1
|
Hanf D, Fasching P, Gass P, Matthias W Beckmann, Hack CC, Heindl F, Lothar Häberle, John N, Erber R, Press MF, Rübner M, Pöschke P. Impact of CCND1 amplification on the prognosis of hormone receptor-positive, HER2-negative breast cancer patients-correlation of clinical and pathological markers. Breast Cancer Res Treat 2025; 210:125-134. [PMID: 39586971 PMCID: PMC11787164 DOI: 10.1007/s10549-024-07545-x] [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: 08/13/2024] [Accepted: 10/30/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE The cyclin D1 gene (CCND1) encodes a key cell-cycle regulatory protein. Resistance to endocrine therapy is reportedly observed more often in patients with CCND1-amplified tumors. CCND1 amplification is known to be a driving event in breast cancer, but contradictory findings are reported for its association with prognosis. This study therefore investigated the prognostic value of CCND1 amplification in hormone receptor (HR)-positive breast cancer patients. METHODS A cohort of 894 unselected breast cancer patients from the Bavarian Breast Cancer Cases and Controls (BBCC) study was included. The CCND1 amplification rate was evaluated in tissue microarrays using fluorescence in situ hybridization. A CCND1/CEP11 ratio ≥ 2.0 was considered amplified. Statistical analysis was conducted on cases with ratios based on a range of 20-100 nuclei analyzed per case. A univariable Cox regression model was fitted with disease-free survival (DFS) and overall survival (OS). RESULTS CCND1 gene status was assessable in 511 patients. The CCND1 amplification rate was 12.9% (66 patients). Most patients with CCND1 amplification had luminal B-Like-(51.5%, n = 34) or luminal A-Like tumors (25.8%, n = 17), 13 patients with HER2-positive disease (19.7%) and only two patients had triple-negative tumors (3.0%). Survival analysis, focused on HR-positive, HER2-negative patients, showed no statistically significant differences in the DFS and OS with and without CCND1 amplification (P = 0.20 and 0.14, respectively, in the unadjusted analysis). CONCLUSIONS CCND1 amplification is a recurring event in breast cancer, occurring most frequently in luminal B-like and HER2-amplified subtypes. A trend toward less favorable outcomes was observed among CCND1-amplified HR-positive, HER2-negative tumors.
Collapse
Affiliation(s)
- Dorothea Hanf
- Department of Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC) and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Translational Medical Oncology, Faculty of Medicine, Carl Gustav Carus University Hospital, TUD Dresden University of Technology, Dresden, Germany
- Partner Site Dresden, German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Fasching
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Paul Gass
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Carolin C Hack
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Felix Heindl
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Biostatistics Unit, Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nelson John
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Biostatistics Unit, Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ramona Erber
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Michael F Press
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Matthias Rübner
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Patrik Pöschke
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN) / Friedrich Alexander University of Erlangen-Nuremberg (FAU), Universitätsstrasse 21-23, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Alliance WERA (CCC), Erlangen, Germany.
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany.
| |
Collapse
|
2
|
Zhang G, Xia G, Zhang C, Li S, Wang H, Zheng D. Combined single cell and spatial transcriptome analysis reveals cellular heterogeneity of hedgehog pathway in gastric cancer. Genes Immun 2024; 25:459-470. [PMID: 39251886 DOI: 10.1038/s41435-024-00297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/04/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
Gastric cancer (GC) is one of the most common and deadly malignancies in the world. Abnormal activation of hedgehog pathway is closely related to tumor development and progression. However, potential therapeutic targets for GC based on the hedgehog pathway have not been clearly identified. In the present study, we combined single-cell sequencing data and spatial transcriptomics to deeply investigate the role of hedgehog pathway in GC. Based on a comprehensive scoring algorithm, we found that fibroblasts from GC tumor tissues were characterized by a highly enriched hedgehog pathway. By analyzing the development process of fibroblasts, we found that CCND1 plays an important role at the end stage of fibroblast development, which may be related to the formation of tumor-associated fibroblasts. Based on spatial transcriptome data, we deeply mined the role of CCND1 in fibroblasts. We found that CCND1-negative and -positive fibroblasts have distinct characteristics. Based on bulk transcriptome data, we verified that highly infiltrating CCND1 + fibroblasts are a risk factor for GC patients and can influence the immune and chemotherapeutic efficacy of GC patients. Our study provides unique insights into GC and hedgehog pathways and also new directions for cancer treatment strategies.
Collapse
Affiliation(s)
- Guoliang Zhang
- Department of General Surgery, Central Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Guojun Xia
- Department of General Surgery, Central Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Chunxu Zhang
- Department of General Surgery, Central Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Shaodong Li
- Department of General Surgery, Central Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Huangen Wang
- Department of General Surgery, Central Hospital of Shaoxing, Shaoxing, Zhejiang, China
| | - Difeng Zheng
- Department of General Surgery, Central Hospital of Shaoxing, Shaoxing, Zhejiang, China.
| |
Collapse
|
3
|
Roy J, Hemavathy N, Saravanan R, Gopinath P, Pugazh P, Jeyaraman J, Venkatraman G, Rayala SK. Spatio-temporal localization of P21-activated kinase in endometrial cancer. Biotechnol Appl Biochem 2024. [PMID: 39506238 DOI: 10.1002/bab.2684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024]
Abstract
Endometrial cancer is the sixth most common gynecologic cancer, and has been reported as a malignancy arising due to the idiopathic effects of certain anticancer agents. Tamoxifen is the drug of choice in ER-positive breast cancer, and several studies have shown better disease-free survival in these patients. However, the long-term usage of tamoxifen has been associated with resistance and risk for endometrial malignancy. A direct mechanistic basis for tamoxifen-induced endometrial tumorigenesis is still unclear. Hyperactivation of PAK1 in endometrial cancer correlates with poor overall survival. The present study demonstrates that tamoxifen treatment induces nuclear localization of PAK1 in endometrial carcinoma cells. This nuclear transit is mediated through JAK2 phosphorylation of PAK1 and binding of β-PIX. In addition, a computational approach involving molecular modeling and simulation of phosphorylated and unphosphorylated forms of PAK1 was used to elucidate the dynamics of nuclear localization. Thus, PAK1 phosphorylation by JAK2 is a prerequisite for its nuclear localization and its tumorigenic effects on endometrial cancer cells.
Collapse
Affiliation(s)
- Joydeep Roy
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| | - Nagarajan Hemavathy
- Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Roshni Saravanan
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Prarthana Gopinath
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| | - Pooja Pugazh
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | | | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu, India
| |
Collapse
|
4
|
Ju G, Sun Y, Wang H, Zhang X, Mu Z, Sun D, Huang L, Lin R, Xing T, Cheng W, Liang J, Lin YS. Fusion Oncogenes in Patients With Locally Advanced or Distant Metastatic Differentiated Thyroid Cancer. J Clin Endocrinol Metab 2024; 109:505-515. [PMID: 37622214 PMCID: PMC10795910 DOI: 10.1210/clinem/dgad500] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
CONTEXT Fusion oncogenes are involved in the underlying pathology of advanced differentiated thyroid cancer (DTC), and even the cause of radioactive iodine (RAI)-refractoriness. OBJECTIVE We aimed to investigation between fusion oncogenes and clinicopathological characteristics involving a large-scale cohort of patients with advanced DTC. METHODS We collected 278 tumor samples from patients with locally advanced (N1b or T4) or distant metastatic DTC. Targeted next-generation sequencing with a 26-gene ThyroLead panel was performed on these samples. RESULTS Fusion oncogenes accounted for 29.86% of the samples (72 rearrangement during transfection (RET) fusions, 7 neurotrophic tropomyosin receptor kinase (NTRK) fusions, 4 anaplastic lymphoma kinase (ALK) fusions) and occurred more frequently in pediatric patients than in their adult counterparts (P = .003, OR 2.411, 95% CI 1.329-4.311) in our cohort. DTCs with fusion oncogenes appeared to have a more advanced American Joint Committee on Cancer (AJCC)_N and AJCC_M stage (P = .0002, OR 15.47, 95% CI 2.54-160.9, and P = .016, OR 2.35, 95% CI 1.18-4.81) than those without. DTCs with fusion oncogenes were associated with pediatric radioactive iodine (RAI) refractoriness compared with those without fusion oncogenes (P = .017, OR 4.85, 95% CI 1.29-15.19). However, in adult DTCs, those with fusion oncogenes were less likely to be associated with RAI refractoriness than those without (P = .029, OR 0.50, 95% CI 0.27-0.95), owing to a high occurrence of the TERT mutation, which was the most prominent genetic risk factor for RAI refractoriness in multivariate logistic regression analysis (P < .001, OR 7.36, 95% CI 3.14-17.27). CONCLUSION Fusion oncogenes were more prevalent in pediatric DTCs than in their adult counterparts and were associated with pediatric RAI refractoriness, while in adult DTCs, TERT mutation was the dominant genetic contributor to RAI refractoriness rather than fusion oncogenes.
Collapse
Affiliation(s)
- Gaoda Ju
- Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
- Department of Oncology, Peking University International Hospital, Peking University, Beijing, 102206, China
| | - Yuqing Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Hao Wang
- Department of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Xin Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Zhuanzhuan Mu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Di Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Lisha Huang
- Department of Medical, Zhejiang Shaoxing Topgen Biomedical Technology Co., Ltd., Shanghai, 201321, China
| | - Ruijue Lin
- Department of Technology, Zhejiang Topgen Clinical Laboratory Co., Ltd., Huzhou, 201914, China
| | - Tao Xing
- Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Wuying Cheng
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| | - Jun Liang
- Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Oncology, Peking University International Hospital, Peking University, Beijing, 102206, China
| | - Yan-Song Lin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, 100730, China
| |
Collapse
|
5
|
Li Z, Tian JM, Chu Y, Zhu HY, Wang JJ, Huang J. Long non-coding RNA PVT1 (PVT1) affects the expression of CCND1 and promotes doxorubicin resistance in osteosarcoma cells. J Bone Oncol 2023; 43:100512. [PMID: 38021073 PMCID: PMC10665705 DOI: 10.1016/j.jbo.2023.100512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Background Acquired drug-resistance is the major risk factor for poor prognosis and short-term survival in patients with osteosarcoma (OS). Accumulating evidence has revealed that long noncoding RNAs (lncRNAs), including plasmacytoma variant translocation 1 (PVT1), play potential regulatory roles in the malignant development of OS. Considering the subcellular distribution of PVT1 as both nuclear and cytoplasmic lncRNA, a thorough exploration of its extensive mechanisms becomes essential. Methods The GEO database was utilized for the acquisition of gene expression data, which was subsequently analyzed to fulfill the research objectives. The subcellular localization of PVT1 in OS cells was determined using fluorescence in situ hybridization (FISH) and quantitative real-time polymerase chain reaction (qRT-PCR). Additionally, the sensitivity of OS cells to doxorubicin was comprehensively evaluated through measurements of cell viability, site-specific proliferation capacity, and the relative expression abundance of multidrug resistance-related proteins (MRPs). In order to investigate the differential response of OS cells with varying levels of PVT1 expression to doxorubicin, pulmonary metastasis mice models were established for in vivo studies. Molecular interactions were further examined using the dual-luciferase assay and RNA immunoprecipitation assay (RIP) to analyze the binding sites of miR-15a-5p and miR-15b-5p on PVT1 and G1/S-specific cyclinD1 (CCND1) mRNA. Furthermore, the chromatin immunoprecipitation (ChIP) and dual-luciferase assay were employed to assess the transcriptional activation of the proto-oncogene c-myc (MYC) on the CCND1 promoter and identify the corresponding binding sites. Results In doxorubicin resistant OS cells, transcription levels of PVT1, MYC and CCND1 were significantly higher than those in original cells. In vivo experiments demonstrated that OS cells rich in PVT1 expression exhibited enhanced tumorigenicity and resistance to doxorubicin. In vitro experiments, it has been observed that overexpression of PVT1 in OS cells is accompanied by an upregulation of CCND1, thereby facilitating resistance to doxorubicin. Nonetheless, this PVT1-induced resistance can be effectively attenuated by the knockdown of CCND1. Mechanistically, PVT1 functions as a competitive endogenous RNA (ceRNA) that influences the expression of CCND1 by inhibiting the degradation function of miR-15a-5p and miR-15b-5p on CCND1 mRNA. Additionally, as a neighboring gene of MYC, PVT1 plays a role in maintaining MYC protein stability, which further enhances MYC-dependent CCND1 transcriptional activity. Conclusion The resistance of OS cells to doxorubicin is facilitated by PVT1, which enhances the expression of CCND1 through a dual mechanism. This findings offer a novel perspective for comprehending the intricate regulatory mechanisms of long non-coding RNA in influencing the expression of coding genes.
Collapse
Affiliation(s)
- Zi Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia-Ming Tian
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Chu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong-Yi Zhu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun-Jie Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Huang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Pyo JS, Min KW, Oh IH, Lim DH, Son BK. Clinicopathological significance and the associated signaling pathway of p21-activated kinase 1 (PAK1) in colorectal cancer. Pathol Res Pract 2023; 251:154820. [PMID: 37801909 DOI: 10.1016/j.prp.2023.154820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 10/08/2023]
Abstract
The aim of this study was to evaluate the clinicopathological significance and associated signaling pathways of p21-activated kinase 1 (PAK1) in colorectal cancer (CRC). PAK1 immunohistochemical expression was investigated in 246 human CRC tissues to evaluate its clinicopathological significance and prognostic role. Correlations between PAK1 and the immunoscore, HIF-1α, and pFOXO1 were also evaluated. PAK1 was expressed in 169 of 246 CRC tissues (68.7%). PAK1 expression significantly correlated with the metastatic lymph node ratio (P = 0.023). However, PAK1 expression did not correlate with tumor size, tumor location, tumor differentiation, lymphovascular and perineural invasion, or distant metastasis. PAK1 expression was significantly higher in CRC with a low immunoscore than in CRC with a high immunoscore (P = 0.017). In addition, there were significant correlations between PAK1, HIF-1α, and pFOXO1 expression (P = 0.001 and P = 0.024, respectively). Patients with PAK1 expression had worse overall and recurrence-free survival than those without PAK1 expression (P 0.001 and P = 0.001, respectively). PAK1 expression was significantly correlated with worse prognosis in CRCs patients. In addition, PAK1 expression was significantly correlated with a low immunoscore and high expression of HIF-1α and pFOXO1 in CRCs.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Department of Pathology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Il Hwan Oh
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Dae Hyun Lim
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea
| | - Byoung Kwan Son
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
7
|
Skjervold AH, Valla M, Ytterhus B, Bofin AM. PAK1 copy number in breast cancer-Associations with proliferation and molecular subtypes. PLoS One 2023; 18:e0287608. [PMID: 37368917 DOI: 10.1371/journal.pone.0287608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION P21-activated kinase 1 (PAK1) is known to be overexpressed in several human tumour types, including breast cancer (BC). It is located on chromosome 11 (11q13.5-q14.1) and plays a significant role in proliferation in BC. In this study we aimed to assess PAK1 gene copy number (CN) in primary breast tumours and their corresponding lymph node metastases, and associations between PAK1 CN and proliferation status, molecular subtype, and prognosis. In addition, we aimed to study associations between CNs of PAK1 and CCND1. Both genes are located on the long arm of chromosome 11 (11q13). METHODS Fluorescence in situ hybridization for PAK1 and Chromosome enumeration probe (CEP)11 were used on tissue microarray sections from a series of 512 BC cases. Copy numbers were estimated by counting the number of fluorescent signals for PAK1 and CEP11 in 20 tumour cell nuclei. Pearson's x2 test was performed to assess associations between PAK1 CN and tumour features, and between PAK1 and CCND1 CNs. Cumulative risk of death from BC and hazard ratios were estimated in analysis of prognosis. RESULTS We found mean PAK1 CN ≥4<6 in 26 (5.1%) tumours, and CN ≥ 6 in 22 (4.3%) tumours. The proportion of cases with copy number increase (mean CN ≥4) was highest among HER2 type and Luminal B (HER2-) tumours. We found an association between PAK1 CN increase, and high proliferation, and high histological grade, but not prognosis. Of cases with PAK1 CN ≥ 6, 30% also had CCND1 CN ≥ 6. CONCLUSIONS PAK1 copy number increase is associated with high proliferation and high histological grade, but not with prognosis. PAK1 CN increase was most frequent in the HER2 type and Luminal B (HER2-) subtype. PAK1 CN increase is associated with CN increase of CCND1.
Collapse
Affiliation(s)
- Anette H Skjervold
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Valla
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Pathology, St. Olav's Hospital, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
8
|
Somanath PR, Chernoff J, Cummings BS, Prasad SM, Homan HD. Targeting P21-Activated Kinase-1 for Metastatic Prostate Cancer. Cancers (Basel) 2023; 15:2236. [PMID: 37190165 PMCID: PMC10137274 DOI: 10.3390/cancers15082236] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic prostate cancer (mPCa) has limited therapeutic options and a high mortality rate. The p21-activated kinase (PAK) family of proteins is important in cell survival, proliferation, and motility in physiology, and pathologies such as infectious, inflammatory, vascular, and neurological diseases as well as cancers. Group-I PAKs (PAK1, PAK2, and PAK3) are involved in the regulation of actin dynamics and thus are integral for cell morphology, adhesion to the extracellular matrix, and cell motility. They also play prominent roles in cell survival and proliferation. These properties make group-I PAKs a potentially important target for cancer therapy. In contrast to normal prostate and prostatic epithelial cells, group-I PAKs are highly expressed in mPCA and PCa tissue. Importantly, the expression of group-I PAKs is proportional to the Gleason score of the patients. While several compounds have been identified that target group-I PAKs and these are active in cells and mice, and while some inhibitors have entered human trials, as of yet, none have been FDA-approved. Probable reasons for this lack of translation include issues related to selectivity, specificity, stability, and efficacy resulting in side effects and/or lack of efficacy. In the current review, we describe the pathophysiology and current treatment guidelines of PCa, present group-I PAKs as a potential druggable target to treat mPCa patients, and discuss the various ATP-competitive and allosteric inhibitors of PAKs. We also discuss the development and testing of a nanotechnology-based therapeutic formulation of group-I PAK inhibitors and its significant potential advantages as a novel, selective, stable, and efficacious mPCa therapeutic over other PCa therapeutics in the pipeline.
Collapse
Affiliation(s)
- Payaningal R. Somanath
- Department of Clinical & Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
- MetasTx LLC, Basking Ridge, NJ 07920, USA
| | - Jonathan Chernoff
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Brian S. Cummings
- MetasTx LLC, Basking Ridge, NJ 07920, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Sandip M. Prasad
- Morristown Medical Center, Atlantic Health System, Morristown, NJ 07960, USA
| | | |
Collapse
|
9
|
Saeki S, Kumegawa K, Takahashi Y, Yang L, Osako T, Yasen M, Otsuji K, Miyata K, Yamakawa K, Suzuka J, Sakimoto Y, Ozaki Y, Takano T, Sano T, Noda T, Ohno S, Yao R, Ueno T, Maruyama R. Transcriptomic intratumor heterogeneity of breast cancer patient-derived organoids may reflect the unique biological features of the tumor of origin. Breast Cancer Res 2023; 25:21. [PMID: 36810117 PMCID: PMC9942352 DOI: 10.1186/s13058-023-01617-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND The intratumor heterogeneity (ITH) of cancer cells plays an important role in breast cancer resistance and recurrence. To develop better therapeutic strategies, it is necessary to understand the molecular mechanisms underlying ITH and their functional significance. Patient-derived organoids (PDOs) have recently been utilized in cancer research. They can also be used to study ITH as cancer cell diversity is thought to be maintained within the organoid line. However, no reports investigated intratumor transcriptomic heterogeneity in organoids derived from patients with breast cancer. This study aimed to investigate transcriptomic ITH in breast cancer PDOs. METHODS We established PDO lines from ten patients with breast cancer and performed single-cell transcriptomic analysis. First, we clustered cancer cells for each PDO using the Seurat package. Then, we defined and compared the cluster-specific gene signature (ClustGS) corresponding to each cell cluster in each PDO. RESULTS Cancer cells were clustered into 3-6 cell populations with distinct cellular states in each PDO line. We identified 38 clusters with ClustGS in 10 PDO lines and used Jaccard similarity index to compare the similarity of these signatures. We found that 29 signatures could be categorized into 7 shared meta-ClustGSs, such as those related to the cell cycle or epithelial-mesenchymal transition, and 9 signatures were unique to single PDO lines. These unique cell populations appeared to represent the characteristics of the original tumors derived from patients. CONCLUSIONS We confirmed the existence of transcriptomic ITH in breast cancer PDOs. Some cellular states were commonly observed in multiple PDOs, whereas others were specific to single PDO lines. The combination of these shared and unique cellular states formed the ITH of each PDO.
Collapse
Affiliation(s)
- Sumito Saeki
- grid.410807.a0000 0001 0037 4131Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550 Japan ,grid.410807.a0000 0001 0037 4131Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kohei Kumegawa
- grid.410807.a0000 0001 0037 4131Cancer Cell Diversity Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoko Takahashi
- grid.410807.a0000 0001 0037 4131Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Liying Yang
- grid.410807.a0000 0001 0037 4131Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550 Japan
| | - Tomo Osako
- grid.410807.a0000 0001 0037 4131Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mahmut Yasen
- grid.410807.a0000 0001 0037 4131Cancer Informatics and Biobanking Platform Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazutaka Otsuji
- grid.410807.a0000 0001 0037 4131Cancer Cell Diversity Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kenichi Miyata
- grid.410807.a0000 0001 0037 4131Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550 Japan
| | - Kaoru Yamakawa
- grid.410807.a0000 0001 0037 4131Cancer Informatics and Biobanking Platform Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Jun Suzuka
- grid.410807.a0000 0001 0037 4131Cancer Cell Diversity Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuri Sakimoto
- grid.410807.a0000 0001 0037 4131Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550 Japan
| | - Yukinori Ozaki
- grid.410807.a0000 0001 0037 4131Breast Medical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshimi Takano
- grid.410807.a0000 0001 0037 4131Breast Medical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Sano
- grid.410807.a0000 0001 0037 4131Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuo Noda
- grid.410807.a0000 0001 0037 4131Director’s Room, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shinji Ohno
- grid.410807.a0000 0001 0037 4131Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryoji Yao
- grid.410807.a0000 0001 0037 4131Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Ueno
- grid.410807.a0000 0001 0037 4131Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Reo Maruyama
- Project for Cancer Epigenomics, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan. .,Cancer Cell Diversity Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
10
|
Soong TR, Dillon DA, Rice-Stitt TL, Wieczorek TJ, Baker GM, Darvishian F, Collins LC, Lester SC, Schnitt SJ, Harrison BT. Invasive lobular carcinoma with extracellular mucin (ILCEM): clinicopathologic and molecular characterization of a rare entity. Mod Pathol 2022; 35:1370-1382. [PMID: 35477749 DOI: 10.1038/s41379-022-01084-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
Abstract
Invasive lobular carcinoma with extracellular mucin (ILCEM) is a rare histologic subtype of breast cancer. Little is known about the pathologic or genomic signatures that distinguish ILCEM from classic invasive lobular carcinoma (ILC) or mucinous carcinoma. We studied 17 breast cancers with lobular morphology and extracellular mucin. Thirteen tumors with sufficient tissue for DNA extraction were analyzed by a next generation sequencing (NGS) assay that interrogates 447 genes for mutations and copy number variations (CNVs). Median patient age was 66 yrs (range: 31-77 yrs). Sixteen patients presented with masses, 7 of which were >2 cm. Seven patients had lymph node metastases. The cases of ILCEM were moderately (n = 13) or poorly differentiated (n = 4), frequently exhibiting variant morphology that has not been previously described or emphasized, including grade 3 nuclei (n = 11), diffuse signet ring cells (n = 10), solid growth (n = 4), tumor necrosis (n = 3) or apocrine features (n = 2). All tumors showed absent or reduced membranous E-cadherin expression. Concurrent lobular carcinoma in situ (LCIS) was seen in 11/17 cases, 1 of which was a striking example of signet ring cell LCIS with extracellular mucin. Receptor profiles were ER+/HER2- (n = 15) and ER+/HER2+ (n = 2). With a median follow-up of 83.5 months (range: 3-171 months) in 12 patients with available information, 8 patients had recurrences resulting in 4 cancer-related deaths. The most common CNVs were 16q loss (n = 11) and 1q gain (n = 9). CDH1 gene-level alterations were detected in all but one case, including frameshift (n = 7), nonsense (n = 2), and donor splice site (n = 1) mutations and indels (n = 2). Recurrent mutations were also seen in PIK3CA (n = 3), POLQ (n = 3), TP53 (n = 3), ERBB3 (n = 3), ERBB2 (n = 2), and RUNX1 (n = 2). Genes with recurrent amplifications included GATA3 (n = 4), FOXA1 (n = 3), CCND1 (n = 2). Our data highlights ILCEM as a distinct variant of ILC that often presents with higher-grade and variant morphologic features and is associated with an aggressive clinical course. NGS data support an overall lobular-type molecular profile and reveal potentially targetable alterations in a subset of cases with recurrence.
Collapse
Affiliation(s)
- T Rinda Soong
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Tad John Wieczorek
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Pathology, Brigham and Women's Faulkner Hospital, Boston, MA, USA
| | - Gabrielle M Baker
- Harvard Medical School, Boston, MA, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Farbod Darvishian
- Department of Pathology, New York University Langone Medical Center, New York, NY, USA
| | - Laura C Collins
- Harvard Medical School, Boston, MA, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Susan C Lester
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Stuart J Schnitt
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Beth T Harrison
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Lei K, Luo M, Tu Z, Lv S, Liu J, Gong C, Ye M, Wu M, Sheng Y, Long X, Li J, Zhu X, Huang K. Comprehensive analysis of the prognostic implications and functional exploration of PAK gene family in human cancer. Cancer Cell Int 2022; 22:275. [PMID: 36064705 PMCID: PMC9442929 DOI: 10.1186/s12935-022-02689-6] [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: 05/05/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
Background The p21-activated kinase (PAK) family (PAKs) plays a key role in the formation and development of human tumors. However, a systematic analysis of PAKs in human cancers is lacking and the potential role of PAKs in cancer immunity has not been explored. Methods We used datasets from in The Cancer Genome Atlas (TCGA) database and Genotype-Tissue Expression database (GTEx). Results Based on TCGA datasets most PAKs show noteworthy differences in expression between tumors and corresponding normal tissues or across different tumor tissues. Patients with high expression of PAKs often show a worse prognosis. However, copy number variation, mutation, and DNA methylation of PAKs have limited impact on tumor development. Further analysis showed that the impact of PAKs on immunity varies with the type of tumor and the respective tumor microenvironment. PAK1 and PAK4 may be stronger predictors of immune characteristics, and are more suitable as drugs and molecular therapeutic targets. Furthermore, Cox regression analysis revealed that a PAK gene signature could be used as an independent prognostic factor for lower grade glioma (LGG) and glioblastoma (GBM). Gene set enrichment analysis (GSEA) analysis indicated that PAK genes may affect the occurrence and development of GBM through the PI3K signaling pathway. Further experiments verified that PAK1 and AKT1 have a significant interaction in GBM cells, and inhibiting the overactivation of PAK1 can significantly inhibit the proliferation of GBM cells. Conclusions Our study provides a rationale for further research on the prognostic and therapeutic potential of PAKs in human tumors. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02689-6.
Collapse
Affiliation(s)
- Kunjian Lei
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Min Luo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao, 334000, Jiangxi, People's Republic of China
| | - Zewei Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao, 334000, Jiangxi, People's Republic of China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Junzhe Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Chuandong Gong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Minhua Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Miaojing Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yilei Sheng
- Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xiaoyan Long
- East China Institute of Digital Medical Engineering, Shangrao, 334000, Jiangxi, People's Republic of China
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China. .,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China. .,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
12
|
Jacobs AT, Martinez Castaneda-Cruz D, Rose MM, Connelly L. Targeted therapy for breast cancer: An overview of drug classes and outcomes. Biochem Pharmacol 2022; 204:115209. [PMID: 35973582 DOI: 10.1016/j.bcp.2022.115209] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 12/20/2022]
Abstract
The last 25 years have seen significant growth in new therapeutic options for breast cancer, termed targeted therapies based on their ability to block specific pathways known to drive breast tumor growth and survival. Introduction of these drugs has been made possible through advances in the understanding of breast cancer biology. While the promise of targeted therapy for breast cancer has been clear for some time, the experience of the clinical use of multiple drugs and drug classes allows us to now present a summary and perspective as to the success and impact of this endeavor. Here we will review breast cancer targeted therapeutics in clinical use. We will provide the rationale for their indications and summarize clinical data in patients with different breast cancer subtypes, their impact on breast cancer progression and survival and their major adverse effects. The focus of this review will be on the development that has occurred within classes of targeted therapies and subsequent impact on breast cancer patient outcomes. We will conclude with a perspective on the role of targeted therapy in breast cancer treatment and highlight future areas of development.
Collapse
Affiliation(s)
- Aaron T Jacobs
- California University of Science and Medicine, 1501 Violet Street, Colton, CA 92324, United States
| | | | - Mark M Rose
- California University of Science and Medicine, 1501 Violet Street, Colton, CA 92324, United States
| | - Linda Connelly
- California University of Science and Medicine, 1501 Violet Street, Colton, CA 92324, United States.
| |
Collapse
|
13
|
Dissecting Molecular Heterogeneity of Circulating Tumor Cells (CTCs) from Metastatic Breast Cancer Patients through Copy Number Aberration (CNA) and Single Nucleotide Variant (SNV) Single Cell Analysis. Cancers (Basel) 2022; 14:cancers14163925. [PMID: 36010918 PMCID: PMC9405921 DOI: 10.3390/cancers14163925] [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: 07/11/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 12/27/2022] Open
Abstract
Circulating tumor cells' (CTCs) heterogeneity contributes to counteract their introduction in clinical practice. Through single-cell sequencing we aim at exploring CTC heterogeneity in metastatic breast cancer (MBC) patients. Single CTCs were isolated using DEPArray NxT. After whole genome amplification, libraries were prepared for copy number aberration (CNA) and single nucleotide variant (SNV) analysis and sequenced using Ion GeneStudio S5 and Illumina MiSeq, respectively. CTCs demonstrate distinctive mutational signatures but retain molecular traces of their common origin. CNA profiling identifies frequent aberrations involving critical genes in pathogenesis: gains of 1q (CCND1) and 11q (WNT3A), loss of 22q (CHEK2). The longitudinal single-CTC analysis allows tracking of clonal selection and the emergence of resistance-associated aberrations, such as gain of a region in 12q (CDK4). A group composed of CTCs from different patients sharing common traits emerges. Further analyses identify losses of 15q and enrichment of terms associated with pseudopodium formation as frequent and exclusive events. CTCs from MBC patients are heterogeneous, especially concerning their mutational status. The single-cell analysis allows the identification of aberrations associated with resistance, and is a candidate tool to better address treatment strategy. The translational significance of the group populated by similar CTCs should be elucidated.
Collapse
|
14
|
Comprehensive characterization of pre- and post-treatment samples of breast cancer reveal potential mechanisms of chemotherapy resistance. NPJ Breast Cancer 2022; 8:60. [PMID: 35523804 PMCID: PMC9076915 DOI: 10.1038/s41523-022-00428-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/12/2022] [Indexed: 12/23/2022] Open
Abstract
When locally advanced breast cancer is treated with neoadjuvant chemotherapy, the recurrence risk is significantly higher if no complete pathologic response is achieved. Identification of the underlying resistance mechanisms is essential to select treatments with maximal efficacy and minimal toxicity. Here we employed gene expression profiles derived from 317 HER2-negative treatment-naïve breast cancer biopsies of patients who underwent neoadjuvant chemotherapy, deep whole exome, and RNA-sequencing profiles of 22 matched pre- and post-treatment tumors, and treatment outcome data to identify biomarkers of response and resistance mechanisms. Molecular profiling of treatment-naïve breast cancer samples revealed that expression levels of proliferation, immune response, and extracellular matrix (ECM) organization combined predict response to chemotherapy. Triple negative patients with high proliferation, high immune response and low ECM expression had a significantly better treatment response and survival benefit (HR 0.29, 95% CI 0.10–0.85; p = 0.02), while in ER+ patients the opposite was seen (HR 4.73, 95% CI 1.51–14.8; p = 0.008). The characterization of paired pre-and post-treatment samples revealed that aberrations of known cancer genes were either only present in the pre-treatment sample (CDKN1B) or in the post-treatment sample (TP53, APC, CTNNB1). Proliferation-associated genes were frequently down-regulated in post-treatment ER+ tumors, but not in triple negative tumors. Genes involved in ECM were upregulated in the majority of post-chemotherapy samples. Genomic and transcriptomic differences between pre- and post-chemotherapy samples are common and may reveal potential mechanisms of therapy resistance. Our results show a wide range of distinct, but related mechanisms, with a prominent role for proliferation- and ECM-related genes.
Collapse
|
15
|
Zhou R, Zhu X, Peng Y, Zhong L, Peng L, Yang B, Meng Y, Chen X, Lu Y. Clinical Impact of 11q13.3 Amplification on Immune Cell Infiltration and Prognosis in Breast Cancer. Int J Gen Med 2022; 15:4037-4052. [PMID: 35444456 PMCID: PMC9014960 DOI: 10.2147/ijgm.s360177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Amplification of the 11q13.3 locus has been observed in various tumors. This study sought to determine the correlation of gene amplification at the 11q13.3 locus with the immune status and survival of breast cancer. Methods Amplification of the 11q13.3 locus was characterized by analyzing a publicly available database from the cBioPortal platform (TCGA). The correlation of amplified genes with immune cell infiltration in breast cancer was further analyzed using the TIMER2.0 platform. Immunohistochemical staining was used to determine the expression levels of Cyclin D1 (CCND1), Fas-associated death domain (FADD) and P53 in 156 clinical breast cancer samples. Results This study revealed that amplification of the 11q13.3 amplicon in breast cancer is likely more frequently detected in luminal B breast cancer. Moreover, high expression or amplification of CCND1, fibroblast growth factor 3 (FGF3), fibroblast growth factor 4 (FGF4), fibroblast growth factor 19 (FGF19) and FADD was inversely correlated with the abundance of CD4+ T cells and dendritic cell infiltration in breast cancer (P < 0.05). Data analysis also demonstrated that high expression of CCND1, FGF4 and FADD mRNA levels was closely correlated with shorter recurrence-free survival (RFS) in patients with breast cancer (P < 0.05). The results of immunohistochemical staining from clinical samples further confirmed that high expression of CCND1 and FADD was frequently detected in luminal B and high-grade breast cancer with shorter metastasis-free survival times (P < 0.05). Conclusion This study demonstrated that coamplification of genes located on the 11q13.3 amplicon is frequently detected in luminal B subtype breast cancer and is closely associated with worse survival in patients with breast cancer. Moreover, coamplification of the CCND1-FGF locus might decrease antitumor immune activity in breast cancer, indicating that coamplification of the 11q13.3 amplicon is likely to be a key determinant of therapeutic resistance and accelerate the aggressive evolution of breast cancer.
Collapse
Affiliation(s)
- Renyu Zhou
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Xiaoxi Zhu
- Department of Oncology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People's Republic of China
| | - Yulong Peng
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Lijuan Zhong
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Lilin Peng
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Bo Yang
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Yuhua Meng
- Department of Pathology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, 528300, People’s Republic of China
| | - Xuanzhao Chen
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Yuanzhi Lu
- Department of Pathology, First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Correspondence: Yuanzhi Lu, Tel +86-20 38688984, Email
| |
Collapse
|
16
|
Valla M, Klæstad E, Ytterhus B, Bofin AM. CCND1 Amplification in Breast Cancer -associations With Proliferation, Histopathological Grade, Molecular Subtype and Prognosis. J Mammary Gland Biol Neoplasia 2022; 27:67-77. [PMID: 35459982 PMCID: PMC9135839 DOI: 10.1007/s10911-022-09516-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
CCND1 is located on 11q13. Increased CCND1 copy number (CN) in breast cancer (BC) is associated with high histopathological grade, high proliferation, and Luminal B subtype. In this study of CCND1 in primary BCs and corresponding axillary lymph node metastases (LNM),we examine associations between CCND1 CN in primary BCs and proliferation status, molecular subtype, and prognosis. Furthermore, we studied associations between CCND1 CN and CNs of FGFR1 and ZNF703, both of which are located on 8p12. Fluorescence in situ hybridization probes for CCND1 and chromosome 11 centromere were used on tissue microarrays comprising 526 BCs and 123 LNM. We assessed associations between CCND1 CN and tumour characteristics using Pearson's χ2 test, and estimated cumulative risks of death from BC and hazard ratios in analysis of prognosis. We found CCND1 CN ≥ 4 < 6 in 45 (8.6%) tumours, and ≥ 6 in 42 (8.0%). CCND1 CN (≥ 6) was seen in all molecular subtypes, most frequently in Luminal B (HER2-) (20/126; 16%). Increased CCND1 CN was associated with high histopathological grade, high Ki-67, and high mitotic count, but not prognosis. CCND1 CN ≥ 6 was accompanied by CN increase of FGFR1 in 6/40 cases (15.0%) and ZNF703 in 5/38 cases (13.2%). Three cases showed CN increase of all three genes. High CCND1 CN was most frequent in Luminal B (HER2-) tumours. Good correlation between CCND1 CNs in BCs and LNM was observed. Despite associations between high CCND1 CN and aggressive tumour characteristics, the prognostic impact of CCND1 CN remains unresolved.
Collapse
Affiliation(s)
- Marit Valla
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's Hospital, Trondheim University Hospital, 7006, Trondheim, Norway
| | - Elise Klæstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Borgny Ytterhus
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna M Bofin
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
17
|
Jeffreys SA, Becker TM, Khan S, Soon P, Neubauer H, de Souza P, Powter B. Prognostic and Predictive Value of CCND1/Cyclin D1 Amplification in Breast Cancer With a Focus on Postmenopausal Patients: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2022; 13:895729. [PMID: 35784572 PMCID: PMC9249016 DOI: 10.3389/fendo.2022.895729] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/10/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Up to 80% of breast cancers (BCa) are estrogen receptor positive and current treatments target the estrogen receptor (endocrine therapies) and/or CDK4/6 (CDK4/6 inhibitors). CCND1 encodes the protein cyclin D1, responsible for regulation of G1 to S phase transition in the cell cycle. CCND1 amplification is common in BCa and contributes to increased cyclin D1 expression. As there are signalling interactions between cyclin D1 and the estrogen receptor, understanding the impact of CCND1 amplification on estrogen receptor positive patients' disease outcomes, is vital. This review aims to evaluate CCND1 amplification as a prognostic and predictive biomarker in BCa. MATERIALS AND METHODS Publications were retrieved from the databases: PubMed, MEDLINE, Embase and Cochrane library. Exclusion criteria were duplication, publication type, non-English language, in vitro and animal studies, not BCa, male BCa, premenopausal BCa, cohort size <35, CCND1 amplification not reported. Publications with cohort duplication, and inadequate recurrence free survival (RFS) and overall survival (OS) data, were also excluded. Included publications were assessed for Risk of Bias (RoB) using the Quality In Prognosis Studies tool. Statistical analyses (Inverse Variance and Mantel-Haenszel) were performed in Review Manager. The PROSPERO registration number is [CRD42020208179]. RESULTS CCND1 amplification was significantly associated with positive estrogen receptor status (OR:1.70, 95% CI:1.19-2.43, p = 0.004) and cyclin D1 overexpression (OR: 5.64, 95% CI: 2.32-13.74, p=0.0001). CCND1 amplification was significantly associated with shorter RFS (OR: 1.64, 95% CI: 1.13-2.38, p = 0.009), and OS (OR: 1.51, 95% CI: 1.19-1.92, p = 0.0008) after removal of studies with a high RoB. In endocrine therapy treated patients specifically, CCND1 amplification predicted shorter RFS (HR: 2.59, 95% CI: 1.96-3.41, p < 0.00001) and OS (HR: 1.59, 95% CI: 1.00-2.49, p = 0.05) also after removal of studies with a high RoB. CONCLUSION While a lack of standardised approach for the detection of CCND1 amplification is to be considered as a limitation, CCND1 amplification was found to be prognostic of shorter RFS and OS in BCa. CCND1 amplification is also predictive of reduced RFS and OS in endocrine therapy treated patients specifically. With standardised methods and cut offs for the detection of CCND1 amplification, CCND1 amplification would have potential as a predictive biomarker in breast cancer patients. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/, identifier CRD42020208179.
Collapse
Affiliation(s)
- Sarah A. Jeffreys
- Centre of Circulating Tumour Cell Diagnostics and Research, Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- *Correspondence: Sarah A. Jeffreys,
| | - Therese M. Becker
- Centre of Circulating Tumour Cell Diagnostics and Research, Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| | - Sarah Khan
- Department of Medical Oncology, Bankstown Cancer Centre, Bankstown, NSW, Australia
| | - Patsy Soon
- Centre of Circulating Tumour Cell Diagnostics and Research, Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
- Department of Surgery, Bankstown Hospital, Bankstown, NSW, Australia
| | - Hans Neubauer
- Department of Obstetrics and Gynaecology, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Paul de Souza
- Centre of Circulating Tumour Cell Diagnostics and Research, Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| | - Branka Powter
- Centre of Circulating Tumour Cell Diagnostics and Research, Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
| |
Collapse
|
18
|
Rajendran S, Swaroop SS, Roy J, Inemai E, Murugan S, Rayala SK, Venkatraman G. p21 activated kinase-1 and tamoxifen - A deadly nexus impacting breast cancer outcomes. Biochim Biophys Acta Rev Cancer 2021; 1877:188668. [PMID: 34896436 DOI: 10.1016/j.bbcan.2021.188668] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022]
Abstract
Tamoxifen is a commonly used drug in the treatment of ER + ve breast cancers since 1970. However, development of resistance towards tamoxifen limits its remarkable clinical success. In this review, we have attempted to provide a brief overview of multiple mechanism that may lead to tamoxifen resistance, with a special emphasis on the roles played by the oncogenic kinase- PAK1. Analysing the genomic data sets available in the cBioPortal, we found that PAK1 gene amplification significantly affects the Relapse Free Survival of the ER + ve breast cancer patients. While PAK1 is known to promote tamoxifen resistance by phosphorylating ERα at Ser305, existing literature suggests that PAK1 can fuel up tamoxifen resistance obliquely by phosphorylating other substrates. We have summarised some of the approaches in the mass spectrometry based proteomics, which would enable us to study the tamoxifen resistance specific phosphoproteomic landscape of PAK1. We also propose that elucidating the multiple mechanisms by which PAK1 promotes tamoxifen resistance might help us discover druggable targets and biomarkers.
Collapse
Affiliation(s)
- Swetha Rajendran
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Srikanth Swamy Swaroop
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Joydeep Roy
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, India
| | - Ezhil Inemai
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, India
| | - Sowmiya Murugan
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, India
| | - Suresh K Rayala
- Department of Biotechnology, Indian Institute of Technology, Madras, Chennai, India.
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| |
Collapse
|
19
|
Jacob S, Davis AA, Gerratana L, Velimirovic M, Shah AN, Wehbe F, Katam N, Zhang Q, Flaum L, Siziopikou KP, Platanias LC, Gradishar WJ, Behdad A, Bardia A, Cristofanilli M. The Use of Serial Circulating Tumor DNA to Detect Resistance Alterations in Progressive Metastatic Breast Cancer. Clin Cancer Res 2020; 27:1361-1370. [PMID: 33323406 DOI: 10.1158/1078-0432.ccr-20-1566] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/18/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Circulating tumor DNA (ctDNA) is a promising tool for noninvasive longitudinal monitoring of genomic alterations. We analyzed serial ctDNA to characterize genomic evolution in progressive metastatic breast cancer. EXPERIMENTAL DESIGN This was a retrospective cohort between 2015 and 2019 obtained under an Institutional Review Board-approved protocol at Northwestern University (Chicago, IL). ctDNA samples were analyzed with Guardant360 next-generation sequencing (NGS) assay. A total of 86 patients had at least two serial ctDNA collections with the second drawn at first post-NGS progression (PN1) by imaging and clinical assessment. A total of 27 participants had ctDNA drawn at second post-NGS clinical progression (PN2). We analyzed alterations, mutant allele frequency (MAF), number of alterations (NOA), and sites of disease on imaging in close proximity to ctDNA evaluation. Matched pairs' variations in MAF, NOA, and alterations at progression were tested through Wilcoxon test. We identified an independent control cohort at Massachusetts General Hospital (Boston, MA) of 63 patients with serial ctDNA sampling and no evidence of progression. RESULTS We identified 44 hormone receptor-positive, 20 HER2+, and 22 triple-negative breast cancer cases. The significant alterations observed between baseline and PN1 were TP53 (P < 0.0075), PIK3CA (P < 0.0126), AR (P < 0.0126), FGFR1 (P < 0.0455), and ESR1 (P < 0.0143). Paired analyses revealed increased MAF and NOA from baseline to PN1 (P = 0.0026, and P < 0.0001, respectively). When compared with controls without progression, patients with ctDNA collection at times of progression were associated with increased MAF and NOA (P = 0.0042 and P < 0.0001, respectively). CONCLUSIONS Serial ctDNA testing identified resistance alterations and increased NOA and MAF were associated with disease progression. Prospective longitudinal ctDNA evaluation could potentially monitor tumor genomic evolution.
Collapse
Affiliation(s)
- Saya Jacob
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Andrew A Davis
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Division of Hematology and Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Lorenzo Gerratana
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Medicine, University of Udine, Udine, Italy
| | | | - Ami N Shah
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Firas Wehbe
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Neelima Katam
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Qiang Zhang
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Lisa Flaum
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Kalliopi P Siziopikou
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Pathology, Northwestern University, Chicago, Illinois
| | - Leonidas C Platanias
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - William J Gradishar
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Amir Behdad
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois.,Department of Pathology, Northwestern University, Chicago, Illinois
| | - Aditya Bardia
- Massachusetts General Hospital, Boston, Massachusetts
| | - Massimo Cristofanilli
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| |
Collapse
|
20
|
Liposomes Targeting P21 Activated Kinase-1 (PAK-1) and Selective for Secretory Phospholipase A 2 (sPLA 2) Decrease Cell Viability and Induce Apoptosis in Metastatic Triple-Negative Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21249396. [PMID: 33321758 PMCID: PMC7764208 DOI: 10.3390/ijms21249396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
P21 activated kinases (or group I PAKs) are serine/threonine kinases whose expression is altered in prostate and breast cancers. PAK-1 activity is inhibited by the small molecule "Inhibitor targeting PAK-1 activation-3" (IPA-3), which has selectivity for PAK-1 but is metabolically unstable. Secretory Group IIA phospholipase A2 (sPLA2) expression correlates to increased metastasis and decreased survival in many cancers. We previously designed novel liposomal formulations targeting both PAK-1 and sPLA2, called Secretory Phospholipase Responsive liposomes or SPRL-IPA-3, and demonstrated their ability to alter prostate cancer growth. The efficacy of SPRL against other types of cancers is not well understood. We addressed this limitation by determining the ability of SPRL to induce cell death in a diverse panel of cells representing different stages of breast cancer, including the invasive but non-metastatic MCF-7 cells, and metastatic triple-negative breast cancer (TNBC) cells such as MDA-MB-231, MDA-MB-468, and MDA-MB-435. We investigated the role of sPLA2 in the disposition of these liposomes by comparing the efficacy of SPRL-IPA-3 to IPA-3 encapsulated in sterically stabilized liposomes (SSL-IPA-3), a formulation shown to be less sensitive to sPLA2. Both SSL-IPA-3 and SPRL-IPA-3 induced time- and dose-dependent decreases in MTT staining in all cell lines tested, but SPRL-IPA-3-induced effects in metastatic TNBC cell lines were superior over SSL-IPA-3. The reduction in MTT staining induced by SPRL-IPA-3 correlated to the expression of Group IIA sPLA2. sPLA2 expression also correlated to increased induction of apoptosis in TNBC cell lines by SPRL-IPA-3. These data suggest that SPRL-IPA-3 is selective for metastatic TNBC cells and that the efficacy of SPRL-IPA-3 is mediated, in part, by the expression of Group IIA sPLA2.
Collapse
|
21
|
PAK1 as a Potential Therapeutic Target in Male Smokers with EGFR-Mutant Non-Small Cell Lung Cancer. Molecules 2020; 25:molecules25235588. [PMID: 33261184 PMCID: PMC7729550 DOI: 10.3390/molecules25235588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022] Open
Abstract
P21-activated kinases (PAKs) are serine/threonine protein kinases that contribute to several cellular processes. Here, we aimed to determine the prognostic value of PAK1 and its correlation with the clinicopathological characteristics and five-year survival rates in patients with non-small cell lung cancer (NSCLC). We evaluated PAK1 mRNA and protein expression in NSCLC cells and resected tumor specimens, as well as in healthy human bronchial epithelial cells and adjacent healthy lung tissues, respectively, for effective comparison. Immunohistochemical tissue microarray analysis of 201 NSCLC specimens showed the correlation of PAK1 expression with clinicopathological characteristics. The mRNA and protein expression of PAK1 were 2.9- and 4.3-fold higher in six of seven NSCLC cell types and human tumors (both, p < 0.001) than in healthy human bronchial epithelial BEAS-2B cells and adjacent healthy lung tissues, respectively. Decreased survival was significantly associated with PAK1 overexpression in the entire cohort (χ2 = 8.48, p = 0.0036), men (χ2 = 17.1, p < 0.0001), and current and former smokers (χ2 = 19.2, p < 0.0001). Notably, epidermal growth factor receptor (EGFR) mutation-positive lung cancer patients with high PAK1 expression showed higher mortality rates than those with low PAK1 expression (91.3% vs. 62.5%, p = 0.02). Therefore, PAK1 overexpression could serve as a molecular target for the treatment of EGFR mutation-positive lung cancer, especially among male patients and current/former smokers.
Collapse
|
22
|
Aleksakhina SN, Kramchaninov MM, Mikushina AD, Kubrina SE, Petkau VV, Ivantsov AO, Moiseyenko VM, Imyanitov EN, Iyevleva AG. CCND1 and FGFR1 gene amplifications are associated with reduced benefit from aromatase inhibitors in metastatic breast cancer. Clin Transl Oncol 2020; 23:874-881. [PMID: 32880048 DOI: 10.1007/s12094-020-02481-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Endocrine therapy is a mainstay for the treatment of hormone receptor-positive breast cancer (BC); however, only a fraction of patients experience a pronounced response to antagonists of estrogen signaling. There is a need to identify predictors for efficacy of this treatment. METHODS This study included 138 patients with newly diagnosed metastatic BC, who received upfront endocrine therapy. Archival biopsy specimens were tested for CCND1 and FGFR1 gene amplification and mRNA expression by PCR-based methods. RESULTS CCND1 and FGFR1 amplification was detected in 24 (17.9%) and 28 (20.9%) of 134 evaluable cases, respectively; 9 carcinomas had concurrent alterations of these two genes. Presence of amplification in at least one locus was more common in tumors of higher grade (p = 0.018) and was associated with higher Ki-67 proliferation index (p = 0.036). CCND1 gene amplification was associated with shorter progression-free survival (PFS) in patients receiving aromatase inhibitors (AI) [16.0 months vs. 32.4 months, HR = 3.16 (95% CI 1.26-7.93), p = 0.014]. FGFR1 status did not significantly affect PFS of AI-treated women; however, objective response to AI was observed less frequently in FGFR1-amplified BC as compared to cases with normal FGFR1 copy number [2/15 (13.3%) vs. 22/46 (47.8%), p = 0.031]. Meanwhile, CCND1/FGFR1 gene status did not influence the outcome of tamoxifen-treated patients. CONCLUSION Presence of CCND1 and/or FGFR1 amplification is associated with worse outcomes of AI therapy in patients with metastatic BC.
Collapse
Affiliation(s)
- S N Aleksakhina
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, Saint-Petersburg, Russia, 197758
| | | | - A D Mikushina
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, Saint-Petersburg, Russia, 197758
| | - S E Kubrina
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, Saint-Petersburg, Russia, 197758
| | - V V Petkau
- Sverdlovskiy Regional Oncological Hospital, Ekatherinburg, Russia, 620036
| | - A O Ivantsov
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, Saint-Petersburg, Russia, 197758
| | | | - E N Imyanitov
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, Saint-Petersburg, Russia, 197758.,Saint-Petersburg Pediatric Medical University, Saint-Petersburg, Russia, 194100.,I.I. Mechnikov North-Western Medical University, Saint-Petersburg, Russia, 191015
| | - A G Iyevleva
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, Saint-Petersburg, Russia, 197758. .,Saint-Petersburg Pediatric Medical University, Saint-Petersburg, Russia, 194100.
| |
Collapse
|
23
|
The CXCR4-Dependent LASP1-Ago2 Interaction in Triple-Negative Breast Cancer. Cancers (Basel) 2020; 12:cancers12092455. [PMID: 32872485 PMCID: PMC7564666 DOI: 10.3390/cancers12092455] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 12/28/2022] Open
Abstract
The CXCR4-LASP1 axis is an emerging target in the field of breast cancer metastasis. C-X-C chemokine receptor type 4 (CXCR4) mediates directed cell migration when activated by its cognate ligand CXCL12. LIM and SH3 Protein 1 (LASP1) is a critical node in the CXCR4 signaling pathway, as its deficiency blocks CXCR4-dependent Matrigel invasion. The mechanism by which LASP1 facilitates this invasive ability of tumor cells when CXCR4 is activated is unknown. Our previous proteomics work had revealed several components of the RNA interference (RNAi) machinery as being potential LASP1 interacting proteins. Here we report that argonaute 2 (Ago2), a protein with central involvement in RNAi, associates with LASP1 in triple-negative breast cancer (TNBC) cells. We demonstrate that LASP1 co-immunoprecipitates with Ago2 endogenously in a CXCL12-dependent manner, with further confirmation of this interaction by proximity ligation assay. Furthermore, this association is specific to CXCR4 as it can be abrogated by the CXCR4 antagonist, AMD3465. By GST-pulldown approach, we identify that LASP1 directly binds to Ago2 through its LIM and SH3 domains, and that this binding is dictated by the S146 and Y171 phosphorylation sites of LASP1. Additionally, the phosphorylation status of LASP1 affected tumor suppressor microRNA (miRNA) Let-7a-guided Ago2 activity. Levels of several endogenous targets of Let-7a were found to be altered including C-C chemokine receptor type 7 (CCR7), which is another critical chemokine receptor involved in metastasis to lymph nodes. Our results suggest a novel role for the LASP1-Ago2 module in shaping the RNAi landscape, functionally impacting the invasive ability of cancer cells.
Collapse
|
24
|
Zhao CC, Zhan MN, Liu WT, Jiao Y, Zhang YY, Lei Y, Zhang TT, Zhang CJ, Du YY, Gu KS, Wei W. Combined LIM kinase 1 and p21-Activated kinase 4 inhibitor treatment exhibits potent preclinical antitumor efficacy in breast cancer. Cancer Lett 2020; 493:120-127. [PMID: 32829006 DOI: 10.1016/j.canlet.2020.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/27/2020] [Accepted: 08/02/2020] [Indexed: 12/20/2022]
Abstract
LIM kinase 1 (LIMK1) and p21-activated kinase 4 (PAK4) are often over-expressed in breast tumors, which causes aggressive cancer phenotypes and unfavorable clinical outcomes. In addition to the well-defined role in regulating cell division, proliferation and invasion, the two kinases promote activation of the MAPK pathway and cause endocrine resistance through phosphorylating estrogen receptor alpha (ERα). PAK4 specifically phosphorylates LIMK1 and its functional partners, indicating possible value of suppressing both kinases in cancers that over-express PAK4 and/or LIMK1. Here, for the first time, we assessed the impact of combining LIMK1 inhibitor LIMKi 3 and PAK4 inhibitor PF-3758309 in preclinical breast cancer models. LIMK1 and PAK4 pharmacological inhibition synergistically reduced the survival of various cancer cell lines, exhibiting specific efficacy in luminal and HER2-enriched models, and suppressed development and ERα-driven signals in a BT474 xenograft model. In silico analysis demonstrated the cell lines with reliance on LIMK1 were the most prone to be susceptible to PAK4 inhibition. Double LIMK1 and PAK4 targeting therapy can be a successful therapeutic strategy for breast cancer, with a unique efficiency in the subtypes of luminal and HER2-enriched tumors.
Collapse
Affiliation(s)
- Chen-Chen Zhao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Meng-Na Zhan
- Department of Pathology, Zhong-Shan Hospital Affiliated to Fudan University, Shanghai, 200023, China
| | - Wan-Ting Liu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Yang Jiao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Yi-Yin Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Yu Lei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Teng-Teng Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Cong-Jun Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Ying-Ying Du
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Kang-Sheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China.
| | - Wei Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China.
| |
Collapse
|
25
|
Kanumuri R, Saravanan R, Pavithra V, Sundaram S, Rayala SK, Venkatraman G. Current trends and opportunities in targeting p21 activated kinase-1(PAK1) for therapeutic management of breast cancers. Gene 2020; 760:144991. [PMID: 32717309 DOI: 10.1016/j.gene.2020.144991] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most frequently diagnosed cancer in women worldwide. Identifying reliable biomarkers and druggable molecular targets pose to be a significant quest in breast cancer research. p21-activated kinase 1 (PAK1) is a serine/threonine kinase that direct cell motility, cytoskeletal remodelling, and has been shown to function as a downstream regulator for various cancer signalling cascades that promote cell proliferation, apoptosis deregulation and hasten mitotic abnormalities, resulting in tumor formation and progression. The heterogeneity and acquired drug resistance are important factors that challenge the treatment of breast cancer. p21-activated kinase 1 signalling is crucial for activation of the Ras/RAF/MEK/ERK, PI3K/Akt/mTOR and Wnt signalling cascades which regulate cell survival, cell cycle progression, differentiation, and proliferation. A study involving proteogenomics analysis on breast cancer tissues showed the PAK1 as outlier kinase. In addition to this, few outlier molecules were identified specific to subtypes of breast cancer. A few substrates of PAK1 in breast cancer are already known. In this paper, we have discussed a similar approach called Kinase Interacting Substrate Screening (KISS) for the identification of novel oncogenic substrates of p21-activated kinase specific to subtypes of breast cancer. Such high throughput approaches are expected to accelerate the process of identifying novel drug targets and biomarkers.
Collapse
Affiliation(s)
- Rahul Kanumuri
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India; Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Roshni Saravanan
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - V Pavithra
- Department of Pathology, Sri Ramachandra Medical College & Research Institute, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Medical College & Research Institute, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India
| | - Suresh K Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| | - Ganesh Venkatraman
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education & Research (Deemed to be University), Porur, Chennai, Tamilnadu, India.
| |
Collapse
|
26
|
Zhu X, Feng J, Fu W, Shu X, Wan X, Liu J. Effects of cisplatin on the proliferation, invasion and apoptosis of breast cancer cells following β‑catenin silencing. Int J Mol Med 2020; 45:1838-1850. [PMID: 32186756 PMCID: PMC7169654 DOI: 10.3892/ijmm.2020.4543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
Resistance to the chemotherapeutic drug cisplatin has been documented in various types of cancer, while the increased expression of β‑catenin has been observed in cisplatin‑resistant ovarian cancer. However, the involvement of β‑catenin in cisplatin resistance is unclear. The present study investigated the antitumor effect of cisplatin on the proliferation, invasion and apoptosis of breast cancer (BC) cells following β‑catenin silencing in BC, which is the most frequent type of malignancy among women. The expression of β‑catenin in BC tissues and cell lines was measured by reverse transcription‑quantitative polymerase chain reaction, and the association between expression levels and clinical characteristics was statistically analyzed. The viability of BC cell lines treated with siR‑β‑catenin or with siR‑β‑catenin and cisplatin in combination was determined using a Cell Counting Kit‑8 assay. The migratory and invasive abilities of BC cells treated with both siR‑β‑catenin and cisplatin were examined with Transwell assays. The CD44 antigen/intercellular adhesion molecule 1 expression ratio, cell cycle distribution and apoptosis levels of BC cells treated with siR‑β‑catenin and cisplatin in combination were detected by flow cytometry. The expression levels of apoptosis‑associated proteins, including caspase‑3/9, in the BC cells treated with both siR‑β‑catenin and cisplatin were investigated by western blot analysis. The levels of apoptosis in the BC cells following combined treatment with siR‑β‑catenin and cisplatin was further quantified by Hoechst 33342 staining. β‑catenin was identified to be highly expressed in BC tissues and cell lines and was associated with pathological stage and lymph node status. Following knockdown of β‑catenin expression, cisplatin treatment suppressed the viabilities, and the migratory and invasive capabilities of the T47D and MCF‑7 cells, and induced extensive apoptosis. β‑catenin knockdown upregulated caspase‑3/9 levels following cisplatin treatment and induced the apoptosis of T47D and MCF‑7 cells. In conclusion, β‑catenin may be of value as a therapeutic target during cisplatin treatment in patients with BC treated with cisplatin.
Collapse
Affiliation(s)
| | - Jia Feng
- Department of Laboratory Medicine
| | - Wenguang Fu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | | | - Xue Wan
- Department of Laboratory Medicine
| | | |
Collapse
|
27
|
Jiang S, Gao Y, Yu QH, Li M, Cheng X, Hu SB, Song ZF, Zheng QC. P-21-activated kinase 1 contributes to tumor angiogenesis upon photodynamic therapy via the HIF-1α/VEGF pathway. Biochem Biophys Res Commun 2020; 526:98-104. [PMID: 32197838 DOI: 10.1016/j.bbrc.2020.03.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 03/07/2020] [Indexed: 02/07/2023]
Abstract
Photodynamic therapy (PDT) is an effective oncotherapy and has been approved for clinical application. Unfortunately, its therapeutic efficacy is usually overshadowed by tumor angiogenesis. Thus, a detailed understanding of the tumor angiogenesis upon PDT is imperative. This study aimed to investigate the potential contribution and mechanism of P-21-activated kinase 1 (PAK1) in PDT-induced tumor angiogenesis. Firstly, we found that PAK1 was upregulated upon PDT and associated with tumor angiogenesis. Then, we elucidated the underlying molecular mechanism. Activation of PAK1 prevents hypoxia-inducible factor 1 alpha (HIF-1α) protein from ubiquitin-mediated degradation. Thereafter, HIF-1α accumulation results in the upregulation of vascular endothelial growth factor (VEGF), thus promoting tumor angiogenesis. More importantly, we determined that PAK1 knockdown effectually repressed tumor angiogenesis, which contributes to enhance the therapeutic effect of PDT. Together, PAK1 is a potential novel pharmaceutical target for inhibiting PDT-induced tumor angiogenesis, and PAK1 suppression in combination with PDT may be a potentially effective strategy for anti-tumor therapy.
Collapse
Affiliation(s)
- Shuai Jiang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Gao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Hong Yu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao Bo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi Fang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qi Chang Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
28
|
Najahi‐Missaoui W, Quach ND, Jenkins A, Dabke I, Somanath PR, Cummings BS. Effect of P21-activated kinase 1 (PAK-1) inhibition on cancer cell growth, migration, and invasion. Pharmacol Res Perspect 2019; 7:e00518. [PMID: 31516713 PMCID: PMC6728842 DOI: 10.1002/prp2.518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
P21-activated kinase-1 (PAK-1) is a serine/threonine kinase involved in multiple signaling pathways that mediate cellular functions such as cytoskeletal motility, cell proliferation, and survival. PAK-1 expression is altered in various cancers, including prostate and breast. Our recent studies showed that prostate cancer cells expressing higher levels of PAK-1 were resistant to the cytotoxic effects of the PAK-1 inhibitor, inhibitor targeting PAK-1 activation-3 (IPA-3), compared to those with lower expression. This study expanded these findings to other cancers (breast and melanoma) by testing the hypothesis that genetic and pharmacological inhibition of PAK-1 alters cell growth, migration, and invasion in prostate, breast, and skin cancer cell lines. We also tested the specificity of IPA-3 for PAK-1 and the hypothesis that gene silencing of PAK-1 altered the efficacy of sterically stabilized liposomes (SSL) containing IPA-3 (SSL-IPA-3). PAK-1 expression was identified in four different breast cancer cell lines, and in a melanoma cell line. The expression of PAK-1 correlated to the IC50 of IPA-3 as measured by MTT staining. PAK-1 inhibition using shRNA correlated with decreased cell migration and invasion in prostate cancer DU-145 and breast cancer MCF-7 cells. Decreased migration and invasion also correlated to decreased expression of E-cadherin and alterations in C-X-C Chemokine Receptor type 4 and Homing Cell Adhesion Molecule expression. PAK-1 inhibition increased the cytotoxicity of IPA-3, and the cytotoxicity of SSL-IPA-3 to levels comparable to that of free drug. These data demonstrate that both pharmacological and molecular inhibition of PAK-1 decreased growth in prostate, breast, and melanoma cancer cell lines, and increased the toxicity of IPA-3 and its liposomal formulation. These data also show the specificity of IPA-3 for PAK-1, are some of the first data suggesting that IPA-3 is a therapeutic treatment for breast cancer and melanoma, and demonstrate the efficacy of liposome-encapsulated IPA-3 in breast cancer cells.
Collapse
Affiliation(s)
- Wided Najahi‐Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
| | - Nhat D. Quach
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Present address:
Department of Molecular Pharmacology, Physiology, & BiotechnologyBrown UniversityProvidenceRIUSA
| | - Amber Jenkins
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Present address:
Cancer Center of Middle GeorgiaDublinGAUSA
| | - Isha Dabke
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Present address:
Medical College of GeorgiaAugustaGAUSA
| | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics, College of PharmacyUniversity of Georgia and Charlie Norwood VA Medical CenterAugustaGAUSA
- Department of Medicine, Vascular Biology Center and Cancer CenterGeorgia Regents UniversityAugustaGAUSA
| | - Brian S. Cummings
- Department of Pharmaceutical and Biomedical Sciences, College of PharmacyUniversity of GeorgiaAthensGAUSA
- Interdisciplinary Toxicology ProgramUniversity of GeorgiaAthensGAUSA
| |
Collapse
|
29
|
Moelans CB, de Ligt J, van der Groep P, Prins P, Besselink NJM, Hoogstraat M, Ter Hoeve ND, Lacle MM, Kornegoor R, van der Pol CC, de Leng WWJ, Barbé E, van der Vegt B, Martens J, Bult P, Smit VTHBM, Koudijs MJ, Nijman IJ, Voest EE, Selenica P, Weigelt B, Reis-Filho JS, van der Wall E, Cuppen E, van Diest PJ. The molecular genetic make-up of male breast cancer. Endocr Relat Cancer 2019; 26:779-794. [PMID: 31340200 PMCID: PMC6938562 DOI: 10.1530/erc-19-0278] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/23/2019] [Indexed: 12/17/2022]
Abstract
Male breast cancer (MBC) is extremely rare and accounts for less than 1% of all breast malignancies. Therefore, clinical management of MBC is currently guided by research on the disease in females. In this study, DNA obtained from 45 formalin-fixed paraffin-embedded (FFPE) MBCs with and 90 MBCs (52 FFPE and 38 fresh-frozen) without matched normal tissues was subjected to massively parallel sequencing targeting all exons of 1943 cancer-related genes. The landscape of mutations and copy number alterations was compared to that of publicly available estrogen receptor (ER)-positive female breast cancers (smFBCs) and correlated to prognosis. From the 135 MBCs, 90% showed ductal histology, 96% were ER-positive, 66% were progesterone receptor (PR)-positive, and 2% HER2-positive, resulting in 50, 46 and 4% luminal A-like, luminal B-like and basal-like cases, respectively. Five patients had Klinefelter syndrome (4%) and 11% of patients harbored pathogenic BRCA2 germline mutations. The genomic landscape of MBC to some extent recapitulated that of smFBC, with recurrent PIK3CA (36%) and GATA3 (15%) somatic mutations, and with 40% of the most frequently amplified genes overlapping between both sexes. TP53 (3%) somatic mutations were significantly less frequent in MBC compared to smFBC, whereas somatic mutations in genes regulating chromatin function and homologous recombination deficiency-related signatures were more prevalent. MDM2 amplifications were frequent (13%), correlated with protein overexpression (P = 0.001) and predicted poor outcome (P = 0.007). In conclusion, despite similarities in the genomic landscape between MBC and smFBC, MBC is a molecularly unique and heterogeneous disease requiring its own clinical trials and treatment guidelines.
Collapse
Affiliation(s)
- Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joep de Ligt
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Petra van der Groep
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Pjotr Prins
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nicolle J M Besselink
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Marlous Hoogstraat
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Miangela M Lacle
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Robert Kornegoor
- Department of Pathology, Gelre Ziekenhuizen, Appeldoorn, The Netherlands
| | - Carmen C van der Pol
- Cancer Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wendy W J de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ellis Barbé
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Bert van der Vegt
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John Martens
- Department of Medical Oncology, Daniel den Hoed Cancer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter Bult
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Marco J Koudijs
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Isaac J Nijman
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - Emile E Voest
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elsken van der Wall
- Cancer Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Edwin Cuppen
- Department of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Cancer Genomics.nl, Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
30
|
Santiago-Gómez A, Kedward T, Simões BM, Dragoni I, NicAmhlaoibh R, Trivier E, Sabin V, Gee JM, Sims AH, Howell SJ, Clarke RB. PAK4 regulates stemness and progression in endocrine resistant ER-positive metastatic breast cancer. Cancer Lett 2019; 458:66-75. [DOI: 10.1016/j.canlet.2019.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/29/2019] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
|
31
|
Mountzios G, Kotoula V, Kolliou GA, Papadopoulou K, Lazaridis G, Christodoulou C, Pentheroudakis G, Skondra M, Koutras A, Linardou H, Razis E, Papakostas P, Chrisafi S, Aravantinos G, Nicolaou I, Goussia A, Kalogeras K, Pectasides D, Fountzilas G. Cyclin D1 differential activation and its prognostic impact in patients with advanced breast cancer treated with trastuzumab. ESMO Open 2019; 4:e000441. [PMID: 31231556 PMCID: PMC6555606 DOI: 10.1136/esmoopen-2018-000441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/20/2019] [Accepted: 01/23/2019] [Indexed: 01/14/2023] Open
Abstract
Introduction We sought to determine the level of activation of the critical components of the cyclin D1-mediated pathway and to evaluate their prognostic significance across the different molecular subtypes of advanced breast cancer. Patients and methods The study population comprised 219 female patients with advanced breast cancer who had been found to have human epidermal growth factor receptor 2 (HER2)-positive disease by local testing and were all treated with trastuzumab-based regimens. For all tumours, central testing for HER2 was performed, and cyclin D1 gene (CCND1) amplification, mRNA and protein expression were assessed by FISH, quantitative real-time-PCR and immunohistochemistry, respectively. Prognostic impact on clinical endpoints was evaluated with Cox regression analyses. Results After central testing, only 134 (61.2%) of 219 patients were confirmed to have HER2 gene amplification by FISH and/or 3+ HER2 protein expression by immunohistochemistry. After a median follow-up time of 136.0 months (95% CI 123.3 to 148.9), 105 (78.4%) HER2-positive patients and 76 (89.4%) HER2-negative patients had died, while 80% of the former and 87.1% of the latter had experienced a disease relapse. Patients with positive oestrogen receptor/progesterone receptor status presented with higher cyclin D1 mRNA expression. In the HER2-negative subgroup, patients with negative cyclin D1 protein expression were at higher risk of progression (HR= 1.66, 95%CI 1.01 to 2.72, Wald’s p=0.045). Among de novo metastatic patients, the risk of progression was higher for patients with non-amplified CCND1 tumours (HR= 2.00, 95% CI 1.03 to 3.90, p=0.041). Conclusion Aberrant activation of the cyclin D1-mediated pathway appears to reduce the risk of progression in HER2-negative tumours, but not in HER2-positive ones.
Collapse
Affiliation(s)
- G Mountzios
- School of Medicine, University of Athens, Athens, Greece
| | - Vassiliki Kotoula
- Laboratory of Molecular Oncology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Lazaridis
- Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Maria Skondra
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, Greece
| | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | - Helena Linardou
- First Department of Medical Oncology, Metropolitan Hospital, Piraeus, Greece
| | - Evangelia Razis
- Third Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | | | - Sofia Chrisafi
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gerasimos Aravantinos
- Second Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| | - Irene Nicolaou
- Department of Histopathology, Agii Anargiri Hospital, Athens, Greece
| | - Anna Goussia
- Pathology, University of Ioannina, Ioannina, Greece
| | | | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, Greece
| | - George Fountzilas
- Department of Medical Oncology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
32
|
Lundberg A, Lindström LS, Li J, Harrell JC, Darai-Ramqvist E, Sifakis EG, Foukakis T, Perou CM, Czene K, Bergh J, Tobin NP. The long-term prognostic and predictive capacity of cyclin D1 gene amplification in 2305 breast tumours. Breast Cancer Res 2019; 21:34. [PMID: 30819233 PMCID: PMC6394106 DOI: 10.1186/s13058-019-1121-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Use of cyclin D1 (CCND1) gene amplification as a breast cancer biomarker has been hampered by conflicting assessments of the relationship between cyclin D1 protein levels and patient survival. Here, we aimed to clarify its prognostic and treatment predictive potential through comprehensive long-term survival analyses. METHODS CCND1 amplification was assessed using SNP arrays from two cohorts of 1965 and 340 patients with matching gene expression array and clinical follow-up data of over 15 years. Kaplan-Meier and multivariable Cox regression analyses were used to determine survival differences between CCND1 amplified vs. non-amplified tumours in clinically relevant patient sets, within PAM50 subtypes and within treatment-specific subgroups. Boxplots and differential gene expression analyses were performed to assess differences between amplified vs. non-amplified tumours within PAM50 subtypes. RESULTS When combining both cohorts, worse survival was found for patients with CCND1-amplified tumours in luminal A (HR = 1.68; 95% CI, 1.15-2.46), luminal B (1.37; 1.01-1.86) and ER+/LN-/HER2- (1.66; 1.14-2.41) subgroups. In gene expression analysis, CCND1-amplified luminal A tumours showed increased proliferation (P < 0.001) and decreased progesterone (P = 0.002) levels along with a large overlap in differentially expressed genes when comparing luminal A and B-amplified vs. non-amplified tumours. CONCLUSIONS Our results indicate that CCND1 amplification is associated with worse 15-year survival in ER+/LN-/HER2-, luminal A and luminal B patients. Moreover, luminal A CCND1-amplified tumours display gene expression changes consistent with a more aggressive phenotype. These novel findings highlight the potential of CCND1 to identify patients that could benefit from long-term treatment strategies.
Collapse
Affiliation(s)
- Arian Lundberg
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Linda S Lindström
- Department of Biosciences and Nutrition, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Jingmei Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
| | - J Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, USA
| | - Eva Darai-Ramqvist
- Department of Pathology and Cytology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Emmanouil G Sifakis
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Theodoros Foukakis
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Charles M Perou
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Department of Public Health, Oxford University, Oxford, UK
| | - Nicholas P Tobin
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| |
Collapse
|
33
|
Haque MM, Desai KV. Pathways to Endocrine Therapy Resistance in Breast Cancer. Front Endocrinol (Lausanne) 2019; 10:573. [PMID: 31496995 PMCID: PMC6712962 DOI: 10.3389/fendo.2019.00573] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/06/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancers with positive expression of Estrogen Receptor (ER+) are treated with anti-hormone/endocrine therapy which targets the activity of the receptor, the half-life of the receptor or the availability of estrogen. This has significantly decreased mortality in women with ER+ breast cancer, however, about 25-30% of treated women run the risk or recurrence due to either intrinsic or acquired resistance to endocrine therapies. While ER itself is a predictor of response to such therapies, there exists a need to find more biomarkers and novel targets to treat resistant tumors. In this review, we summarize the known mechanisms and describe the ability of genomics in unraveling rare mutations and gene rearrangements that may impact the development of resistance and therefore treatment of ER+ breast cancer in the near future.
Collapse
|
34
|
Beelen K, Opdam M, Severson T, Koornstra R, Vincent A, Wesseling J, Sanders J, Vermorken J, van Diest P, Linn S. Mitotic count can predict tamoxifen benefit in postmenopausal breast cancer patients while Ki67 score cannot. BMC Cancer 2018; 18:761. [PMID: 30041599 PMCID: PMC6057037 DOI: 10.1186/s12885-018-4516-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background Controversy exists for the use of Ki67 protein expression as a predictive marker to select patients who do or do not derive benefit from adjuvant endocrine therapy. Whether other proliferation markers, like Cyclin D1, and mitotic count can also be used to identify those estrogen receptor α (ERα) positive breast cancer patients that derive benefit from tamoxifen is not well established. We tested the predictive value of these markers for tamoxifen benefit in ERα positive postmenopausal breast cancer patients. Methods We collected primary tumor blocks from 563 ERα positive patients who had been randomized between tamoxifen (1 to 3 years) vs. no adjuvant therapy (IKA trial) with a median follow-up of 7.8 years. Mitotic count, Ki67 and Cyclin D1 protein expression were centrally assessed by immunohistochemistry on tissue microarrays. In addition, we tested the predictive value of CCND1 gene copy number variation using MLPA technology. Multivariate Cox proportional hazard models including interaction between marker and treatment were used to test the predictive value of these markers. Results Patients with high Ki67 (≥5%) as well as low (< 5%) expressing tumors equally benefitted from adjuvant tamoxifen (adjusted hazard ratio (HR) 0.5 for both groups)(p for interaction 0.97). We did not observe a significant interaction between either Cyclin D1 or Ki67 and tamoxifen, indicating that the relative benefit from tamoxifen was not dependent on the level of these markers. Patients with tumors with low mitotic count derived substantial benefit from tamoxifen (adjusted HR 0.24, p < 0.0001), while patients with tumors with high mitotic count derived no significant benefit (adjusted HR 0.64, p = 0.14) (p for interaction 0.03). Conclusion Postmenopausal breast cancer patients with high Ki67 counts do significantly benefit from adjuvant tamoxifen, while those with high mitotic count do not. Mitotic count is a better selection marker for reduced tamoxifen benefit than Ki67. Electronic supplementary material The online version of this article (10.1186/s12885-018-4516-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karin Beelen
- Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Opdam
- Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tesa Severson
- Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rutger Koornstra
- Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andrew Vincent
- Departments of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jelle Wesseling
- Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joyce Sanders
- Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan Vermorken
- Department of Medical Oncology, University Hospital Antwerpen, Edegem, Belgium
| | - Paul van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sabine Linn
- Molecular Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands. .,Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Patel HK, Bihani T. Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther 2018; 186:1-24. [DOI: 10.1016/j.pharmthera.2017.12.012] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Zhang X, Xue D, Hao F, Xie L, He J, Gai J, Liu Y, Xu H, Li Q, Wang E. Remodeling and spacing factor 1 overexpression is associated with poor prognosis in renal cell carcinoma. Oncol Lett 2018; 15:3852-3857. [PMID: 29467902 DOI: 10.3892/ol.2018.7797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/02/2017] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to assess the expression and prognostic significance of remodeling and spacing factor 1 (RSF1; HBXAP) in renal cell carcinoma (RCC). RSF1 expression was analyzed using immunohistochemistry on tissue samples from a consecutive series of 137 patients with RCC who underwent tumor resection between November 2000 and March 2004. The associations between RSF1 expression, clinicopathological factors and patient survival were investigated. Immunohistochemistry revealed that RSF1 was highly expressed in 43.1% (59/137) of the RCC samples. RSF1 expression levels were associated with the T stage of the Tumor-Node-Metastasis grading system. Kaplan-Meier survival analysis indicated that high RSF1 expression in RCC was significantly associated with a poor prognosis. Multivariate analysis revealed that RSF1 expression is an independent prognostic parameter for the duration of overall survival of patients with RCC. The results demonstrated that a high expression level of RSF1 in RCC is associated with advanced tumor stages and a poor prognosis. To the best of our knowledge, the present study provides novel evidence of the biological significance of RSF1 expression in RCC.
Collapse
Affiliation(s)
- Xiuwei Zhang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China.,Department of Pathology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Dongwei Xue
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Fengxia Hao
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lingling Xie
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jiani He
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Junda Gai
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuhui Liu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hongtao Xu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Qingchang Li
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Enhua Wang
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
37
|
Fumagalli D, Wilson TR, Salgado R, Lu X, Yu J, O'Brien C, Walter K, Huw LY, Criscitiello C, Laios I, Jose V, Brown DN, Rothé F, Maetens M, Zardavas D, Savas P, Larsimont D, Piccart-Gebhart MJ, Michiels S, Lackner MR, Sotiriou C, Loi S. Somatic mutation, copy number and transcriptomic profiles of primary and matched metastatic estrogen receptor-positive breast cancers. Ann Oncol 2017; 27:1860-6. [PMID: 27672107 DOI: 10.1093/annonc/mdw286] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 07/14/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Estrogen receptor-positive (ER+) breast cancers (BCs) constitute the most frequent BC subtype. The molecular landscape of ER+ relapsed disease is not well characterized. In this study, we aimed to describe the genomic evolution between primary (P) and matched metastatic (M) ER+ BCs after failure of adjuvant therapy. MATERIALS AND METHODS A total of 182 ER+ metastatic BC patients with long-term follow-up were identified from a single institution. P tumor tissue was available for all patients, with 88 having matched M material. According to the availability of tumor material, samples were characterized using a 120 mutational hotspot qPCR, a 29 gene copy number aberrations (CNA) and a 400 gene expression panels. ESR1 mutations were assayed by droplet digital PCR. Molecular alterations were correlated with overall survival (OS) using the Cox proportional hazards regression models. RESULTS The median follow-up was 6.4 years (range 0.5-26.6 years). Genomic analysis of P tumors revealed somatic mutations in PIK3CA, KRAS, AKT1, FGFR3, HRAS and BRAF at frequencies of 41%, 6%, 5%, 2%, 1% and 2%, respectively, and CN amplification of CCND1, ZNF703, FGFR1, RSF1 and PAK1 at 23%, 19%, 17%, 12% and 11%, respectively. Mutations and CN amplifications were largely concordant between P and matched M (>84%). ESR1 mutations were found in 10.8% of the M but none of the P. Thirteen genes, among which ESR1, FOXA1, and HIF1A, showed significant differential expression between P and M. In P, the differential expression of 18 genes, among which IDO1, was significantly associated with OS (FDR < 0.1). CONCLUSIONS Despite the large concordance between P and matched M for the evaluated molecular alterations, potential actionable targets such as ESR1 mutations were found only in M. This supports the importance of characterizing the M disease. Other targets we identified, such as HIF1A and IDO1, warrant further investigation in this patient population.
Collapse
Affiliation(s)
- D Fumagalli
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - T R Wilson
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - R Salgado
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - X Lu
- Department of Biostatistics, Genentech Inc., South San Francisco, CA, USA
| | - J Yu
- Department of Biostatistics, Genentech Inc., South San Francisco, CA, USA
| | - C O'Brien
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - K Walter
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - L Y Huw
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - C Criscitiello
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy
| | - I Laios
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium
| | - V Jose
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - D N Brown
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - F Rothé
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - M Maetens
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - D Zardavas
- Breast International Group, Brussels, Belgium
| | - P Savas
- Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - D Larsimont
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium
| | | | - S Michiels
- Division of Biostatistics and Epidemiology, Institut Gustave Roussy, Villejuif, France INSERM U1018, CESP, University of Paris, Villejuif, France
| | - M R Lackner
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - C Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium Division of Medical Oncology, Institut Jules Bordet, Brussels, Belgium
| | - S Loi
- Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Cloning and functional characterization of human Pak1 promoter by steroid hormones. Gene 2017; 646:120-128. [PMID: 29274909 DOI: 10.1016/j.gene.2017.12.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/29/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022]
Abstract
P21-activated kinase 1 (Pak1) is known to be involved in a plethora of functions including cell growth, survival and can lead to cell transformation and tumor progression especially in breast tissue. Multiple studies have shown Pak1 dysregulation as a change in DNA copy number as well as gene expression levels, suggesting many regulatory mechanisms at transcriptional and translational level. However, very little is known about the transcriptional regulation of the human Pak1 promoter. Here, we focus on Pak1 promoter regulation by steroid hormones along with their respective receptors that are also crucial players in breast tissue function and tumorigenesis. Our results show high Pak1 expression in breast cancer cell lines and in breast tumor tissue. It also suggests that Pak1 is hormone responsive, whose expression can be modulated by steroid hormones namely, estrogen in the form of 17β-estradiol (E2) and progesterone (P4). Sequence analysis of a 3.2kb Pak1 proximal promoter region shows the presence of PRE (progesterone response element) and ERE (estrogen response element) half sites, that were further cloned and characterized. Results from promoter analysis showed that Pak1 promoter activity is mediated by PR via its binding to PRE present on the Pak1 promoter that was further reaffirmed in vitro by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation assay (ChIP). Our results together suggest that it is the PR isoform B regulates Pak1 promoter. To our knowledge, this is the first study to report the detailed characterization and transcriptional regulation of the human Pak1 promoter by steroid hormones.
Collapse
|
39
|
Physical Confirmation and Comparative Genomics of the Rat Mammary carcinoma susceptibility 3 Quantitative Trait Locus. G3-GENES GENOMES GENETICS 2017; 7:1767-1773. [PMID: 28391240 PMCID: PMC5473756 DOI: 10.1534/g3.117.039388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Human breast and rat mammary cancer susceptibility are complex phenotypes where complete sets of risk associated loci remain to be identified for both species. We tested multiple congenic rat strains to physically confirm and positionally map rat Mammary carcinoma susceptibility 3 (Mcs3)-a mammary cancer resistance allele previously predicted at Rattus norvegicus chromosome 1 (RNO1). The mammary cancer susceptible Wistar Furth (WF) strain was the recipient, and the mammary cancer resistant Copenhagen (Cop) strain was the RNO1-segment donor for congenics. Inbred WF females averaged 6.3 carcinogen-induced mammary carcinomas per rat. Two WF.Cop congenic strains averaged 2.8 and 3.4 mammary carcinomas per rat, which confirmed Mcs3 as an independently acting allele. Two other WF.Cop congenic strains averaged 6.6 and 8.1 mammary carcinomas per rat, and, thus, did not contain Mcs3 Rat Mcs3 was delimited to 27.8 Mb of RNO1 from rs8149408 to rs105131702 (RNO1:143700228-171517317 of RGSC 6.0/rn6). Human genetic variants with p values for association to breast cancer risk below 10-7 had not been reported for Mcs3 orthologous loci; however, human variants located in Mcs3-orthologous regions with potential association to risk (10-7 < p < 10-3) were listed in some population-based studies. Further, rat Mcs3 contains sequence orthologous to human 11q13/14-a region frequently amplified in female breast cancer. We conclude that Mcs3 is an independently acting mammary carcinoma resistance allele. Human population-based, genome-targeted association studies interrogating Mcs3 orthologous loci may yield novel breast cancer risk associated variants and genes.
Collapse
|
40
|
High expression of cyclin D1 is associated to high proliferation rate and increased risk of mortality in women with ER-positive but not in ER-negative breast cancers. Breast Cancer Res Treat 2017; 164:667-678. [PMID: 28528450 PMCID: PMC5495873 DOI: 10.1007/s10549-017-4294-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/13/2017] [Indexed: 12/19/2022]
Abstract
Purpose Cyclin D1 has a central role in cell cycle control and is an important component of estrogen regulation of cell cycle progression. We have previously shown that high cyclin D expression is related to aggressive features of ER-positive but not ER-negative breast cancer. The aims of the present study were to validate this differential ER-related effect and furthermore explore the relationship between cyclin D overexpression and CCND1 gene amplification status in a node-negative breast cancer case–control study. Methods Immunohistochemical nuclear expression of cyclin D1 (n = 364) and amplification of the gene CCND1 by fluorescent in situ hybridization (n = 255) was performed on tissue microarray sections from patients with T1-2N0M0 breast cancer. Patients given adjuvant chemotherapy were excluded. The primary event was defined as breast cancer death. Breast cancer-specific survival was analyzed in univariate and multivariable models using conditional logistic regression. Results Expression of cyclin D1 above the median (61.7%) in ER breast cancer was associated with an increased risk for breast cancer death (OR 3.2 95% CI 1.5–6.8) also when adjusted for tumor size and grade (OR 3.1). No significant prognostic impact of cyclin D1 expression was found among ER-negative cases. Cyclin D1 overexpression was significantly associated to high expression of the proliferation markers cyclins A (ρ 0.19, p = 0.006) and B (ρ 0.18, p = 0.003) in ER-positive tumors, but not in ER-negative cases. There was a significant association between CCND1 amplification and cyclin D1 expression (p = 0.003), but CCND1 amplification was not statistically significantly prognostic (HR 1.4, 95% CI 0.4–4.4). Conclusion We confirmed our previous observation that high cyclin D1 expression is associated to high proliferation and a threefold higher risk of death from breast cancer in ER-positive breast cancer.
Collapse
|
41
|
Overexpression of Rsf-1 correlates with poor survival and promotes invasion in non-small cell lung cancer. Virchows Arch 2017; 470:553-560. [PMID: 28289901 DOI: 10.1007/s00428-017-2102-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 02/01/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
Rsf-1 (HBXAP) was recently reported to play roles in tumorigenesis and tumor progression. There have been many reports referred to Rsf-1 overexpression in various cancers and associated with the malignant behavior of cancer cells. However, the molecular mechanism of Rsf-1 in non-small cell lung cancer aggressiveness remains ambiguous. In the present study, we found that there was a significant association between Rsf-1 overexpression and poor overall survival (p = 0.028) in lung cancer. Furthermore, knockdown of Rsf-1 expression in H1299 and H460 cells with high endogenous Rsf-1 expression inhibited cell migration and invasion and downregulated MMP2 expression and nuclear levels of NF-κB. NF-κB inhibitor could also block the effect of Rsf-1 in regulation of MMP2 expression. Further experiments demonstrated that Rsf-1 depletion restrained NF-κB reporter luciferase activity and downregulated bcl-2 and p-IκB protein level. In conclusion, we demonstrated that Rsf-1 was overexpressed in lung cancer and associated with poor survival. Rsf-1 regulated cell invasion through MMP2 and NF-κB pathway.
Collapse
|
42
|
Pharmacological inhibition of Rac1-PAK1 axis restores tamoxifen sensitivity in human resistant breast cancer cells. Cell Signal 2017; 30:154-161. [DOI: 10.1016/j.cellsig.2016.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 12/05/2016] [Accepted: 12/05/2016] [Indexed: 11/22/2022]
|
43
|
Kumar R, Sanawar R, Li X, Li F. Structure, biochemistry, and biology of PAK kinases. Gene 2016; 605:20-31. [PMID: 28007610 DOI: 10.1016/j.gene.2016.12.014] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/24/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
PAKs, p21-activated kinases, play central roles and act as converging junctions for discrete signals elicited on the cell surface and for a number of intracellular signaling cascades. PAKs phosphorylate a vast number of substrates and act by remodeling cytoskeleton, employing scaffolding, and relocating to distinct subcellular compartments. PAKs affect wide range of processes that are crucial to the cell from regulation of cell motility, survival, redox, metabolism, cell cycle, proliferation, transformation, stress, inflammation, to gene expression. Understandably, their dysregulation disrupts cellular homeostasis and severely impacts key cell functions, and many of those are implicated in a number of human diseases including cancers, neurological disorders, and cardiac disorders. Here we provide an overview of the members of the PAK family and their current status. We give special emphasis to PAK1 and PAK4, the prototypes of groups I and II, for their profound roles in cancer, the nervous system, and the heart. We also highlight other family members. We provide our perspective on the current advancements, their growing importance as strategic therapeutic targets, and our vision on the future of PAKs.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA; Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India.
| | - Rahul Sanawar
- Cancer Biology Program, Rajiv Gandhi Center of Biotechnology, Thiruvananthapuram 695014, India
| | - Xiaodong Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Chinese Ministry of Education, China Medical University, Shenyang 110122, China.
| |
Collapse
|
44
|
Gene expression, signal transduction pathways and functional networks associated with growth of sporadic vestibular schwannomas. J Neurooncol 2016; 131:283-292. [PMID: 27752882 DOI: 10.1007/s11060-016-2292-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/09/2016] [Indexed: 10/20/2022]
Abstract
The objective of this study was to determine global gene expression in relation to Vestibular schwannomas (VS) growth rate and to identify signal transduction pathways and functional molecular networks associated with growth. Repeated magnetic resonance imaging (MRI) prior to surgery determined tumor growth rate. Following tissue sampling during surgery, mRNA was extracted from 16 sporadic VS. Double stranded cDNA was synthesized from the mRNA and used as template for in vitro transcription reaction to synthesize biotin-labeled antisense cRNA, which was hybridized to Affymetrix HG-U133A arrays and analyzed by dChip software. Differential gene expression was defined as a 1.5-fold difference between fast and slow growing tumors (><0.5 ccm/year), employing a p-value <0.01. Deregulated transcripts were matched against established gene ontology. Ingenuity Pathway Analysis was used for identification of signal transduction pathways and functional molecular networks associated with tumor growth. In total 109 genes were deregulated in relation to tumor growth rate. Genes associated with apoptosis, growth and cell proliferation were deregulated. Gene ontology included regulation of the cell cycle, cell differentiation and proliferation, among other functions. Fourteen pathways were associated with tumor growth. Five functional molecular networks were generated. This first study on global gene expression in relation to vestibular schwannoma growth rate identified several genes, signal transduction pathways and functional networks associated with tumor progression. Specific genes involved in apoptosis, cell growth and proliferation were deregulated in fast growing tumors. Fourteen pathways were associated with tumor growth. Generated functional networks underlined the importance of the PI3K family, among others.
Collapse
|
45
|
Zhuang T, Zhu J, Li Z, Lorent J, Zhao C, Dahlman-Wright K, Strömblad S. p21-activated kinase group II small compound inhibitor GNE-2861 perturbs estrogen receptor alpha signaling and restores tamoxifen-sensitivity in breast cancer cells. Oncotarget 2016; 6:43853-68. [PMID: 26554417 PMCID: PMC4791272 DOI: 10.18632/oncotarget.6081] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/26/2015] [Indexed: 01/07/2023] Open
Abstract
Estrogen receptor alpha (ERα) is highly expressed in most breast cancers. Consequently, ERα modulators, such as tamoxifen, are successful in breast cancer treatment, although tamoxifen resistance is commonly observed. While tamoxifen resistance may be caused by altered ERα signaling, the molecular mechanisms regulating ERα signaling and tamoxifen resistance are not entirely clear. Here, we found that PAK4 expression was consistently correlated to poor patient outcome in endocrine treated and tamoxifen-only treated breast cancer patients. Importantly, while PAK4 overexpression promoted tamoxifen resistance in MCF-7 human breast cancer cells, pharmacological treatment with a group II PAK (PAK4, 5, 6) inhibitor, GNE-2861, sensitized tamoxifen resistant MCF-7/LCC2 breast cancer cells to tamoxifen. Mechanistically, we identified a regulatory positive feedback loop, where ERα bound to the PAK4 gene, thereby promoting PAK4 expression, while PAK4 in turn stabilized the ERα protein, activated ERα transcriptional activity and ERα target gene expression. Further, PAK4 phosphorylated ERα-Ser305, a phosphorylation event needed for the PAK4 activation of ERα-dependent transcription. In conclusion, PAK4 may be a suitable target for perturbing ERα signaling and tamoxifen resistance in breast cancer patients.
Collapse
Affiliation(s)
- Ting Zhuang
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Jian Zhu
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Zhilun Li
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Julie Lorent
- Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden.,Science for Life Laboratory (SciLifeLab), Karolinska Institutet, Solna, Sweden
| | - Staffan Strömblad
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
46
|
Yellapu NK, Pulaganti M, Pakala SB. Bioinformatics exploration of PAK1 (P21-activated kinase-1) revealed potential network gene elements in breast invasive carcinoma. J Biomol Struct Dyn 2016; 35:2269-2279. [DOI: 10.1080/07391102.2016.1216894] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Nanda Kumar Yellapu
- Biomedical Informatics Centre, Vector Control Research Centre (VCRC)-ICMR, Pondicherry 605006, India
| | - Madhusudana Pulaganti
- Multi-Disciplinary Research Unit, Sri Venkateswara Medical College, Tirupati 517501, India
| | - Suresh Babu Pakala
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517507, India
| |
Collapse
|
47
|
Girotra S, Yeghiazaryan K, Golubnitschaja O. Potential biomarker panels in overall breast cancer management: advancements by multilevel diagnostics. Per Med 2016; 13:469-484. [PMID: 29767597 DOI: 10.2217/pme-2016-0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Breast cancer (BC) prevalence has reached an epidemic scale with half a million deaths annually. Current deficits in BC management include predictive and preventive approaches, optimized screening programs, individualized patient profiling, highly sensitive detection technologies for more precise diagnostics and therapy monitoring, individualized prediction and effective treatment of BC metastatic disease. To advance BC management, paradigm shift from delayed to predictive, preventive and personalized medical services is essential. Corresponding step forwards requires innovative multilevel diagnostics procuring specific panels of validated biomarkers. Here, we discuss current instrumental advancements including genomics, proteomics, epigenetics, miRNA, metabolomics, circulating tumor cells and cancer stem cells with a focus on biomarker discovery and multilevel diagnostic panels. A list of the recommended biomarker candidates is provided.
Collapse
|
48
|
Ahern TP, Cronin-Fenton DP, Lash TL, Sørensen HT, Ording AG, Hamilton-Dutoit SJ, Hellberg Y. Pak1, adjuvant tamoxifen therapy, and breast cancer recurrence risk in a Danish population-based study. Acta Oncol 2016; 55:734-41. [PMID: 27056567 DOI: 10.3109/0284186x.2016.1150606] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Adjuvant tamoxifen therapy approximately halves the risk of estrogen receptor-positive (ER+) breast cancer recurrence, but many women do not respond to therapy. Observational studies nested in clinical trial populations suggest that overexpression or nuclear localization of p21-activated kinase 1 (Pak1) in primary tumors predicts tamoxifen failure. Material and methods We measured the association between Pak1 expression and breast cancer recurrence in a Danish population-based case-control study. Pak1 cytoplasmic expression level and nuclear positivity were determined by immunohistochemical staining of primary breast tumors from recurrence cases and matched controls from two breast cancer populations; women diagnosed with ER-positive tumors who received at least one year of tamoxifen therapy (ER+/TAM+), and women diagnosed with ER-negative tumors who survived for at least one year (ER-/TAM-). Pak1 staining was assessed by a single, blinded pathologist, and associations were estimated with conditional logistic regression models. Results We included 541 recurrence cases and 1:1 matched controls from the ER+/TAM + group and 300 recurrence cases and 1:1 matched controls from the ER-/TAM - group. Pak1 cytoplasmic intensity was not associated with breast cancer recurrence in either group (ER+/TAM + ORadj for strong vs. no cytoplasmic staining = 0.91, 95% CI 0.57, 1.5; ER-/TAM - ORadj for strong vs. no cytoplasmic staining = 0.74, 95% CI 0.39, 1.4). Associations between Pak1 nuclear positivity and breast cancer recurrence were similarly near null in both groups. Conclusion Pak1 positivity in primary breast tumors was neither predictive nor prognostic in this prospective, population-based study.
Collapse
Affiliation(s)
- Thomas P. Ahern
- Department of Surgery, University of Vermont College of Medicine, Burlington, Vermont, USA
| | | | - Timothy L. Lash
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Gulbech Ording
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Ylva Hellberg
- Institute of Pathology, Aarhus University Hospital, Aarhus, Denmark
- Department of Pathology, Hvidovre Hospital, Hvidovre Municipality, Denmark
| |
Collapse
|
49
|
Parvathy M, Sreeja S, Kumar R, Pillai MR. Potential role of p21 Activated Kinase 1 (PAK1) in the invasion and motility of oral cancer cells. BMC Cancer 2016; 16 Suppl 1:293. [PMID: 27229476 PMCID: PMC4896241 DOI: 10.1186/s12885-016-2263-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Oral cancer malignancy consists of uncontrolled division of cells primarily in and around the floor of the oral cavity, gingiva, oropharynx, lower lip and base of the tongue. According to GLOBOCAN 2012 report, oral cancer is one of the most common cancers among males and females in India. Even though significant advancements have been made in the field of oral cancer treatment modalities, the overall prognosis for the patients has not improved in the past few decades and hence, this demands a new thrust for the identification of novel therapeutic targets in oral cancer. p21 Activated Kinases (PAKs) are potential therapeutic targets that are involved in numerous physiological functions. PAKs are serine-threonine kinases and they serve as important regulators of cytoskeletal dynamics and cell motility, transcription through MAP kinase cascades, death and survival signalling, and cell-cycle progression. Although PAKs are known to play crucial roles in cancer progression, the role and clinical significance of PAKs in oral cancer remains poorly understood. RESULTS Our results suggest that PAK1 is over-expressed in oral cancer cell lines. Stimulation of Oral Squamous Cell Carcinoma (OSCC) cells with serum growth factors leads to PAK1 re-localization and might cause a profound cytoskeletal remodelling. PAK1 was also found to be involved in the invasion, migration and cytoskeletal remodelling of OSCC cells. CONCLUSIONS Our study revealed that PAK1 may play a crucial role in the progression of OSCC. Studying the role of PAK1 and its substrates is likely to enhance our understanding of oral carcinogenesis and potential therapeutic value of PAKs in oral cancer.
Collapse
Affiliation(s)
- Muraleedharan Parvathy
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Sreeharshan Sreeja
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Rakesh Kumar
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Department of Biochemistry and Molecular Medicine, George Washington University, Washington DC, USA
| | | |
Collapse
|
50
|
Gautam P, Karhinen L, Szwajda A, Jha SK, Yadav B, Aittokallio T, Wennerberg K. Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells. Mol Cancer 2016; 15:34. [PMID: 27165605 PMCID: PMC4862054 DOI: 10.1186/s12943-016-0517-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/30/2016] [Indexed: 01/23/2023] Open
Abstract
Background Triple negative breast cancer (TNBC) is a highly heterogeneous and aggressive type of cancer that lacks effective targeted therapy. Despite detailed molecular profiling, no targeted therapy has been established. Hence, with the aim of gaining deeper understanding of the functional differences of TNBC subtypes and how that may relate to potential novel therapeutic strategies, we studied comprehensive anticancer-agent responses among a panel of TNBC cell lines. Method The responses of 301 approved and investigational oncology compounds were measured in 16 TNBC cell lines applying a functional profiling approach. To go beyond the standard drug viability effect profiling, which has been used in most chemosensitivity studies, we utilized a multiplexed readout for both cell viability and cytotoxicity, allowing us to differentiate between cytostatic and cytotoxic responses. Results Our approach revealed that most single-agent anti-cancer compounds that showed activity for the viability readout had no or little cytotoxic effects. Major compound classes that exhibited this type of response included anti-mitotics, mTOR, CDK, and metabolic inhibitors, as well as many agents selectively inhibiting oncogene-activated pathways. However, within the broad viability-acting classes of compounds, there were often subsets of cell lines that responded by cell death, suggesting that these cells are particularly vulnerable to the tested substance. In those cases we could identify differential levels of protein markers associated with cytotoxic responses. For example, PAI-1, MAPK phosphatase and Notch-3 levels associated with cytotoxic responses to mitotic and proteasome inhibitors, suggesting that these might serve as markers of response also in clinical settings. Furthermore, the cytotoxicity readout highlighted selective synergistic and synthetic lethal drug combinations that were missed by the cell viability readouts. For instance, the MEK inhibitor trametinib synergized with PARP inhibitors. Similarly, combination of two non-cytotoxic compounds, the rapamycin analog everolimus and an ATP-competitive mTOR inhibitor dactolisib, showed synthetic lethality in several mTOR-addicted cell lines. Conclusions Taken together, by studying the combination of cytotoxic and cytostatic drug responses, we identified a deeper spectrum of cellular responses both to single agents and combinations that may be highly relevant for identifying precision medicine approaches in TNBC as well as in other types of cancers. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0517-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Prson Gautam
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Leena Karhinen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Agnieszka Szwajda
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sawan Kumar Jha
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Bhagwan Yadav
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
| |
Collapse
|