hospital in Plzen, Charles University, Plzen, Czech Republic
- Bioptical Laboratory, Ltd., Plzen, Czech Republic
| | | | - Karol Krawczyk
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Michal Michal
- Sikl's Institute of Pathology, Faculty of Medicine and Teaching Hospital in Plzen, Charles University, Plzen, Czech Republic
- Bioptical Laboratory, Ltd., Plzen, Czech Republic
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
5
|
Masuda K, Yoshida T, Motoi N, Shinno Y, Matsumoto Y, Okuma Y, Goto Y, Horinouchi H, Yamamoto N, Watanabe S, Hoshino T, Yatabe Y, Ohe Y. Schlafen 11 Expression in Patients With Small Cell Lung Cancer and Its Association With Clinical Outcomes. Thorac Cancer 2025; 16:e15529. [PMID: 39809728 PMCID: PMC11732703 DOI: 10.1111/1759-7714.15529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/25/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Schlafen 11 (SLFN-11) has been identified as a sensitizer of tumor cells to DNA-damaging agents. However, the relationship between SLFN-11 expression and clinical outcomes in patients with small cell lung cancer (SCLC) remains unexplored. Thus, we aimed to evaluate the impact of SLFN-11 expression on survival in patients with limited-stage (LS) SCLC. METHODS We conducted a retrospective review of data from patients pathologically diagnosed with LS-SCLC post-surgery between January 2008 and December 2018. SLFN-11 expression was assessed using immunohistochemistry in tissue microarrays and scored using a histology (H)-score (range: 0-300). RESULTS Overall, 86 patients were included in the analysis with a median H-score of 43 for SLFN-11 expression. Among the patients, 44 had high SLFN-11 expression (provisionally defined as H-score ≥ 43). No significant differences in clinical profiles were observed between the two groups (high and low SLFN expression). The median survival durations were not reached (NR; 95% confidence interval [CI]: 65.1 months to NR) and 33.5 months (95% CI: 24.2 months to NR) for patients with high and low SLFN-11 expression, respectively (hazard ratio [HR]: 0.40, 95% CI: 0.19-0.81; p = 0.012). Among patients who relapsed post-surgery (n = 21), the median survival durations were 22.0 (95% CI: 7.6-44.9 months) and 8.1 (95% CI: 1.8-24.6 months) months in patients with high and low SLFN-11 expression, respectively (HR: 0.22, 95% CI: 0.06-0.84; p = 0.026). CONCLUSIONS High SLFN-11 expression is associated with relatively longer survival in patients with LS-SCLC in both those undergoing surgery and those who have relapsed.
Collapse
Affiliation(s)
- Ken Masuda
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Tatsuya Yoshida
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | - Noriko Motoi
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of PathologySaitama Cancer CenterSaitamaJapan
| | - Yuki Shinno
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Yuji Matsumoto
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Yusuke Okuma
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Yasushi Goto
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | | | - Noboru Yamamoto
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | | | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of MedicineKurume University School of MedicineFukuokaJapan
| | - Yasushi Yatabe
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
| | - Yuichiro Ohe
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
6
|
König P, Eichhorn JM, Suparman E, Bückreiß N, Cinatl J, Michaelis M, Bendas G. SLFN11 and ATR as targets for overcoming cisplatin resistance in ovarian cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167448. [PMID: 39117290 DOI: 10.1016/j.bbadis.2024.167448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
The levels and activities of the DNA/RNA helicase schlafen11 (SLFN11) and the serine/threonine-protein kinase ataxia telangiectasia and Rad3-related protein (ATR) may determine cancer cell sensitivity to DNA damaging agents, including platinum drugs. Here, we studied the roles of SLFN11 and ATR in cisplatin resistance of ovarian cancer using cell lines displaying acquired or intrinsic cisplatin resistance. W1CR, the cisplatin-resistant subline of W1 ovarian cancer cells, displayed reduced SLFN11 levels. HDAC inhibition using entinostat returned an epigenetic downregulation of SLFN11 in W1CR cells, caused SLFN11 re-expression and re-sensitized these cells to cisplatin. Moreover, entinostat also sensitized intrinsically resistant EFO21 ovarian cancer cells to cisplatin by upregulating SLFN11. However, SLFN11 was not involved in cisplatin resistance in all other cell models. Thus, SLFN11 expression is not a general cisplatin resistance marker in ovarian cancer. In contrast, inhibition of the DNA damage repair master regulator ATR using sub-toxic concentrations of elimusertib sensitized parental cell lines as well as intrinsically resistant EFO21 cells to cisplatin, and fully reversed acquired cisplatin resistance in cisplatin-adapted sublines W1CR, A2780cis, and KuramochirCDDP2000. Mechanisms underlying ATR-mediated cisplatin resistance differed between the cell lines and included CHK1/WEE1 signaling and induction of homologous recombination. In conclusion, SLFN11 and ATR are involved in ovarian cancer cisplatin resistance. Although our data identify ATR as key target for tackling cisplatin resistance in ovarian cancer, future studies are needed to identify biomarkers that indicate, which individual ovarian cancers benefit from SLFN11 re-activation and/or ATR inhibition.
Collapse
Affiliation(s)
- Philipp König
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany
| | | | - Eloy Suparman
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany
| | - Nico Bückreiß
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany
| | - Jindrich Cinatl
- Institute of Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany; Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60528 Frankfurt am Main, Germany
| | - Martin Michaelis
- Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60528 Frankfurt am Main, Germany; School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
| | - Gerd Bendas
- Department of Pharmacy, University Bonn, 53121 Bonn, Germany.
| |
Collapse
|
7
|
Moosavi F, Hassani B, Nazari S, Saso L, Firuzi O. Targeting DNA damage response in pancreatic ductal adenocarcinoma: A review of preclinical and clinical evidence. Biochim Biophys Acta Rev Cancer 2024; 1879:189185. [PMID: 39326802 DOI: 10.1016/j.bbcan.2024.189185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with one of the most unfavorable prognoses across all malignancies. In this review, we investigate the role of inhibitors targeting crucial regulators of DNA damage response (DDR) pathways, either as single treatments or in combination with chemotherapeutic agents and targeted therapies in PDAC. The most prominent clinical benefit of PARP inhibitors' monotherapy is related to the principle of synthetic lethality in individuals harboring BRCA1/2 and other DDR gene mutations as predictive biomarkers. Moreover, induction of BRCAness with inhibitors of RTKs, including VEGFR and c-MET and their downstream signaling pathways, RAS/RAF/MEK/ERK and PI3K/AKT/mTOR in order to expand the application of PARP inhibitors in patients without DDR mutations, has also been addressed. Other DDR-targeting agents beyond PARP inhibitors, including inhibitors of ATM, ATR, CHEK1/2, and WEE1 have also demonstrated their potential in preclinical models of PDAC and may hold promise in future studies.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Hassani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Bedore S, van der Eerden J, Boghani F, Patel SJ, Yassin S, Aguilar K, Lokeshwar VB. Protein-Based Predictive Biomarkers to Personalize Neoadjuvant Therapy for Bladder Cancer-A Systematic Review of the Current Status. Int J Mol Sci 2024; 25:9899. [PMID: 39337385 PMCID: PMC11432686 DOI: 10.3390/ijms25189899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The clinical outcome of patients with muscle-invasive bladder cancer (MIBC) is poor despite the approval of neoadjuvant chemotherapy or immunotherapy to improve overall survival after cystectomy. MIBC subtypes, immune, transcriptome, metabolomic signatures, and mutation burden have the potential to predict treatment response but none have been incorporated into clinical practice, as tumor heterogeneity and lineage plasticity influence their efficacy. Using the PRISMA statement, we conducted a systematic review of the literature, involving 135 studies published within the last five years, to identify studies reporting on the prognostic value of protein-based biomarkers for response to neoadjuvant therapy in patients with MIBC. The studies were grouped based on biomarkers related to molecular subtypes, cancer stem cell, actin-cytoskeleton, epithelial-mesenchymal transition, apoptosis, and tumor-infiltrating immune cells. These studies show the potential of protein-based biomarkers, especially in the spatial context, to reduce the influence of tumor heterogeneity on a biomarker's prognostic capability. Nevertheless, currently, there is little consensus on the methodology, reagents, and the scoring systems to allow reliable assessment of the biomarkers of interest. Furthermore, the small sample size of several studies necessitates the validation of potential prognostic biomarkers in larger multicenter cohorts before their use for individualizing neoadjuvant therapy regimens for patients with MIBC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vinata B. Lokeshwar
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA; (S.B.); (J.v.d.E.); (S.J.P.); (S.Y.); (K.A.)
| |
Collapse
|
9
|
Kaczorowski M, Ylaya K, Chłopek M, Taniyama D, Pommier Y, Lasota J, Miettinen M. Immunohistochemical Evaluation of Schlafen 11 (SLFN11) Expression in Cancer in the Search of Biomarker-Informed Treatment Targets: A Study of 127 Entities Represented by 6658 Tumors. Am J Surg Pathol 2024:00000478-990000000-00409. [PMID: 39185596 DOI: 10.1097/pas.0000000000002299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Schlafen 11 (SLFN11), a DNA/RNA helicase, acts as a regulator of cellular response to replicative stress and irreversibly triggers replication block and cell death. Several preclinical in vitro studies and clinical trials established that SLFN11 expression predicts outcomes in patients with advanced cancer treated with DNA-damaging chemotherapeutics and more recently with poly(ADP-ribose) polymerase inhibitors. SLFN11 expression status remains unknown in many cancer types, especially in mesenchymal tumors. This study evaluated a cohort of well characterized 3808 epithelial and 2850 mesenchymal and neuroectodermal tumors for SLFN11 expression using immunohistochemistry. Nuclear SLFN11 expression was rare in some of the most common carcinomas, for example, hepatocellular (1%), prostatic (2%), colorectal (5%), or breast (16%) cancers. In contrast, other epithelial tumors including mesotheliomas (92%), clear cell renal cell carcinomas (79%), small cell lung cancers (76%), squamous cell carcinomas of the tonsil (89%) and larynx (71%), or ovarian serous carcinomas (69%) were mostly SLFN11-positive. Compared with epithelial malignancies, SLFN11 expression was overall higher in neuroectodermal and mesenchymal tumors. Most positive entities included desmoplastic small round cell tumor (100%), Ewing sarcoma (92%), undifferentiated sarcoma (92%), solitary fibrous tumor (91%), dedifferentiated liposarcoma (89%), synovial sarcoma (86%), and malignant peripheral nerve sheath tumor (85%). Also, this study identifies tumors with potentially worse response to DNA-damaging drugs including antibody drug conjugates due to the absence of SLFN11 expression. Such entities may benefit from alternative treatments or strategies to overcome SLFN11 deficiency-related drug resistance. Our approach and results should serve as a foundation for future biomarker-associated clinical trials.
Collapse
Affiliation(s)
- Maciej Kaczorowski
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, Wrocław, Poland
| | - Kris Ylaya
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | | | - Daiki Taniyama
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| |
Collapse
|
10
|
Perez RE, Eckerdt F, Platanias LC. Schlafens: Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:1805. [PMID: 38791884 PMCID: PMC11119473 DOI: 10.3390/cancers16101805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
The interferon (IFN) family of immunomodulatory cytokines has been a focus of cancer research for over 50 years with direct and indirect implications in cancer therapy due to their properties to inhibit malignant cell proliferation and modulate immune responses. Among the transcriptional targets of the IFNs is a family of genes referred to as Schlafens. The products of these genes, Schlafen proteins, exert important roles in modulating cellular proliferation, differentiation, immune responses, viral replication, and chemosensitivity of malignant cells. Studies have demonstrated that abnormal expression of various Schlafens contributes to the pathophysiology of various cancers. Schlafens are now emerging as promising biomarkers and potentially attractive targets for drug development in cancer research. Here, we highlight research suggesting the use of Schlafens as cancer biomarkers and the rationale for the development of specific drugs targeting Schlafen proteins.
Collapse
Affiliation(s)
- Ricardo E. Perez
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (R.E.P.); (F.E.)
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Frank Eckerdt
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (R.E.P.); (F.E.)
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (R.E.P.); (F.E.)
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Akashi H, Yachida N, Ueda H, Yamaguchi M, Yamawaki K, Tamura R, Suda K, Ishiguro T, Adachi S, Nagase Y, Ueda Y, Ueda M, Abiko K, Kagabu M, Baba T, Nakaoka H, Enomoto T, Murai J, Yoshihara K. SLFN11 is a BRCA Independent Biomarker for the Response to Platinum-Based Chemotherapy in High-Grade Serous Ovarian Cancer and Clear Cell Ovarian Carcinoma. Mol Cancer Ther 2024; 23:106-116. [PMID: 37717249 DOI: 10.1158/1535-7163.mct-23-0257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/12/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
BRCA1/2 mutations are robust biomarkers for platinum-based chemotherapy in epithelial ovarian cancers. However, BRCA1/2 mutations in clear cell ovarian carcinoma (CCC) are less frequent compared with high-grade serous ovarian cancer (HGSC). The discovery of biomarkers that can be applied to CCC is an unmet need in chemotherapy. Schlafen 11 (SLFN11) has attracted attention as a novel sensitizer for DNA-damaging agents including platinum. In this study, we investigated the utility of SLFN11 in HGSC and CCC for platinum-based chemotherapy. SLFN11 expression was analyzed retrospectively by IHC across 326 ovarian cancer samples. The clinicopathologic significance of SLFN11 expression was analyzed across 57 advanced HGSC as a discovery set, 96 advanced HGSC as a validation set, and 57 advanced CCC cases, all of whom received platinum-based chemotherapy. BRCA1/2 mutation was analyzed using targeted-gene sequencing. In the HGSC cohort, the SLFN11-positive and BRCA mutation group showed significantly longer whereas the SLFN11-negative and BRCA wild-type group showed significantly shorter progression-free survival and overall survival. Moreover, SLFN11-positive HGSC shrunk significantly better than SLFN11-negative HGSC after neoadjuvant chemotherapy. Comparable results were obtained with CCC but without consideration of BRCA1/2 mutation due to a small population. Multivariate analysis identified SLFN11 as an independent factor for better survival in HGSC and CCC. The SLFN11-dependent sensitivity to platinum and PARP inhibitors were validated with genetically modified non-HGSC ovarian cancer cell lines. Our study reveals that SLFN11 predicts platinum sensitivity in HGSC and CCC independently of BRCA1/2 mutation status, indicating that SLFN11 assessment can guide treatment selection in HGSC and CCC.
Collapse
Affiliation(s)
- Hidehiko Akashi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Nozomi Yachida
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Haruka Ueda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Manako Yamaguchi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kaoru Yamawaki
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryo Tamura
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuaki Suda
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuya Ishiguro
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sosuke Adachi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshikazu Nagase
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masashi Ueda
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Masahiro Kagabu
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Shiwa, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Shiwa, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation Chiyoda-ku, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Junko Murai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
12
|
Fujiwara K, Maekawa M, Iimori Y, Ogawa A, Urano T, Kono N, Takeda H, Higashiyama S, Arita M, Murai J. The crucial role of single-stranded DNA binding in enhancing sensitivity to DNA-damaging agents for Schlafen 11 and Schlafen 13. iScience 2023; 26:108529. [PMID: 38125019 PMCID: PMC10730379 DOI: 10.1016/j.isci.2023.108529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/19/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Schlafen (SLFN) 11 enhances cellular sensitivity to various DNA-damaging anticancer agents. Among the human SLFNs (SLFN5/11/12/13/14), SLFN11 is unique in its drug sensitivity and ability to block replication under DNA damage. In biochemical analysis, SLFN11 binds single-stranded DNA (ssDNA), and this binding is enhanced by the dephosphorylation of SLFN11. In this study, human cell-based assays demonstrated that a point mutation at the ssDNA-binding site of SLFN11 or a constitutive phosphorylation mutant abolished SLFN11-dependent drug sensitivity. Additionally, we discovered that nuclear SLFN13 with a point mutation mimicking the DNA-binding site of SLFN11 was recruited to chromatin, blocked replication, and enhanced drug sensitivity. Through generating multiple mutants and structure analyses of SLFN11 and SLFN13, we identified protein phosphatase 2A as a binding partner of SLFN11 and the putative binding motif in SLFN11. These findings provide crucial insights into the unique characteristics of SLFN11, contributing to a better understanding of its mechanisms.
Collapse
Affiliation(s)
- Kohei Fujiwara
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-Ku, Tokyo 105-8512, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Masashi Maekawa
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-Ku, Tokyo 105-8512, Japan
| | - Yuki Iimori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Akane Ogawa
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Takeshi Urano
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane 693-8501, Japan
- Center for Vaccines and Therapeutic Antibodies for Emerging Infectious Diseases, Shimane University, Izumo, Shimane 693-8501, Japan
| | - Nobuaki Kono
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa 252-0882, Japan
| | - Hiroyuki Takeda
- Division of Proteo-Drug-Discovery, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
- Department of Oncogenesis and Tumor Regulation, Osaka International Cancer Institute, Chuo-Ku, Osaka 541-8567, Japan
| | - Makoto Arita
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Minato-Ku, Tokyo 105-8512, Japan
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa 230-0045, Japan
| | - Junko Murai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| |
Collapse
|
13
|
Cathomas R, Rothschild SI, Hayoz S, Bubendorf L, Özdemir BC, Kiss B, Erdmann A, Aeppli S, Mach N, Strebel RT, Hadaschik B, Berthold D, John H, Zihler D, Schmid M, Alborelli I, Schneider M, Musilova J, Spahn M, Petrausch U. Perioperative Chemoimmunotherapy With Durvalumab for Muscle-Invasive Urothelial Carcinoma: Primary Analysis of the Single-Arm Phase II Trial SAKK 06/17. J Clin Oncol 2023; 41:5131-5139. [PMID: 37590894 PMCID: PMC10666980 DOI: 10.1200/jco.23.00363] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/02/2023] [Accepted: 07/05/2023] [Indexed: 08/19/2023] Open
Abstract
PURPOSE The integration of immunotherapy in the perioperative setting of muscle-invasive urothelial carcinoma (MIUC) appears promising. SAKK 06/17 investigated the addition of neoadjuvant durvalumab to gemcitabine/cisplatin (GC) chemotherapy followed by radical surgery and adjuvant checkpoint inhibition with durvalumab. PATIENTS AND METHODS SAKK 06/17 was an investigator-initiated, open-label, single-arm phase II study including cisplatin-fit patients with stage cT2-T4a cN0-1 operable MIUC. Four cycles of neoadjuvant GC in combination with four cycles of durvalumab (start with GC cycle 2) were administered, followed by radical surgery. Adjuvant durvalumab was given for 10 cycles. The primary end point was event-free survival (EFS) at 2 years. RESULTS Sixty one patients were accrued at 12 sites. The full analysis set consisted of 57 patients, 54 (95%) had bladder cancer. Median follow-up was 40 months. The primary end point was met, with EFS at 2 years of 76% (one-sided 90% CI [lower bound], 67%; two-sided 95% CI, 62 to 85). EFS at 3 years was 73% (95% CI, 59 to 83). Complete pathologic response in resected patients (N = 52) was achieved in 17 patients (33%), and 31 (60%) had pathologic response CONCLUSION The addition of perioperative durvalumab to the standard of care for patients with resectable MIUC results in a high EFS and OS at 2 years.
Collapse
Affiliation(s)
- Richard Cathomas
- Division of Oncology/Hematology, Kantonsspital Graubünden, Chur, Switzerland
| | - Sacha I Rothschild
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital Basel, Basel, Switzerland
- Department of Oncology/Hematology, Kantonsspital Baden, Baden, Switzerland
| | | | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Berna C Özdemir
- Department of Oncology, Bern University Hospital, Bern, Switzerland
| | - Bernhard Kiss
- Department of Urology, Bern University Hospital, Bern, Switzerland
| | - Andreas Erdmann
- Department of Oncology/Hematology, Kantonsspital Baden, Baden, Switzerland
| | - Stefanie Aeppli
- Department of Oncology, University Hospital HUG, Geneva, Switzerland
| | - Nicolas Mach
- Department of Oncology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Räto T Strebel
- Division of Urology, Kantonsspital Graubünden, Chur, Switzerland
| | - Boris Hadaschik
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Dominik Berthold
- Department of Oncology, University Hospital CHUV, Lausanne, Switzerland
| | - Hubert John
- Department of Urology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Deborah Zihler
- Department of Oncology, Kantonsspital Aarau, Aarau, Switzerland
| | - Mathias Schmid
- Department of Oncology, Triemlispital, Zürich, Switzerland
| | - Ilaria Alborelli
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | | | | | - Martin Spahn
- Department of Urology, Bern University Hospital, Bern, Switzerland
- Hirslanden Klinik, Zürich, Switzerland
- Lindenhofspital Bern, Bern, Switzerland
| | | |
Collapse
|
14
|
Miladinov M, Rosic J, Eric K, Guzonjic A, Jelenkovic J, Bogavac-Stanojevic N, Dimitrijevic I, Kotur-Stevuljevic J, Barisic G. Analysis of the Prognostic Potential of Schlafen 11, Programmed Death Ligand 1, and Redox Status in Colorectal Cancer Patients. Int J Mol Sci 2023; 24:15083. [PMID: 37894765 PMCID: PMC10606719 DOI: 10.3390/ijms242015083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
The Schlafen 11 (SLFN11) protein has recently emerged as pivotal in DNA damage conditions, with predictive potential for tumor response to cytotoxic chemotherapies. Recent discoveries also showed that the programmed death ligand 1 (PD-L1) protein can be found on malignant cells, providing an immune evasion mechanism exploited by different tumors. Additionally, excessive generation of free radicals, redox imbalance, and consequential DNA damage can affect intestinal cell homeostasis and lead to neoplastic transformation. Therefore, our study aimed to investigate the significance of SLFN11 and PD-L1 proteins and redox status parameters as prognostic biomarkers in CRC patients. This study included a total of 155 CRC patients. SLFN11 and PD-L1 serum levels were measured with ELISA and evaluated based on redox status parameters, sociodemographic and clinical characteristics, and survival. The following redox status parameters were investigated: spectrophotometrically measured superoxide dismutase (SOD), sulfhydryl (SH) groups, advanced oxidation protein products (AOPP), malondialdehyde (MDA), pro-oxidant-antioxidant balance (PAB), and superoxide anion (O2•-). The prooxidative score, antioxidative score, and OXY-SCORE were also calculated. The results showed significantly shorter survival in patients with higher OXY-SCOREs and higher levels of serum SLFN11, while only histopathology-analysis-related factors showed significant prognostic value. OXY-SCORE and SLFN11 levels may harbor prognostic potential in CRC patients.
Collapse
Affiliation(s)
- Marko Miladinov
- Clinic for Digestive Surgery-First Surgical Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Jovana Rosic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Katarina Eric
- Department of Pathology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Azra Guzonjic
- Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelenko Jelenkovic
- Clinic for Digestive Surgery-First Surgical Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | | | - Ivan Dimitrijevic
- Clinic for Digestive Surgery-First Surgical Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Goran Barisic
- Clinic for Digestive Surgery-First Surgical Clinic, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
15
|
Murai J, Ceribelli M, Fu H, Redon CE, Jo U, Murai Y, Aladjem MI, Thomas CJ, Pommier Y. Schlafen 11 (SLFN11) Kills Cancer Cells Undergoing Unscheduled Re-replication. Mol Cancer Ther 2023; 22:985-995. [PMID: 37216280 PMCID: PMC10524552 DOI: 10.1158/1535-7163.mct-22-0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/24/2022] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Schlafen 11 (SLFN11) is an increasingly prominent predictive biomarker and a molecular sensor for a wide range of clinical drugs: topoisomerases, PARP and replication inhibitors, and platinum derivatives. To expand the spectrum of drugs and pathways targeting SLFN11, we ran a high-throughput screen with 1,978 mechanistically annotated, oncology-focused compounds in two isogenic pairs of SLFN11-proficient and -deficient cells (CCRF-CEM and K562). We identified 29 hit compounds that selectively kill SLFN11-proficient cells, including not only previously known DNA-targeting agents, but also the neddylation inhibitor pevonedistat (MLN-4924) and the DNA polymerase α inhibitor AHPN/CD437, which both induced SLFN11 chromatin recruitment. By inactivating cullin-ring E3 ligases, pevonedistat acts as an anticancer agent partly by inducing unscheduled re-replication through supraphysiologic accumulation of CDT1, an essential factor for replication initiation. Unlike the known DNA-targeting agents and AHPN/CD437 that recruit SLFN11 onto chromatin in 4 hours, pevonedistat recruited SLFN11 at late time points (24 hours). While pevonedistat induced unscheduled re-replication in SLFN11-deficient cells after 24 hours, the re-replication was largely blocked in SLFN11-proficient cells. The positive correlation between sensitivity to pevonedistat and SLFN11 expression was also observed in non-isogenic cancer cells in three independent cancer cell databases (NCI-60, CTRP: Cancer Therapeutics Response Portal and GDSC: Genomic of Drug Sensitivity in Cancer). The present study reveals that SLFN11 not only detects stressed replication but also inhibits unscheduled re-replication induced by pevonedistat, thereby enhancing its anticancer efficacy. It also suggests SLFN11 as a potential predictive biomarker for pevonedistat in ongoing and future clinical trials.
Collapse
Affiliation(s)
- Junko Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- Department of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon 791-0295, Japan
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon 791-0295, Japan
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Haiqing Fu
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Christophe E. Redon
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yasuhisa Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Mirit I. Aladjem
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
He Z, Gu J, Luan T, Li H, Li C, Chen Z, Luo E, Wang J, Huang Y, Ding M. Comprehensive analyses of a tumor-infiltrating lymphocytes-related gene signature regarding the prognosis and immunologic features for immunotherapy in bladder cancer on the basis of WGCNA. Front Immunol 2022; 13:973974. [PMID: 36211333 PMCID: PMC9540212 DOI: 10.3389/fimmu.2022.973974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor-infiltrating lymphocyte (TIL) is a class of cells with important immune functions and plays a crucial role in bladder cancer (BCa). Several studies have shown the clinical significance of TIL in predicting the prognosis and immunotherapy efficacy. TIL-related gene module was screened utilizing weighted gene coexpression network analysis. We screened eight TIL-related genes utilizing univariate Cox regression analysis, least absolute shrinkage and selection operator (LASSO) Cox regression analysis, and multivariate Cox regression analysis. Then, we established a TIL-related signature model containing the eight selected genes and subsequently classified all patients into two groups, that is, the high-risk as well as low-risk groups. Gene mutation status, prognosis, immune cell infiltration, immune subtypes, TME, clinical features, and immunotherapy response were assessed among different risk subgroups. The results affirmed that the TIL-related signature model was a reliable predictor of overall survival (OS) for BCa and was determined as an independent risk factor for BCa patients in two cohorts. Moreover, the risk score was substantially linked to age, tumor staging, TNM stage, and pathological grade. And there were different mutational profiles, biological pathways, immune scores, stromal scores, and immune cell infiltration in the tumor microenvironment (TME) between the two risk groups. In particular, immune checkpoint genes’ expression was remarkably different between the two risk groups, with patients belonging to the low-risk group responding better to immune checkpoint inhibition (ICI) therapy. In conclusion, our study demonstrates that the TIL-related model was a reliable signature in anticipating prognosis, immune status, and immunotherapy response, which can help in screening patients who respond to immunotherapy.
Collapse
Affiliation(s)
- Zexi He
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Gu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ting Luan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haihao Li
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Charles Li
- Zhongke Jianlan Medical Research Institute, Beijing, China
| | - Zhenjie Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Enxiu Luo
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yinglong Huang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Mingxia Ding, ; Yinglong Huang,
| | - Mingxia Ding
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Mingxia Ding, ; Yinglong Huang,
| |
Collapse
|
17
|
Onji H, Murai J. Reconsidering the mechanisms of action of PARP inhibitors based on clinical outcomes. Cancer Sci 2022; 113:2943-2951. [PMID: 35766436 PMCID: PMC9459283 DOI: 10.1111/cas.15477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/19/2022] [Accepted: 06/25/2022] [Indexed: 11/30/2022] Open
Abstract
PARP inhibitors (PARPis) were initially developed as DNA repair inhibitors that inhibit the catalytic activity of PARP1 and PARP2 and are expected to induce synthetic lethality in BRCA‐ or homologous recombination (HR)‐deficient tumors. However, the clinical indications for PARPis are not necessarily limited to BRCA mutations or HR deficiency; BRCA wild‐type and HR‐proficient cancers can also derive some benefit from PARPis. These facts are interpretable by an additional primary antitumor mechanism of PARPis named PARP trapping, resulting from the stabilization of PARP‐DNA complexes. Favorable response to platinum derivatives (cisplatin and carboplatin) in preceding treatment is used as a clinical biomarker for some PARPis, implying that sensitivity factors for platinum derivatives and PARPis are mainly common. Such common sensitivity factors include not only HR defects (HRD) but also additional factors. One of them is Schlafen 11 (SLFN11), a putative DNA/RNA helicase, that sensitizes cancer cells to a broad type of DNA‐damaging agents, including platinum and topoisomerase inhibitors. Mechanistically, SLFN11 induces a lethal replication block in response to replication stress (ie, DNA damage). As SLFN11 acts upon replication stress, trapping PARPis can activate SLFN11. Preclinical models show the importance of SLFN11 in PARPi sensitivity. However, the relevance of SLFN11 in PARPi response is less evident in clinical data compared with the significance of SLFN11 for platinum sensitivity. In this review, we consider the reasons for variable indications of PARPis resulting from clinical outcomes and review the mechanisms of action for PARPis as anticancer agents.
Collapse
Affiliation(s)
- Hiroshi Onji
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Japan.,Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Japan
| | - Junko Murai
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Japan.,Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| |
Collapse
|
18
|
Can Schlafen 11 Help to Stratify Ovarian Cancer Patients Treated with DNA-Damaging Agents? Cancers (Basel) 2022; 14:cancers14102353. [PMID: 35625957 PMCID: PMC9139752 DOI: 10.3390/cancers14102353] [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] [Received: 03/25/2022] [Revised: 04/25/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
Platinum-based chemotherapy has been the cornerstone of systemic treatment in ovarian cancer. Since no validated molecular predictive markers have been identified yet, the response to platinum-based chemotherapy has been evaluated clinically, based on platinum-free interval. The new promising marker Schlafen 11 seems to correlate with sensitivity or resistance to DNA-damaging agents, including platinum compounds or PARP inhibitors in various types of cancer. We provide background information about the function of Schlafen 11, its evaluation in tumor tissue, and its prevalence in ovarian cancer. We discuss the current evidence of the correlation of Schlafen 11 expression in ovarian cancer with treatment outcomes and the potential use of Schlafen 11 as the key predictive and prognostic marker that could help to better stratify ovarian cancer patients treated with platinum-based chemotherapy or PARP inhibitors. We also provide perspectives on future directions in the research on this promising marker.
Collapse
|