1
|
Liu G, Lin W, Zhang K, Chen K, Niu G, Zhu Y, Liu Y, Li P, Li Z, An Y. Elucidating the prognostic and therapeutic significance of TOP2A in various malignancies. Cancer Genet 2024; 288-289:68-81. [PMID: 39454521 DOI: 10.1016/j.cancergen.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Topoisomerase IIα (TOP2A) is a crucial enzyme that plays a vital role in DNA replication and transcription mechanisms. Dysregulated expression of TOP2A has been associated with various malignancies, including hepatocellular carcinoma, prostate cancer, colon cancer, lung cancer and breast cancer. In this review, we summarized the prognostic relevances of TOP2A in various types of cancer. The increased expression of TOP2A has been linked to resistance to therapy and reduced survival rates. Therefore, evaluating TOP2A levels could assist in identifying patients who may derive advantages from molecular targeted therapy. The amplification of TOP2A has been linked to a positive response to chemotherapy regimens that contain anthracycline. Nevertheless, the overexpression of TOP2A also indicates a heightened likelihood of disease recurrence and unfavorable prognosis. The prognostic significance of TOP2A has been extensively studied in various types of cancer. The increased expression of TOP2A is associated with poor clinical outcomes, indicating its potential as a valuable biomarker for assessing risk and stratifying treatment in these malignancies. However, further investigation is needed to elucidate the underlying mechanisms by which TOP2A influences cancer progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Wenlong Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Kangxu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Guanglin Niu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key laboratory of cell signal transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
2
|
Imyanitov EN, Iyevleva AG. Molecular tests for prediction of tumor sensitivity to cytotoxic drugs. Cancer Lett 2022; 526:41-52. [PMID: 34808283 DOI: 10.1016/j.canlet.2021.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/15/2022]
Abstract
Chemotherapy constitutes the backbone of cancer treatment. Several predictive assays assist personalized administration of cytotoxic drugs and are recommended for use in a clinical setting. The deficiency of DNA repair by homologous recombination (HRD), which is caused by inactivation of BRCA1/2 genes or other genetic events, is associated with high tumor responsiveness to platinum compounds, bifunctional alkylating agents and topoisomerase II poisons. Low activity of MGMT predicts the efficacy of nitrosoureas and tetrazines. Some clinically established pharmacogenetic tests allow for the adjustment of drug dosage, for example, the analysis of DPYD allelic variants for administration of fluoropyrimidines and UGT1A1 genotyping for the use of irinotecan. While there are promising molecular predictors of tumor sensitivity to pemetrexed, gemcitabine and taxanes, they remain in the investigational stage and require additional validation. Comprehensive molecular analysis of tumors obtained from drug responders and non-responders is likely to reveal new clinically useful predictive markers for cytotoxic therapy.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia; Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 191015, Russia.
| | - Aglaya G Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia
| |
Collapse
|
3
|
Luan S, Gao Y, Liang X, Zhang L, Wu Q, Hu Y, Yin L, He C, Liu S. Aconitine linoleate, a natural lipo-diterpenoid alkaloid, stimulates anti-proliferative activity reversing doxorubicin resistance in MCF-7/ADR breast cancer cells as a selective topoisomerase IIα inhibitor. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:65-76. [PMID: 34727218 DOI: 10.1007/s00210-021-02172-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/21/2021] [Indexed: 11/27/2022]
Abstract
Aconitine linoleate (1) is a lipo-diterpenoid alkaloid, isolated from Aconitum sinchiangense W. T. Wang. The study aimed at investigating the anti-proliferative efficacy and the underlying mechanisms of 1 against MCF-7 and MCF-7/ADR cells, as well as obvious the safety evaluation in vivo. The cytotoxic activities of 1 were measured in vitro. Also, we investigated the latent mechanism of 1 by cell cycle analysis in MCF-7/ADR cells and topo I and topo IIα inhibition assay. Molecular docking is done by Discovery Studio 3.5 and Autodock vina 1.1.2. Finally, the acute toxicity of 1 was detected on mice. 1 exhibited significant antitumor activity against both MCF-7 and MCF-7/ADR cells, with IC50 values of 7.58 and 7.02 μM, which is 2.38 times and 5.05 times more active, respectively than etoposide in both cell lines, and being 9.63 times more active than Adriamycin in MCF-7/ADR cell lines. The molecular docking and the topo inhibition test found that it is a selective inhibitor of topoisomerase IIα. Moreover, activation of the damage response pathway of the DNA leads to cell cycle arrest at the G0G1 phase. Furthermore, the in vivo acute toxicity of 1 in mice displayed lower toxicity than aconitine, with LD50 of 2.2 × 105 nmol/kg and only slight pathological changes in liver and lung tissue, 489 times safer than aconitine. In conclusion, compared with aconitine, 1 has more significant anti-proliferative activity against MCF-7 and MCF-7/ADR cells and greatly reduces in vivo toxicity, which suggests this kind of lipo-alkaloids is powerful and promising antitumor compounds for breast cancer.
Collapse
Affiliation(s)
- Shangxian Luan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yingying Gao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China.
| | - Li Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Qiang Wu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yunkai Hu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Shixi Liu
- School of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China
| |
Collapse
|
4
|
Carvalho RF, do Canto LM, Cury SS, Frøstrup Hansen T, Jensen LH, Rogatto SR. Drug Repositioning Based on the Reversal of Gene Expression Signatures Identifies TOP2A as a Therapeutic Target for Rectal Cancer. Cancers (Basel) 2021; 13:5492. [PMID: 34771654 PMCID: PMC8583090 DOI: 10.3390/cancers13215492] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/21/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Rectal cancer is a common disease with high mortality rates and limited therapeutic options. Here we combined the gene expression signatures of rectal cancer patients with the reverse drug-induced gene-expression profiles to identify drug repositioning candidates for cancer therapy. Among the predicted repurposable drugs, topoisomerase II inhibitors (doxorubicin, teniposide, idarubicin, mitoxantrone, and epirubicin) presented a high potential to reverse rectal cancer gene expression signatures. We showed that these drugs effectively reduced the growth of colorectal cancer cell lines closely representing rectal cancer signatures. We also found a clear correlation between topoisomerase 2A (TOP2A) gene copy number or expression levels with the sensitivity to topoisomerase II inhibitors. Furthermore, CRISPR-Cas9 and shRNA screenings confirmed that loss-of-function of the TOP2A has the highest efficacy in reducing cellular proliferation. Finally, we observed significant TOP2A copy number gains and increased expression in independent cohorts of rectal cancer patients. These findings can be translated into clinical practice to evaluate TOP2A status for targeted and personalized therapies based on topoisomerase II inhibitors in rectal cancer patients.
Collapse
Affiliation(s)
- Robson Francisco Carvalho
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
- Department of Functional and Structural Biology—Institute of Bioscience, São Paulo State University (UNESP), Botucatu 18618-689, Brazil;
| | - Luisa Matos do Canto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Sarah Santiloni Cury
- Department of Functional and Structural Biology—Institute of Bioscience, São Paulo State University (UNESP), Botucatu 18618-689, Brazil;
| | - Torben Frøstrup Hansen
- Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (T.F.H.); (L.H.J.)
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, University Hospital of Southern Denmark, 7100 Vejle, Denmark; (T.F.H.); (L.H.J.)
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark
| | - Silvia Regina Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark;
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark
| |
Collapse
|
5
|
Tarpgaard LS, Qvortrup C, Nielsen SL, Stenvang J, Detlefsen S, Brünner N, Pfeiffer P. New use for old drugs: Epirubicin in colorectal cancer. Acta Oncol 2021; 60:954-956. [PMID: 33783307 DOI: 10.1080/0284186x.2021.1904519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
| | - Camilla Qvortrup
- Department of Oncology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Signe L. Nielsen
- Faculty of Health and Medical Sciences, Institute of Drug Design and Pharmacology, University of Copenhagen, Frederiksberg, Denmark
| | - Jan Stenvang
- Faculty of Health and Medical Sciences, Institute of Drug Design and Pharmacology, University of Copenhagen, Frederiksberg, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Nils Brünner
- Faculty of Health and Medical Sciences, Institute of Drug Design and Pharmacology, University of Copenhagen, Frederiksberg, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
6
|
Ogino M, Fujii T, Nakazawa Y, Higuchi T, Koibuchi Y, Oyama T, Horiguchi J, Shirabe K. Implications of Topoisomerase (TOP1 and TOP2α) Expression in Patients With Breast Cancer. In Vivo 2021; 34:3483-3487. [PMID: 33144457 DOI: 10.21873/invivo.12188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND/AIM We evaluated the usefulness of topoisomerases (TOPs) expression as prognostic predictors in breast cancer. PATIENTS AND METHODS We retrospectively investigated sixty cases with primary breast cancer. We evaluated the tumor and non-tumor mRNA levels of TOP1 and TOP2α using quantitative reverse-transcription polymerase chain reaction. TOP1/TOP2α positivity was defined as the ratio of the mRNA expression of cancer/normal tissue of >1 for both TOP1 and TOP2α. RESULTS TOP1 and TOP2α were markedly overexpressed in breast cancer tissues compared to normal breast tissues. Of the 60 cases, 46 (76.7%) were positive for TOP1/TOP2α. The relapse-free survival was relatively shorter for patients with positive TOP1/TOP2α. There was no recurrent disease among the 14 patients who were negative for TOP1/TOP2α, whereas four of the 46 TOP1/TOP2α-positive patients had disease recurrence. CONCLUSION Negative TOP1 or TOP2α expression may be useful for predicting better prognoses in breast cancer patients.
Collapse
Affiliation(s)
- Misato Ogino
- Division of Breast and Endocrine Surgery, Graduate School of Medicine, Gunma University, Gunma, Japan.,Department of General Surgical Science, Graduate School of Medicine, Gunma University, Gunma, Japan.,Department of Breast and Endocrine Surgery, National Hospital Organization Takasaki General Medical Center, Gunma, Japan
| | - Takaaki Fujii
- Division of Breast and Endocrine Surgery, Graduate School of Medicine, Gunma University, Gunma, Japan .,Department of General Surgical Science, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Yuko Nakazawa
- Division of Breast and Endocrine Surgery, Graduate School of Medicine, Gunma University, Gunma, Japan.,Department of General Surgical Science, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Toru Higuchi
- Department of Breast Surgery, Japanese Red Cross Saitama Hospital, Saitama, Japan
| | - Yukio Koibuchi
- Department of Breast and Endocrine Surgery, National Hospital Organization Takasaki General Medical Center, Gunma, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University, Gunma, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Chiba, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Gunma, Japan
| |
Collapse
|
7
|
Zhong W, Yang Y, Zhang A, Lin W, Liang G, Ling Y, Zhong J, Yong J, Liu Z, Tian Z, Lin Q, Luo Q, Li Y, Gong C. Prognostic and predictive value of the combination of TOP2A and HER2 in node-negative tumors 2 cm or smaller (T1N0) breast cancer. Breast Cancer 2020; 27:1147-1157. [PMID: 32780321 DOI: 10.1007/s12282-020-01142-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/31/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND We aim to evaluate the prognostic and predictive value of TOP2A and HER2 expression in T1N0 breast cancer patients. METHODS 299 cases with T1N0 breast cancer were obtained from the Oncomine database (Cohort 1) and 963 of T1N0 breast cancer patients from Sun Yat-sen Memorial Hospital (Cohort 2) were retrospectively enrolled. Kaplan-Meier product was applied to estimate survival curve. Cox proportional hazard models was used to identify prognostic factors. We used PSM (propensity score matching) to balance clinicopathologic characteristics among four groups of different HER2/TOP2A status. Survival between groups and chemotherapy regimens were analyzed, before and after PSM. RESULTS In Cohort 1, we found that the group with HER2+ and higher expression of TOP2A mRNA was associated with poor breast cancer-specific survival (BCSS) compared to the group of HER2- with lower expression of TOP2A mRNA. In Cohort 2, HER2+ patients with higher TOP2A protein expression had greater risk of recurrence and distant recurrence compared to HER2- patients with lower expression of TOP2A protein. Among the patients who developed both HER2+ and higher expression of TOP2A protein and received chemotherapy, patients who received an anthracycline-based regimen had a significantly better recurrence-free survival (RFS) than those with a non-anthracycline-based regime. CONCLUSION Patients with both HER2+ and high expression level of TOP2A protein predicts poor prognosis in T1N0 breast cancer patients. Patients with double positive for TOP2A protein and HER2 may benefit from anthracycline-based regimens.
Collapse
Affiliation(s)
- Wenjing Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Yaping Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Ailing Zhang
- Department of Breast Surgery, Dongguan City People's Hospital, Dongguan, People's Republic of China
| | - Wanyi Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Gehao Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Yun Ling
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Jiajie Zhong
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Juanjuan Yong
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zihao Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Zhenluan Tian
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Qun Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Qing Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China
| | - Yangyang Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chang Gong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China. .,Department of Breast Surgery, Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
8
|
Nakazawa Y, Nakazawa S, Kurozumi S, Ogino M, Koibuchi Y, Odawara H, Oyama T, Horiguchi J, Fujii T, Shirabe K. The pathological complete response and secreted protein acidic and rich in cysteine expression in patients with breast cancer receiving neoadjuvant nab-paclitaxel chemotherapy. Oncol Lett 2020; 19:2705-2712. [PMID: 32218821 PMCID: PMC7068243 DOI: 10.3892/ol.2020.11354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 12/03/2019] [Indexed: 01/01/2023] Open
Abstract
Biomarkers that can accurately predict treatment response are required for indicating optimal neoadjuvant treatments. The current study assessed the predictive value of secreted protein acidic and rich in cysteine (SPARC) mRNA expression for the response to neoadjuvant nab-paclitaxel (nab-PTX) therapy in patients with breast cancer. It was hypothesized that SPARC expression can affect the response to albumin-bound taxanes, including nab-PTX since SPARC binds albumin with a high affinity. Pre-therapeutic specimens of core needle biopsies were analyzed from 50 patients in a phase II trial of neoadjuvant nab-PTX and the factors that were associated with a pathological complete response (pCR) were assessed. The pre-therapeutic tumor mRNA levels of chemotherapy-related proteins were quantified, including SPARC, and the correlations with post-therapeutic clinicopathological factors were assessed, including with pCR. The results demonstrated that pre-therapeutic SPARC mRNA expression was significantly higher in non-pCR patients compared with patients with pCR (92.37±55.33 vs. 56.53±30.19; P=0.027). A cutoff point of 48.5 was determined using receiver operating characteristic (ROC) curve analysis (sensitivity, 83.3%; specificity, 50.0%), and patients were classified into low and high SPARC expression groups. High SPARC expression was associated with histological grade (P=0.035), estrogen receptor expression (P=0.037), and progesterone receptor expression (P=0.002) but not with HER2 (P=0.895), and Ki-67 LI (P=0.743) expression. The results of the current study indicated that a high SPARC mRNA expression was a negative predictor of pCR following neoadjuvant nab-PTX therapy regardless of breast cancer subtype. The phase II study was conducted in accordance with the Declaration of Helsinki, and the protocol was approved by the Ethics Committee of the National Hospital Organization Takasaki General Medical Center (Registration nos. H23-9 and H23-33).
Collapse
Affiliation(s)
- Yuko Nakazawa
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Department of Breast and Endocrine Surgery, National Hospital Organization Takasaki General Medical Center, Takasaki, Gunma 370-0829, Japan.,Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Seshiru Nakazawa
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Sasagu Kurozumi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Misato Ogino
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.,Department of Breast and Endocrine Surgery, National Hospital Organization Takasaki General Medical Center, Takasaki, Gunma 370-0829, Japan
| | - Yukio Koibuchi
- Department of Breast and Endocrine Surgery, National Hospital Organization Takasaki General Medical Center, Takasaki, Gunma 370-0829, Japan
| | - Hiroki Odawara
- Department of Surgery, Toho Hospital, Midori, Gunma 376-0121, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Chiba 286-8686, Japan
| | - Takaaki Fujii
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
9
|
Abstract
DNA topoisomerases are enzymes that catalyze changes in the torsional and flexural strain of DNA molecules. Earlier studies implicated these enzymes in a variety of processes in both prokaryotes and eukaryotes, including DNA replication, transcription, recombination, and chromosome segregation. Studies performed over the past 3 years have provided new insight into the roles of various topoisomerases in maintaining eukaryotic chromosome structure and facilitating the decatenation of daughter chromosomes at cell division. In addition, recent studies have demonstrated that the incorporation of ribonucleotides into DNA results in trapping of topoisomerase I (TOP1)–DNA covalent complexes during aborted ribonucleotide removal. Importantly, such trapped TOP1–DNA covalent complexes, formed either during ribonucleotide removal or as a consequence of drug action, activate several repair processes, including processes involving the recently described nuclear proteases SPARTAN and GCNA-1. A variety of new TOP1 inhibitors and formulations, including antibody–drug conjugates and PEGylated complexes, exert their anticancer effects by also trapping these TOP1–DNA covalent complexes. Here we review recent developments and identify further questions raised by these new findings.
Collapse
Affiliation(s)
- Mary-Ann Bjornsti
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294-0019, USA
| | - Scott H Kaufmann
- Departments of Oncology and Molecular Pharmacolgy & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
10
|
Capelôa T, Benyahia Z, Zampieri LX, Blackman MCNM, Sonveaux P. Metabolic and non-metabolic pathways that control cancer resistance to anthracyclines. Semin Cell Dev Biol 2019; 98:181-191. [PMID: 31112797 DOI: 10.1016/j.semcdb.2019.05.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Anthracyclines Doxorubicin, Epirubicin, Daunorubicin and Idarubicin are used to treat a variety of tumor types in the clinics, either alone or, most often, in combination therapies. While their cardiotoxicity is well known, the emergence of chemoresistance is also a major issue accounting for treatment discontinuation. Resistance to anthracyclines is associated to the acquisition of multidrug resistance conferred by overexpression of permeability glycoprotein-1 or other efflux pumps, by altered DNA repair, changes in topoisomerase II activity, cancer stemness and metabolic adaptations. This review further details the metabolic aspects of resistance to anthracyclines, emphasizing the contributions of glycolysis, the pentose phosphate pathway and nucleotide biosynthesis, glutathione, lipid metabolism and autophagy to the chemoresistant phenotype.
Collapse
Affiliation(s)
- Tânia Capelôa
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Zohra Benyahia
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Luca X Zampieri
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marine C N M Blackman
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium.
| |
Collapse
|
11
|
Tarasov VV, Chubarev VN, Ashraf GM, Dostdar SA, Sokolov AV, Melnikova TI, Sologova SS, Grigorevskich EM, Makhmutovа A, Kinzirsky AS, Klochkov SG, Aliev G. How Cancer Cells Resist Chemotherapy: Design and Development of Drugs Targeting Protein-Protein Interactions. Curr Top Med Chem 2019; 19:394-412. [DOI: 10.2174/1568026619666190305130141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/20/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
Background:Resistance toward chemotherapeutics is one of the main obstacles on the way to effective cancer treatment. Personalization of chemotherapy could improve clinical outcome. However, despite preclinical significance, most of the potential markers have failed to reach clinical practice partially due to the inability of numerous studies to estimate the marker’s impact on resistance properly.Objective:The analysis of drug resistance mechanisms to chemotherapy in cancer cells, and the proposal of study design to identify bona fide markers.Methods:A review of relevant papers in the field. A PubMed search with relevant keywords was used to gather the data. An example of a search request: drug resistance AND cancer AND paclitaxel.Results:We have described a number of drug resistance mechanisms to various chemotherapeutics, as well as markers to underlie the phenomenon. We also proposed a model of a rational-designed study, which could be useful in determining the most promising potential biomarkers.Conclusion:Taking into account the most reasonable biomarkers should dramatically improve clinical outcome by choosing the suitable treatment regimens. However, determining the leading biomarkers, as well as validating of the model, is a work for further investigations.
Collapse
Affiliation(s)
- Vadim V. Tarasov
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Vladimir N. Chubarev
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samira A. Dostdar
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Alexander V. Sokolov
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Tatiana I. Melnikova
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Susanna S. Sologova
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Ekaterina M. Grigorevskich
- Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russian Federation
| | - Alfiya Makhmutovа
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Alexander S. Kinzirsky
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Sergey G. Klochkov
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Gjumrakch Aliev
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| |
Collapse
|
12
|
Abstract
Drug resistance is a well-known phenomenon that occurs when initially responsive to chemotherapy cancer cells become tolerant and elude further effectiveness of anticancer drugs. Based on their mechanism of action, anticancer drugs can be divided into cytotoxic-based agents and target-based agents. An important role among the therapeutics of the second group is played by drugs targeting topoisomerases, nuclear enzymes critical to DNA function and cell survival. These enzymes are cellular targets of several groups of anticancer agents which generate DNA damage in rapidly proliferating cancer cells. Drugs targeting topoisomerase I are mostly analogs of camtothecin, a natural compound isolated from the bark of a tree growing in China. Drugs targeting topoisomerase II are divided into poisons, such as anthracycline antibiotics, whose action is based on intercalation between DNA bases, and catalytic inhibitors that block topoisomerase II at different stages of the catalytic cycle. Unfortunately, chemotherapy is often limited by the induction of drug resistance. Identifying mechanisms that promote drug resistance is critical for the improvement of patient prognosis. Cancer drug resistance is a complex phenomenon that may be influenced by many factors. Here we discuss various mechanisms by which cancer cells can develop resistance to topoisomerase-directed drugs, which include enhanced drug efflux, mutations in topoisomerase genes, hypophosphorylation of topoisomerase II catalytic domain, activation of NF-κB transcription factor and drug inactivation. All these events may lead to the ineffective induction of cancer cell death. Attempts at circumventing drug resistance through the inhibition of cellular efflux pumps, use of silencing RNAs or inhibition of some important mechanisms, which can allow cancer cells to survive therapy, are also presented.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| | - Angelika Długosz
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Łódź, Poland
| |
Collapse
|
13
|
Liposomal therapies in oncology: does one size fit all? Cancer Chemother Pharmacol 2018; 82:741-755. [DOI: 10.1007/s00280-018-3668-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/09/2018] [Indexed: 12/23/2022]
|
14
|
Pajic M, Froio D, Daly S, Doculara L, Millar E, Graham PH, Drury A, Steinmann A, de Bock CE, Boulghourjian A, Zaratzian A, Carroll S, Toohey J, O'Toole SA, Harris AL, Buffa FM, Gee HE, Hollway GE, Molloy TJ. miR-139-5p Modulates Radiotherapy Resistance in Breast Cancer by Repressing Multiple Gene Networks of DNA Repair and ROS Defense. Cancer Res 2018; 78:501-515. [PMID: 29180477 DOI: 10.1158/0008-5472.can-16-3105] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/12/2017] [Accepted: 11/02/2017] [Indexed: 11/16/2022]
Abstract
Radiotherapy is essential to the treatment of most solid tumors and acquired or innate resistance to this therapeutic modality is a major clinical problem. Here we show that miR-139-5p is a potent modulator of radiotherapy response in breast cancer via its regulation of genes involved in multiple DNA repair and reactive oxygen species defense pathways. Treatment of breast cancer cells with a miR-139-5p mimic strongly synergized with radiation both in vitro and in vivo, resulting in significantly increased oxidative stress, accumulation of unrepaired DNA damage, and induction of apoptosis. Several miR-139-5p target genes were also strongly predictive of outcome in radiotherapy-treated patients across multiple independent breast cancer cohorts. These prognostically relevant miR-139-5p target genes were used as companion biomarkers to identify radioresistant breast cancer xenografts highly amenable to sensitization by cotreatment with a miR-139-5p mimetic.Significance: The microRNA described in this study offers a potentially useful predictive biomarker of radiosensitivity in solid tumors and a generally applicable druggable target for tumor radiosensitization. Cancer Res; 78(2); 501-15. ©2017 AACR.
Collapse
Affiliation(s)
- Marina Pajic
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Danielle Froio
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Sheridan Daly
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Louise Doculara
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Ewan Millar
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Anatomical Pathology, South Eastern Area Laboratory Service (SEALS), St George Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Peter H Graham
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Alison Drury
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Angela Steinmann
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Charles E de Bock
- Laboratory for the Molecular Biology of Leukemia, Center for Human Genetics, KU Leuven and Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Alice Boulghourjian
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Anaiis Zaratzian
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Susan Carroll
- The Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Joanne Toohey
- The Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Sandra A O'Toole
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Adrian L Harris
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Francesca M Buffa
- Growth Factor Group, Cancer Research UK, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Harriet E Gee
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- The Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Georgina E Hollway
- Cancer Research Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Timothy J Molloy
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.
- St Vincent's Centre for Applied Medical Research, Darlinghurst, New South Wales, Australia
| |
Collapse
|
15
|
Weerts MJA, Hollestelle A, Sieuwerts AM, Foekens JA, Sleijfer S, Martens JWM. Low Tumor Mitochondrial DNA Content Is Associated with Better Outcome in Breast Cancer Patients Receiving Anthracycline-Based Chemotherapy. Clin Cancer Res 2017; 23:4735-4743. [PMID: 28420722 DOI: 10.1158/1078-0432.ccr-17-0032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/20/2017] [Accepted: 04/11/2017] [Indexed: 11/16/2022]
Abstract
Purpose: In this study, we aimed to explore whether low levels of mitochondrial DNA (mtDNA) content in the primary tumor could predict better outcome for breast cancer patients receiving anthracycline-based therapies. We hypothesized that tumor cells with low mtDNA content are more susceptible to mitochondrial damage induced by anthracyclines, and thus are more susceptible to anthracycline treatment.Experimental Design: We measured mtDNA content by a qPCR approach in 295 primary breast tumor specimens originating from two well-defined cohorts: 174 lymph node-positive patients who received adjuvant chemotherapy and 121 patients with advanced disease who received chemotherapy as first-line palliative treatment. The chemotherapy regimens given were either anthracycline-based (FAC/FEC) or methotrexate-based (CMF).Results: In both the adjuvant and advanced settings, we observed increased benefit for patients with low mtDNA content in their primary tumor, but only when treated with FAC/FEC. In multivariable Cox regression analysis for respectively distant metastasis-free survival and progression-free survival, the HR for the FAC/FEC-treated mtDNA low group in the adjuvant setting was 0.46 [95% confidence interval (CI), 0.24-0.89; P = 0.020] and in the advanced setting 0.49 (95% CI, 0.27-0.90; P = 0.022) compared with the FAC/FEC-treated mtDNA high group. We did not observe these associations in the patients treated with CMF.Conclusions: In our two study cohorts, breast cancer patients with low mtDNA content in their primary tumor had better outcome from anthracycline-containing chemotherapy. The frequently observed decrease in mtDNA content in primary breast tumors may be exploited by guiding chemotherapeutic regimen decision making. Clin Cancer Res; 23(16); 4735-43. ©2017 AACR.
Collapse
Affiliation(s)
- Marjolein J A Weerts
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Antoinette Hollestelle
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John A Foekens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
16
|
Hou JY, Baptiste C, Hombalegowda RB, Tergas AI, Feldman R, Jones NL, Chatterjee-Paer S, Bus-Kwolfski A, Wright JD, Burke WM. Vulvar and vaginal melanoma: A unique subclass of mucosal melanoma based on a comprehensive molecular analysis of 51 cases compared with 2253 cases of nongynecologic melanoma. Cancer 2016; 123:1333-1344. [PMID: 28026870 DOI: 10.1002/cncr.30473] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND Optimal treatments for vulvar and vaginal melanomas (VVMs) have not been identified. Herein, the authors compare molecular profiles between VVM and nongynecologic melanoma (NGM) subtypes with the objective of identifying novel, targetable biomarkers. METHODS In total, 2304 samples of malignant melanoma that were submitted to Caris Life Sciences between 2009 and 2015 were reviewed. In situ hybridization and immunohistochemistry were used to assess copy numbers and protein expression of selected genes. Sequenced variants were analyzed using a proprietary cancer panel. RESULTS In total, 51 VVMs (14 vaginal and 37 vulvar melanomas) were compared with 2253 malignant NGMs, including 2127 cutaneous, 105 mucosal, and 21 acral melanomas. In VVMs, B-Raf proto-oncogene serine/threonine kinase (BRAF) was the most frequently mutated gene (26%) compared with 8.3% of mucosal NGMs (P = .008). In BRAF-mutated tumors, fewer VVMs (50%), compared with NGMs (82.1%), had a variant within the valine codon 600 (V600) domain. The KIT mutation rate was highest in VVMs (22%) compared with 3% in cutaneous (P < .001) and 8.8% in mucosal (P = .05) melanoma subtypes. NRAS mutations were rare in VVMs compared with cutaneous (25.9%; P = .009) and acral (40.6%; P = .002) melanoma subtypes. PD-L1 (56%) and PD-1 (75%) were frequently expressed in VVM, whereas PI3KCA pathway mutations and estrogen receptor/progesterone receptor expression were rare. Compared with VVMs that had KIT mutations, wild-type KIT VVMs were more likely to express molecular markers suggestive of platinum resistance (ERCC1), alkylating sensitivity (MGMT), and anthracycline sensitivity (TOP2A). CONCLUSIONS The unique molecular features of VVM render this disease a distinct subtype of melanoma. Gene-based molecular therapy and immunotherapies may be promising and should be evaluated in clinical trials. Cancer 2017;123:1333-1344. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- June Y Hou
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Caitlin Baptiste
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Radhika Bangalore Hombalegowda
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Ana I Tergas
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Rebecca Feldman
- Medical Affairs Department, Caris Life Sciences, Phoenix, Arizona
| | - Nathaniel L Jones
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Sudeshna Chatterjee-Paer
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Ama Bus-Kwolfski
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Jason D Wright
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - William M Burke
- Department of Obstetrics and Gynecology and Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| |
Collapse
|
17
|
Hansen SN, Ehlers NS, Zhu S, Thomsen MBH, Nielsen RL, Liu D, Wang G, Hou Y, Zhang X, Xu X, Bolund L, Yang H, Wang J, Moreira J, Ditzel HJ, Brünner N, Schrohl AS, Stenvang J, Gupta R. The stepwise evolution of the exome during acquisition of docetaxel resistance in breast cancer cells. BMC Genomics 2016; 17:442. [PMID: 27277198 PMCID: PMC4899892 DOI: 10.1186/s12864-016-2749-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/17/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Resistance to taxane-based therapy in breast cancer patients is a major clinical problem that may be addressed through insight of the genomic alterations leading to taxane resistance in breast cancer cells. In the current study we used whole exome sequencing to discover somatic genomic alterations, evolving across evolutionary stages during the acquisition of docetaxel resistance in breast cancer cell lines. RESULTS Two human breast cancer in vitro models (MCF-7 and MDA-MB-231) of the step-wise acquisition of docetaxel resistance were developed by exposing cells to 18 gradually increasing concentrations of docetaxel. Whole exome sequencing performed at five successive stages during this process was used to identify single point mutational events, insertions/deletions and copy number alterations associated with the acquisition of docetaxel resistance. Acquired coding variation undergoing positive selection and harboring characteristics likely to be functional were further prioritized using network-based approaches. A number of genomic changes were found to be undergoing evolutionary selection, some of which were likely to be functional. Of the five stages of progression toward resistance, most resistance relevant genomic variation appeared to arise midway towards fully resistant cells corresponding to passage 31 (5 nM docetaxel) for MDA-MB-231 and passage 16 (1.2 nM docetaxel) for MCF-7, and where the cells also exhibited a period of reduced growth rate or arrest, respectively. MCF-7 cell acquired several copy number gains on chromosome 7, including ABC transporter genes, including ABCB1 and ABCB4, as well as DMTF1, CLDN12, CROT, and SRI. For MDA-MB-231 numerous copy number losses on chromosome X involving more than 30 genes was observed. Of these genes, CASK, POLA1, PRDX4, MED14 and PIGA were highly prioritized by the applied network-based gene ranking approach. At higher docetaxel concentration MCF-7 subclones exhibited a copy number loss in E2F4, and the gene encoding this important transcription factor was down-regulated in MCF-7 resistant cells. CONCLUSIONS Our study of the evolution of acquired docetaxel resistance identified several genomic changes that might explain development of docetaxel resistance. Interestingly, the most relevant resistance-associated changes appeared to originate midway through the evolution towards fully resistant cell lines. Our data suggest that no single genomic event sufficiently predicts resistance to docetaxel, but require genomic alterations affecting multiple pathways that in concert establish the final resistance stage.
Collapse
Affiliation(s)
- Stine Ninel Hansen
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Natasja Spring Ehlers
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet building 208, DK-2800, Lyngby, Denmark
| | - Shida Zhu
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Mathilde Borg Houlberg Thomsen
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Department of Molecular Medicine, Aarhus University Hospital, Brendstrupgaardsvej 100, DK-8200, Aarhus N, Denmark
| | - Rikke Linnemann Nielsen
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet building 208, DK-2800, Lyngby, Denmark
| | - Dongbing Liu
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Guangbiao Wang
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Yong Hou
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Xiuqing Zhang
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Xun Xu
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Lars Bolund
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Department of Biomedicine, Aarhus University, Bartholins Allé 6, DK-8000, Aarhus C, Denmark
| | - Huanming Yang
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China
| | - Jun Wang
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen, 518083, China.,Macau University of Science and Technology, Avenida Wai long, Taipa, Macau, 999078, China.,Department of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.,Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloews Vej 25, DK-5000, Odense, Denmark.,Department of Oncology, Odense University Hospital, Sdr. Boulevard 29, DK-5000, Odense, Denmark
| | - Jose Moreira
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Henrik J Ditzel
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen, Denmark.,Princess Al Jawhara Albrahim Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nils Brünner
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Anne-Sofie Schrohl
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Jan Stenvang
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark. .,Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology, University of Copenhagen, Strandboulevarden 49, DK-2100, Copenhagen, Denmark.
| | - Ramneek Gupta
- Sino Danish Breast Cancer Research Center, Copenhagen, Denmark. .,Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet building 208, DK-2800, Lyngby, Denmark.
| |
Collapse
|
18
|
Harris LN, Ismaila N, McShane LM, Andre F, Collyar DE, Gonzalez-Angulo AM, Hammond EH, Kuderer NM, Liu MC, Mennel RG, Van Poznak C, Bast RC, Hayes DF. Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2016; 34:1134-50. [PMID: 26858339 PMCID: PMC4933134 DOI: 10.1200/jco.2015.65.2289] [Citation(s) in RCA: 592] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To provide recommendations on appropriate use of breast tumor biomarker assay results to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer. METHODS A literature search and prospectively defined study selection sought systematic reviews, meta-analyses, randomized controlled trials, prospective-retrospective studies, and prospective comparative observational studies published from 2006 through 2014. Outcomes of interest included overall survival and disease-free or recurrence-free survival. Expert panel members used informal consensus to develop evidence-based guideline recommendations. RESULTS The literature search identified 50 relevant studies. One randomized clinical trial and 18 prospective-retrospective studies were found to have evaluated the clinical utility, as defined by the guideline, of specific biomarkers for guiding decisions on the need for adjuvant systemic therapy. No studies that met guideline criteria for clinical utility were found to guide choice of specific treatments or regimens. RECOMMENDATIONS In addition to estrogen and progesterone receptors and human epidermal growth factor receptor 2, the panel found sufficient evidence of clinical utility for the biomarker assays Oncotype DX, EndoPredict, PAM50, Breast Cancer Index, and urokinase plasminogen activator and plasminogen activator inhibitor type 1 in specific subgroups of breast cancer. No biomarker except for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 was found to guide choices of specific treatment regimens. Treatment decisions should also consider disease stage, comorbidities, and patient preferences.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents, Hormonal/therapeutic use
- Biomarkers, Tumor/analysis
- Breast Neoplasms/chemistry
- Breast Neoplasms/drug therapy
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/chemistry
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Chemotherapy, Adjuvant
- Clinical Decision-Making/methods
- Comorbidity
- Disease-Free Survival
- Evidence-Based Medicine
- Female
- Humans
- Neoplasm Staging
- Plasminogen Activator Inhibitor 1/analysis
- Predictive Value of Tests
- Randomized Controlled Trials as Topic
- Receptor, ErbB-2/analysis
- Receptors, Estrogen/analysis
- Receptors, Progesterone/analysis
- Reproducibility of Results
- Survival Analysis
- Urokinase-Type Plasminogen Activator/analysis
Collapse
Affiliation(s)
- Lyndsay N Harris
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Nofisat Ismaila
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI.
| | - Lisa M McShane
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Fabrice Andre
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Deborah E Collyar
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Ana M Gonzalez-Angulo
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Elizabeth H Hammond
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Nicole M Kuderer
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Minetta C Liu
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Robert G Mennel
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Catherine Van Poznak
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Robert C Bast
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Daniel F Hayes
- Lyndsay N. Harris, Case Western Reserve University, Cleveland, OH; Nofisat Ismaila, American Society of Clinical Oncology, Alexandria, VA; Lisa M. McShane, National Cancer Institute, Bethesda, MD; Fabrice Andre, Institute Gustave Roussy, Paris, France; Deborah E. Collyar, Patient Advocates in Research; Elizabeth H. Hammond, University of Utah and Intermountain Health Care, Salt Lake City, UT; Ana M. Gonzalez-Angulo and Robert C. Bast, The University of Texas MD Anderson Cancer Center, Houston; Robert G.Mennel, Baylor University Medical Center and Texas Oncology PA, Dallas, TX; Nicole M. Kuderer, University of Washington Medical Center, Seattle, WA; Minetta C. Liu, Mayo Clinic College of Medicine, Rochester, MN; and Catherine Van Poznak and Daniel F. Hayes, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| |
Collapse
|
19
|
Tarpgaard LS, Qvortrup C, Nygård SB, Nielsen SL, Andersen DR, Jensen NF, Stenvang J, Detlefsen S, Brünner N, Pfeiffer P. A phase II study of Epirubicin in oxaliplatin-resistant patients with metastatic colorectal cancer and TOP2A gene amplification. BMC Cancer 2016; 16:91. [PMID: 26867764 PMCID: PMC4750171 DOI: 10.1186/s12885-016-2124-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/03/2016] [Indexed: 12/28/2022] Open
Abstract
ᅟ The overall purpose of this study is to provide proof of concept for introducing the anthracycline epirubicin as an effective, biomarker-guided treatment for metastatic colorectal cancer (mCRC) patients who are refractory to treatment with oxaliplatin-based chemotherapy and have TOP2A gene amplification in their tumor cells. Background Epirubicin is an anthracycline that targets DNA topoisomerase 2-α enzyme encoded by the TOP2A gene. It is used for treatment of several malignancies, but currently not in CRC. TOP2A gene amplifications predict improved efficacy of epirubicin in patients with breast cancer and thus could be an alternative option for patients with CRC and amplified TOP2A gene. We have previously analysed the frequency of TOP2A gene aberrations in CRC and found that 46.6 % of these tumors had TOP2A copy gain and 2.0 % had loss of TOP2A when compared to adjacent normal tissue. The TOP2A gene is located on chromosome 17 and when the TOP2A/CEN-17 ratio was applied to identify tumors with gene loss or amplifications, 10.5 % had a ratio ≥ 1.5 consistent with gene amplification and 2.6 % had a ratio ≤ 0.8 suggesting gene deletions. Based on these observations and the knowledge gained from treatment of breast cancer patients, we have initiated a prospective clinical, phase II protocol using epirubicin (90 mg/m2 iv q 3 weeks) in mCRC patients, who are refractory to treatment with oxaliplatin. Methods/Design The study is an open label, single arm, phase II study, investigating the efficacy of epirubicin in patients with oxaliplatin refractory mCRC and with a cancer cell TOP2A/CEN-17 ratio ≥ 1.5. TOP2A gene amplification measured by fluorescence in situ hybridization. A total of 25 evaluable patients (15 + 10 in two steps) will be included (Simon’s two-stage minimax design). Every nine weeks, response is measured by computed tomography imaging and evaluated according to RECIST 1.1. The primary end-point of the study is progression-free survival. Trial registration Eudract no. 2013-001648-79.
Collapse
Affiliation(s)
- Line S Tarpgaard
- Department of Oncology, Odense University Hospital, Odense, Denmark.
| | - Camilla Qvortrup
- Department of Oncology, Odense University Hospital, Odense, Denmark.
| | - Sune B Nygård
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark. .,Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Signe L Nielsen
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark.
| | - Diana R Andersen
- Department of Oncology, Odense University Hospital, Odense, Denmark.
| | - Niels Frank Jensen
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark.
| | - Jan Stenvang
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark.
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark.
| | - Nils Brünner
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, University of Copenhagen, Frederiksberg, Denmark.
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
20
|
Nogi H, Uchida K, Kamio M, Kato K, Toriumi Y, Akiba T, Morikawa T, Suzuki M, Kobayashi T, Takeyama H. Triple-negative breast cancer exhibits a favorable response to neoadjuvant chemotherapy independent of the expression of topoisomerase IIα. Mol Clin Oncol 2015; 4:383-389. [PMID: 26998288 DOI: 10.3892/mco.2015.719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/23/2015] [Indexed: 12/31/2022] Open
Abstract
The present study retrospectively analyzed the utility of topoisomerase IIα expression as a prognostic marker to predict the neoadjuvant chemotherapeutic response and survival among different breast cancer subtypes. The patients were subtyped and the expression of topoisomerase IIα was determined using immunohistochemistry. All patients (n=147) received an anthracycline-containing regimen preoperatively, and 139 (95%) patients also received docetaxel. Of the 147 patients, 25 (17%) were triple-negative and 20 (17%) were human epidermal growth factor receptor 2 (HER2)-positive. Among these subtypes, a significantly higher a rate (P<0.0001) and higher incidence of topoisomerase IIα expression (P=0.036) were observed compared with that in the hormone receptor-positive and HER2-negative breast cancer types. However, the expression of topoisomerase IIα revealed no correlation with the treatment response or survival in any of the subtypes. Therefore, these results indicated that the favorable response to anthracycline-containing chemotherapy among triple-negative and HER2-positive breast cancer was independent of the expression of topoisomerase IIα.
Collapse
Affiliation(s)
- Hiroko Nogi
- Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Ken Uchida
- Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Makiko Kamio
- Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Kumiko Kato
- Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Yasuo Toriumi
- Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Tadashi Akiba
- Department of Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Toshiaki Morikawa
- Department of Thoracic Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Masaaki Suzuki
- Department of Pathology, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Tadashi Kobayashi
- Department of Medical Oncology and Hematology, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroshi Takeyama
- Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
21
|
Sønderstrup IMH, Nygård SB, Poulsen TS, Linnemann D, Stenvang J, Nielsen HJ, Bartek J, Brünner N, Nørgaard P, Riis L. Topoisomerase-1 and -2A gene copy numbers are elevated in mismatch repair-proficient colorectal cancers. Mol Oncol 2015; 9:1207-17. [PMID: 25777966 DOI: 10.1016/j.molonc.2015.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/19/2015] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Topoisomerase 1 (TOP1) and 2A (TOP2A) are potential predictive biomarkers for irinotecan and anthracycline treatment, respectively, in colorectal cancer (CRC), and we have recently reported a high frequency of gene gain of the TOP1 and TOP2A genes in CRC. Furthermore, Mismatch Repair (MMR) subtypes of CRC have been associated with benefit from adjuvant chemotherapy of primary CRC. Given the involvement of the topoisomerase enzymes in DNA replication and repair, we raised the hypothesis that an association may exist between TOP gene copy numbers and MMR proficiency/deficiency in CRC. MATERIAL AND METHODS Test cohort: FISH analysis with an in-house TOP1/CEN20 probe mix and a commercially available TOP2A/CEN17 (Dako, Glostrup, Denmark) probe mix was performed on archival formalin fixed paraffin embedded (FFPE) tissue samples from 18 patients with proficient MMR (pMMR) CRC and 18 patients with deficient MMR (dMMR) CRC. TOP1 and TOP2A gene copy numbers and their ratios per nucleus were correlated with MMR status using the Mann-Whitney test. Validation cohort: FFPE samples from 154 patients with primary stage III CRC (originally included in the RANX05 study) were classified according to MMR status by immunohistochemical analysis using validated antibodies for MLH1, MLH2, MSH6 and PMS2, and information on TOP1, CEN20, TOP2A and CEN17 status was previously published for this cohort. RESULTS The observed TOP1 gene copy numbers in the 36 CRC test cohort were significantly greater (p < 0.01) in the pMMR subgroup (mean: 3.84, SD: 2.03) than in the dMMR subgroup (mean: 1.50, SD: 0.12). Similarly, the TOP2A copy numbers were significantly greater (p < 0.01) in the pMMR subgroup (mean: 1.99, SD: 0.52) than in the dMMR subgroup (mean: 1.52, SD: 0.10). These findings were confirmed in the validation cohort, where in the pMMR subgroup 51% had ≥2 extra TOP1 copies per cell, while all tumors classified as dMMR had diploid TOP1 status and mean TOP2A copy numbers were 2.30 (SD: 1.36) and 1.80 (SD: 0.31) (p = 0.01) in the pMMR subgroup vs. dMMR subgroup, respectively. DISCUSSION AND CONCLUSION Our results show that TOP1 and TOP2A gene copy numbers are increased in the pMMR subgroup. We propose that this preference may reflect a selective pressure to gain and/or maintain the gained extra copies of topoisomerase genes whose products are required to cope with high replication stress present in the pMMR tumors, thereby providing a survival advantage selectively in pMMR tumors. Future studies should test this concept and explore potential differences between pMMR and dMMR tumors in response to Top1 and Top2 inhibitors.
Collapse
Affiliation(s)
| | - Sune Boris Nygård
- Section of Molecular Disease Biology, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark.
| | - Tim Svenstrup Poulsen
- Department of Pathology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Dorte Linnemann
- Department of Pathology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Jan Stenvang
- Section of Molecular Disease Biology, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark.
| | - Hans Jørgen Nielsen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Kettegårds Allé 30, DK-2650 Hvidovre, Denmark.
| | - Jiri Bartek
- Danish Cancer Research Center, The Danish Cancer Society, Strandboulevarden 49, DK-2100 Copenhagen Ø, Denmark; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Tr Svobody 8, 771 26 Olomouc, Czech Republic.
| | - Nils Brünner
- Section of Molecular Disease Biology, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark.
| | - Peter Nørgaard
- Department of Pathology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Lene Riis
- Department of Pathology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| |
Collapse
|
22
|
Susini T, Berti B, Carriero C, Tavella K, Nori J, Vanzi E, Molino C, Di Tommaso M, Santini M, Saladino V, Bianchi S. Topoisomerase II alpha and TLE3 as predictive markers of response to anthracycline and taxane-containing regimens for neoadjuvant chemotherapy in breast cancer. Onco Targets Ther 2014; 7:2111-20. [PMID: 25484594 PMCID: PMC4240193 DOI: 10.2147/ott.s71646] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Anthracyclines and taxanes are considered the standard for neoadjuvant chemotherapy of breast cancer, although they are often associated with serious side effects and wide variability of individual response. In this study, we analyzed the value of topoisomerase II alpha (TOP2A) and transducin-like enhancer of split 3 (TLE3) as predictive markers of response to therapy with anthracyclines and taxanes. Materials and methods TOP2A and TLE3 protein expressions were evaluated using immunohistochemistry on 28 samples, obtained by core needle biopsy in patients with locally advanced breast carcinoma, subsequently subjected to epirubicin- and paclitaxel-based neoadjuvant chemotherapy. The immunohistochemical staining was correlated with the clinical response measured by the tumor size reduction evaluated by breast magnetic resonance imaging, prior and after chemotherapy, and by pathologic evaluation of the surgical specimen. Results Neoadjuvant chemotherapy achieved a size reduction in 26/28 tumors (92.9%), with an average percentage decrease of 45.6%. A downstaging was achieved in 71.4% of the cases of locally advanced carcinoma. TOP2A positivity was correlated with a greater reduction in tumor diameter (P=0.06); negative staining for TLE3 was predictive of a better response to neoadjuvant chemotherapy (P=0.07). A higher reduction in tumor diameter (P=0.03) was also found for tumors that were concurrently TLE3-negative and TOP2A-positive. Conclusion TOP2A and TLE3 showed a correlation with response to neoadjuvant chemotherapy. While TOP2A is a well-known marker of response to anthracyclines-based chemotherapy, TLE3 is a new putative predictor of response to taxanes. Data from the current study suggest that TOP2A and TLE3 warrant further investigation in a larger series as predictors of response to neoadjuvant chemotherapy for locally advanced breast carcinoma.
Collapse
Affiliation(s)
- Tommaso Susini
- Department of Health Science, Gynecology Section, University of Florence, Italy
| | - Barbara Berti
- Department of Health Science, Gynecology Section, University of Florence, Italy
| | - Carlo Carriero
- Department of Health Science, Gynecology Section, University of Florence, Italy
| | - Ketty Tavella
- Department of Health Science, Chemotherapy Section, University of Florence, Italy
| | - Jacopo Nori
- Diagnostic Senology Unit, Azienda Ospedaliera-Universitaria Careggi, Florence, Italy
| | - Ermanno Vanzi
- Diagnostic Senology Unit, Azienda Ospedaliera-Universitaria Careggi, Florence, Italy
| | - Cecilia Molino
- Department of Health Science, Gynecology Section, University of Florence, Italy
| | | | - Marco Santini
- Department of Health Science, Gynecology Section, University of Florence, Italy
| | - Valeria Saladino
- Department of Surgery and Translational Medicine, Pathology Unit, University of Florence, Italy
| | - Simonetta Bianchi
- Department of Surgery and Translational Medicine, Pathology Unit, University of Florence, Italy
| |
Collapse
|
23
|
Nygård SB, Christensen IJ, Smith DH, Nielsen SL, Jensen NF, Nielsen HJ, Vainer B, Brünner N. Underpinning the repurposing of anthracyclines towards colorectal cancer: assessment of topoisomerase II alpha gene copy number alterations in colorectal cancer. Scand J Gastroenterol 2013; 48:1436-43. [PMID: 24138107 DOI: 10.3109/00365521.2013.848230] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE. We propose a repurposing strategy where anthracyclines are reintroduced to a subgroup of patients with metastatic colorectal cancer with the highest likelihood of response. In breast cancer, DNA topoisomerase II alpha gene (TOP2A) alterations predict incremental benefit of anthracyclines, but this association has not been investigated in colorectal cancer. Frequency analysis of TOP2A gene alterations in colorectal cancer and the association with prognosis are evaluated and the challenges of using a TOP2A/CEN-17 FISH probe combination are addressed. MATERIAL AND METHODS. Formalin-fixed, paraffin-embedded material from 154 stage III colorectal cancer patients included in the RANX05 clinical trial was retrospectively assessed for TOP2A gene alterations using FISH. The TOP2A/CEN-17 ratio as well as the TOP2A gene copy number alone was used to define gene alterations and associations between gene status and outcomes were analyzed. RESULTS. TOP2A gene gain was a frequent finding with 9.8 % having a total of ≥4 TOP2A copies per cell. According to the TOP2A/CEN-17 ratio, 10.5 % had TOP2A gene gain. Polysomy or gain of the centromere region of chromosome-17 was not as frequent as reported in breast cancer. No prognostic characteristic of TOP2A was identified. CONCLUSION. TOP2A gene gain is present in numbers relevant to identify a subgroup of patients who may benefit from anthracycline therapy. Based on the present findings, we will initiate a prospective clinical trial designed to evaluate this hypothesis in patients with metastatic colorectal cancer who have failed 5-fluorouracil and oxaliplatin chemotherapy.
Collapse
Affiliation(s)
- Sune Boris Nygård
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Section for Molecular Disease Biology , Copenhagen Ø , Denmark
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Yu Y, Xiang H, He XM, Yang HJ, Zong XY. Predictive factors determining neoadjuvant chemotherapy outcomes in breast cancer - a single center experience. Asian Pac J Cancer Prev 2013; 14:2401-6. [PMID: 23725148 DOI: 10.7314/apjcp.2013.14.4.2401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
From January 1, 2008 to March 31, 2010, 101 patients with stage II-III breast cancer were enrolled in this study and subjected to an anthracycline-based neoadjuvant chemotherapy regimen with or without docetaxel. Surgery was performed after 2-6 cycles of chemotherapy, and the clinical response was determined by pathological and histochemical assessments. The clinical response rate, as indicated by complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD), were 6.9, 52.5, 36.6, and 4.0%, respectively. A multivariable correlation analysis indicated that the overall clinical response rate correlated with the number of metastatic lymph nodes, number of chemotherapy cycles, and vessel invasion status. Importantly, the CR rate was only associated with the number of chemotherapy cycles. Nonparametric tests failed to detect a correlation between HER2 or Topo IIα status and clinical response to neoadjuvant chemotherapy in these patients. When they were stratified by HER2 or HR status, for HER2-positive patients the CR rate was associated with vessel invasion and Topo IIα status. Based on our findings, we propose that HR, HER-2 and Topo IIα are not putative predictive biomarkers of chemotherapy outcome for breast cancer patients. Topo IIα expression level was only inversely correlated with CR rate among HR-positive patients. Importantly, the achievement of CR was largely related to the number of chemotherapy cycles.
Collapse
Affiliation(s)
- Yang Yu
- Department of Breast Surgery, Zhejiang Cancer Hospital, China
| | | | | | | | | |
Collapse
|
25
|
Ganapathi RN, Ganapathi MK. Mechanisms regulating resistance to inhibitors of topoisomerase II. Front Pharmacol 2013; 4:89. [PMID: 23914174 PMCID: PMC3729981 DOI: 10.3389/fphar.2013.00089] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 06/24/2013] [Indexed: 11/13/2022] Open
Abstract
Inhibitors of topoisomerase II (topo II) are clinically effective in the management of hematological malignancies and solid tumors. The efficacy of anti-tumor drugs targeting topo II is often limited by resistance and studies with in vitro cell culture models have provided several insights on potential mechanisms. Multidrug transporters that are involved in the efflux and consequently reduced cytotoxicity of diverse anti-tumor agents suggest that they play an important role in resistance to clinically active drugs. However, in clinical trials, modulating the multidrug-resistant phenotype with agents that inhibit the efflux pump has not had an impact. Since reduced drug accumulation per se is insufficient to explain tumor cell resistance to topo II inhibitors several studies have focused on characterizing mechanisms that impact on DNA damage mediated by drugs that target the enzyme. Mammalian topo IIα and topo IIβ isozymes exhibit similar catalytic, but different biologic, activities. Whereas topo IIα is associated with cell division, topo IIβ is involved in differentiation. In addition to site specific mutations that can affect drug-induced topo II-mediated DNA damage, post-translation modification of topo II primarily by phosphorylation can potentially affect enzyme-mediated DNA damage and the downstream cytotoxic response of drugs targeting topo II. Signaling pathways that can affect phosphorylation and changes in intracellular calcium levels/calcium dependent signaling that can regulate site-specific phosphorylation of topoisomerase have an impact on downstream cytotoxic effects of topo II inhibitors. Overall, tumor cell resistance to inhibitors of topo II is a complex process that is orchestrated not only by cellular pharmacokinetics but more importantly by enzymatic alterations that govern the intrinsic drug sensitivity.
Collapse
Affiliation(s)
- Ram N Ganapathi
- Levine Cancer Institute, Carolinas HealthCare System Charlotte, NC, USA
| | | |
Collapse
|
26
|
Wang Y, Liu Y, Du Y, Yin W, Lu J. The predictive role of phosphatase and tensin homolog (PTEN) loss, phosphoinositol-3 (PI3) kinase (PIK3CA) mutation, and PI3K pathway activation in sensitivity to trastuzumab in HER2-positive breast cancer: a meta-analysis. Curr Med Res Opin 2013; 29:633-42. [PMID: 23574264 DOI: 10.1185/03007995.2013.794775] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Phosphatase and tensin homolog (PTEN) loss or activating mutations of phosphoinositol-3 (PI3) kinase (PIK3CA) may be related to trastuzumab resistance in in vitro studies; however, this issue in clinical studies is controversial. Therefore, we conducted a meta-analysis to assess the association between PTEN loss, PIK3CA mutation and the efficacy of trastuzumab-based treatment in HER2-positive breast cancer patients. METHODS A computerized search was performed through the PubMed database, the online proceedings of the American Society of Clinical Oncology Annual Meetings, the San Antonio Breast Cancer Symposium and the International St. Gallen Breast Cancer Conference. Ten eligible studies including 1889 cases were identified. RESULTS In HER2-positive locally advanced breast cancer patients, neither PTEN loss, PIK3CA mutation nor PI3K activation was associated with the response rate of trastuzumab-based neoadjuvant treatment (PTEN loss: RR = 0.687, 95% CI: 0.439-1.074, P = 0.099; PIK3CA mutation: RR = 1.114, 95% CI: 0.453-2.735, P = 0.814; PI3K activation: RR = 0.787, 95% CI: 0.417-1.484, P = 0.459; RR = 0.772, 95% CI: 0.387-1.539, P = 0.462). In HER2-positive early stage breast cancer patients, PTEN loss was not associated with the disease-free survival (DFS) rate of trastuzumab-based adjuvant treatment (HR = 1.096, 95% CI: 0.706-1.700, P = 0.684). In HER2-positive recurrent or metastatic breast cancer patients, PTEN loss was significantly correlated with poorer efficacy of trastuzumab-based salvage treatment (RR = 0.682, 95% CI: 0.550-0.846, P = 0.000). CONCLUSIONS In HER2-positive recurrent or metastatic breast cancer patients PTEN loss might indicate resistance to trastuzumab-based salvage treatment. Due to the small sample size and the considerable heterogeneity in the chemotherapy treatment regimens, further research is needed to clarify the association between PTEN loss, PIK3CA mutation and the efficacy of trastuzumab-based treatment in neoadjuvant and adjuvant settings.
Collapse
Affiliation(s)
- Yaohui Wang
- Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
27
|
The HER2 amplicon in breast cancer: Topoisomerase IIA and beyond. Biochim Biophys Acta Rev Cancer 2013; 1836:146-57. [PMID: 23628726 DOI: 10.1016/j.bbcan.2013.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 04/17/2013] [Accepted: 04/19/2013] [Indexed: 12/20/2022]
Abstract
HER2 gene amplification is observed in about 15% of breast cancers. The subgroup of HER2-positive breast cancers appears to be heterogeneous and presents complex patterns of gene amplification at the locus on chromosome 17q12-21. The molecular variations within the chromosome 17q amplicon and their clinical implications remain largely unknown. Besides the well-known TOP2A gene encoding Topoisomerase IIA, other genes might also be amplified and could play functional roles in breast cancer development and progression. This review will focus on the current knowledge concerning the HER2 amplicon heterogeneity, its clinical and biological impact and the pitfalls associated with the evaluation of gene amplifications at this locus, with particular attention to TOP2A and the link between TOP2A and anthracycline benefit. In addition it will discuss the clinical and biological implications of the amplification of ten other genes at this locus (MED1, STARD3, GRB7, THRA, RARA, IGFPB4, CCR7, KRT20, KRT19 and GAST) in breast cancer.
Collapse
|