1
|
Kanu GA, Mouselly A, Mohamed AA. Foundations and applications of computational genomics. DEEP LEARNING IN GENETICS AND GENOMICS 2025:59-75. [DOI: 10.1016/b978-0-443-27574-6.00007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Sun Y, Wang D, Yuan C, Lang X, Fu S. Lapatinib: A Potential Therapeutic Agent for Colon Cancer Targeting Ferroptosis. Anticancer Agents Med Chem 2025; 25:114-123. [PMID: 39238394 DOI: 10.2174/0118715206327756240830062531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Colon cancer poses a significant threat to the lives of several patients, impacting their quality of life, thus necessitating its urgent treatment. Lapatinib, a new generation of targeted anti-tumor drugs for clinical application, has yet to be studied for its molecular mechanisms in treating colon cancer. OBJECTIVES This study aimed to uncover the underlying molecular mechanisms through which lapatinib exerts its therapeutic effects in colon cancer treatment. METHODS We accessed pertinent data on patients with colon cancer from the Cancer Genome Atlas (TCGA) database and performed bioinformatics analysis to derive valuable insights. The cell counting kit-8 (CCK8) assay was employed to assess whether lapatinib has a potential inhibitory effect on the growth and proliferation of HT- 29 cells. Additionally, we employed western blot and real-time quantitative polymerase chain reaction methods to investigate whether lapatinib regulates the expression of the ferroptosis-associated protein GPX4 in HT-29 cells. Furthermore, we utilized specific assay kits to measure the levels of reactive oxygen species (ROS) and malondialdehyde in HT-29 cells treated with lapatinib, aiming to elucidate the precise pattern of cell damage induced by this compound. RESULTS GPX4 exhibited high expression levels in tissues from patients with colon cancer and was significantly associated with patient prognosis and diagnosis. Lapatinib inhibited the growth and proliferation of the colon cancer cell line HT-29. Additionally, lapatinib suppressed the expression of GPX4 in HT-29 cells, while the ferroptosis inhibitor ferrostatin-1 (Fer-1) partially restored its expression. Lapatinib induced an increase in intracellular ROS levels and malondialdehyde content in HT-29 cells, with Fer-1 partially restoring these levels. CONCLUSION Our findings demonstrated that lapatinib could effectively suppress the mRNA and protein expression of GPX4 in colon cancer cells, which elevates intracellular levels of ROS and malondialdehyde, ultimately inducing ferroptosis in these cells. This mechanism underscores the potential of lapatinib as a therapeutic strategy for targeting tumors.
Collapse
Affiliation(s)
- Yue Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China
| | - Dan Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China
| | - Chen Yuan
- Basic Medical College, Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Xiujuan Lang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang, 150081, China
| | - Songbo Fu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Sun H, Kang EY, Chen H, Sweeney KJ, Suchko M, Wu Y, Wen J, Krishnamurthy S, Albarracin CT, Ding QQ, Foo WC, Sahin AA. Immunohistochemical assessment of HER2 low breast cancer: interobserver reproducibility and correlation with digital image analysis. Breast Cancer Res Treat 2024; 205:403-411. [PMID: 38441847 DOI: 10.1007/s10549-024-07256-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/18/2024] [Indexed: 05/19/2024]
Abstract
PURPOSE The recent findings from the DESTINY-Breast04 trial highlighted the clinical importance of distinguishing between HER2 immunohistochemistry (IHC) scores 0 and 1 + in metastatic breast cancer (BC). However, pathologist interpretation of HER2 IHC scoring is subjective, and standardized methodology is needed. We evaluated the consistency of HER2 IHC scoring among pathologists and the accuracy of digital image analysis (DIA) in interpreting HER2 IHC staining in cases of HER2-low BC. METHODS Fifty whole-slide biopsies of BC with HER2 IHC staining were evaluated, comprising 25 cases originally reported as IHC score 0 and 25 as 1 +. These slides were digitally scanned. Six pathologists with breast expertise independently reviewed and scored the scanned images, and DIA was applied. Agreement among pathologists and concordance between pathologist scores and DIA results were statistically analyzed using Kendall coefficient of concordance (W) tests. RESULTS Substantial agreement among at least five of the six pathologists was found for 18 of the score 0 cases (72%) and 15 of the score 1 + cases (60%), indicating excellent interobserver agreement (W = 0.828). DIA scores were highly concordant with pathologist scores in 96% of cases (47/49), indicating excellent concordance (W = 0.959). CONCLUSION Although breast subspecialty pathologists were relatively consistent in evaluating BC with HER2 IHC scores of 0 and 1 +, DIA may be a reliable supplementary tool to enhance the standardization and quantification of HER2 IHC assessment, especially in challenging cases where results may be ambiguous (i.e., scores 0-1 +). These findings hold promise for improving the accuracy and consistency of HER2 testing.
Collapse
Affiliation(s)
- Hongxia Sun
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Eun Young Kang
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Hui Chen
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Keith J Sweeney
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Michael Suchko
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Yun Wu
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Jianguo Wen
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Savitri Krishnamurthy
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Constance T Albarracin
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Qing-Qing Ding
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Wai Chin Foo
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA
| | - Aysegul A Sahin
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, USA.
| |
Collapse
|
4
|
Bozorgchami N, Ahmadzadeh M, Hatamabadi D, Yazdani A, Shahhosseini S, Mohit E. Preparation, Characterization, and Radiolabeling of Anti-HER2 scFv With Technetium Tricarbonyl and Stability Studies. J Labelled Comp Radiopharm 2024; 67:168-179. [PMID: 38485465 DOI: 10.1002/jlcr.4090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 05/14/2024]
Abstract
Breast cancer is the most common diagnosed cancer, and the second cause of cancer death among women, worldwide. HER2 overexpression occurred in approximately 15% to 20% of breast cancers. Invasive biopsy method has been used for detection of HER2 overexpression. HER2-targeted imaging via an appropriate radionuclide is a promising method for sensitive and accurate identification of HER2+ primary and metastatic lesions. 99mTc-anti-HER2 scFv can specifically target malignancies and be used for diagnosis of the cancer type and metastasis as well as treatment of breast cancer. We radiolabeled anti-HER2 scFv that was expressed in Escherichia coli and purified through Ni-NTA resin under native condition with 99mTc-tricarbonyl formed from boranocarbonate. HER2-based ELISA, BCA, TLC, and HPLC were used in this study. In the current study, anti-HER2 scFv was lyophilized before radiolabeling. It was found that freeze-drying did not change the binding activity of anti-HER2 scFv to HER2. Results demonstrated direct anti-HER2 scFv radiolabeling by 99mTc-tricarbonyl to hexahistidine sequence (His-tag) without any changes in biological activity and radiochemical purity of around 98%. Stability analysis revealed that 99mTc-anti-HER2 scFv is stable for at least 24 h in PBS buffer, normal saline, human plasma proteins, and histidine solution.
Collapse
Affiliation(s)
- Negar Bozorgchami
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Ahmadzadeh
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Food and Drug Laboratory Research Center, Food and Drug Administration, The Ministry of Health and Medical Education, Tehran, Iran
| | - Dara Hatamabadi
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Yazdani
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soraya Shahhosseini
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Abada E, Singh K, Hansen K, Quddus MR. Low-level expression of human Epidermal growth Factor Receptor-2 (HER2) in High-Grade Mullerian Tumors: Implications for therapy decision making. Gynecol Oncol Rep 2023; 49:101251. [PMID: 37600081 PMCID: PMC10432814 DOI: 10.1016/j.gore.2023.101251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/17/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Reclassification of HER2-negative breast cancers to HER2 low-level expression allowed targeted anti-HER2 therapy in about 60% of patients, improving outcome. The high recurrence rates and often dismal outcomes with current therapies of high-grade Mullerian carcinomas, offers opportunity to explore anti-HER2 therapies in the gynecologic tract carcinomas. We investigated HER2 low expression as currently defined in breast carcinomas. Methods We reviewed all high-grade Mullerian cancers between 2016 and 2021, where HER2 by IHC and/or FISH tests were available. Additional clinical information was recorded, and statistical analysis was performed using SPSS (version 27). Results Forty (49.4%) tumors were endometrial, 20 (24.7%) ovarian, 16 (19.8%) fallopian tubal, and 5 (6.2%) primarily peritoneal. Overall, 17 (21.0%) were HER2 positive (IHC 3+/IHC 2+/FISH amplified), 31 (38.3%) HER2 low (IHC 1+/2+/FISH non-amplified), and 30 (37.0%) were HER2 negative (IHC = 0). HER2 low expression was noted in 15% ovarian, 25% fallopian tubal, 53% endometrial, and 60% peritoneal tumors; 34% and 21% of serous carcinomas, 63% and 13% of carcinosarcomas, and 67% and 33% of endometrioid carcinomas were HER2 low and HER2 positive respectively. HER2 negative and HER2 low expression had a significant association with primary tumor location (p = 0.001); endometrium and peritoneal tumors were more likely to be HER2 low and HER2 negative. During a mean follow-up of 13.2 months (range: 1-34), 5% of the patients were deceased. Conclusions Based on the current HER2-low recommendations in the breast, about one-third of patients with high-grade Mullerian carcinomas might qualify for anti-HER2 therapy with a potential for improved progression-free and overall survival.
Collapse
Affiliation(s)
- Evi Abada
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital and The Warren Alpert Medical School of Brown University, USA
| | - Kamaljeet Singh
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital and The Warren Alpert Medical School of Brown University, USA
| | - Katrine Hansen
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital and The Warren Alpert Medical School of Brown University, USA
| | - M. Ruhul Quddus
- Department of Pathology and Laboratory Medicine, Women & Infants Hospital and The Warren Alpert Medical School of Brown University, USA
| |
Collapse
|
6
|
Liu Y, Han D, Parwani AV, Li Z. Applications of Artificial Intelligence in Breast Pathology. Arch Pathol Lab Med 2023; 147:1003-1013. [PMID: 36800539 DOI: 10.5858/arpa.2022-0457-ra] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 02/19/2023]
Abstract
CONTEXT.— Increasing implementation of whole slide imaging together with digital workflow and advances in computing capacity enable the use of artificial intelligence (AI) in pathology, including breast pathology. Breast pathologists often face a significant workload, with diagnosis complexity, tedious repetitive tasks, and semiquantitative evaluation of biomarkers. Recent advances in developing AI algorithms have provided promising approaches to meet the demand in breast pathology. OBJECTIVE.— To provide an updated review of AI in breast pathology. We examined the success and challenges of current and potential AI applications in diagnosing and grading breast carcinomas and other pathologic changes, detecting lymph node metastasis, quantifying breast cancer biomarkers, predicting prognosis and therapy response, and predicting potential molecular changes. DATA SOURCES.— We obtained data and information by searching and reviewing literature on AI in breast pathology from PubMed and based our own experience. CONCLUSIONS.— With the increasing application in breast pathology, AI not only assists in pathology diagnosis to improve accuracy and reduce pathologists' workload, but also provides new information in predicting prognosis and therapy response.
Collapse
Affiliation(s)
- Yueping Liu
- From the Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China (Liu, Han)
| | - Dandan Han
- From the Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China (Liu, Han)
| | - Anil V Parwani
- The Department of Pathology, The Ohio State University, Columbus (Parwani, Li)
| | - Zaibo Li
- From the Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China (Liu, Han)
| |
Collapse
|
7
|
Qiu J, Liu Q, Li P, Jiang Q, Chen W, Li D, Li G, Shan G. Ligand-Directed Photodegradation of Interacting Proteins: Oxidative HER2/HER3 Heterodimer Degradation with a Lapatinib-Derived Photosensitizer. J Med Chem 2023; 66:10265-10272. [PMID: 37421416 DOI: 10.1021/acs.jmedchem.3c00252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
In this work, we described a photocatalytic approach, termed ligand-directed photodegradation of interacting proteins (LDPIP), for efficient protein-protein heterodimer degradation. This LDPIP approach utilizes a combination of a photosensitizing protein ligand and appropriate light and molecular oxygen to induce oxidative damage to the ligand-binding protein as well as its interacting protein partner. As a showcase study, a photosensitizing HER2 ligand HER-PS-I was rationally designed based on the FDA-approved HER2 inhibitor lapatinib to efficiently degrade HER2 together with its interacting protein partner HER3, which is thought to induce HER2-targeted therapy resistance and difficult to target by small molecules. HER-PS-I exhibited excellent anticancer activity against drug-resistant MDA-MB-453 cells and its three-dimensional multicellular spheroids. We hope that this LDPIP approach would find more applications in degrading proteins that are thought undruggable or difficult to drug.
Collapse
Affiliation(s)
- Jingru Qiu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, P. R. China
| | - Qinghong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, P. R. China
| | - Peixia Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, P. R. China
| | - Qiaoyun Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, P. R. China
| | - Weijia Chen
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, P. R. China
| | - Donghai Li
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Guiling Li
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P. R. China
| | - Gang Shan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, P. R. China
| |
Collapse
|
8
|
Li F, Liang Y, Luo M, Shen J, Zhou T, Liang Y, Tang X, Yuan H, Zeng J. The efficacy and safety of epirubicin and cyclophosphamide combined with pyrotinib in neoadjuvant treatment for HER2-positive breast cancer: A real-world study. Front Oncol 2023; 13:1041111. [PMID: 36793595 PMCID: PMC9922885 DOI: 10.3389/fonc.2023.1041111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Purpose Long-term survival benefit of anthracyclines for human epidermal growth factor receptor 2 (HER2)-positive breast cancer is clear. In the neoadjuvant treatment, compared with the monoclonal antibody such as trastuzumab and pertuzumab, the clinical benefit of pyrotinib, a new small-molecule tyrosine kinase inhibitor (TKI), as the main anti-HER2 strategy currently requires more research to determine. Our real-world study is the first prospective observational study in China to evaluate the efficacy and safety of epirubicin (E) and cyclophosphamide (C) with pyrotinib as anti-HER2 therapy in the neoadjuvant setting of patients with stage II-III HER2-positive breast cancer. Methods From May 2019 to December 2021, 44 untreated patients with HER2-positive nonspecific invasive breast cancer who received 4 cycles of neoadjuvant EC with pyrotinib. The primary endpoint was pathological complete response (pCR) rate. Secondary endpoints included the overall clinical response, breast pathological complete response rate (bpCR), the rate of axillary lymph nodes pathological negativity and adverse events (AEs). Other objective indicators were the rate of surgical breast-conserving, the negative conversion ratios of tumor markers. Results Thirty-seven (84.1%) of 44 patients completed this neoadjuvant therapy, and 35 (79.5%) had surgery and were included in the primary endpoint assessment. The objective response rate (ORR) of 37 patients was 97.3%. Two patients reached clinical complete response, 34 obtained clinical partial response, 1 sustained stable disease, and no one had progressive disease. Eleven (31.4%) of 35 patients who had surgery achieved bpCR and the rate of axillary lymph nodes pathological negativity was 61.3%. The tpCR rate was 28.6% (95% CI: 12.8-44.3%). Safety was evaluated in all 44 patients. Thirty-nine (88.6%) had diarrhea, and 2 developed grade 3 diarrhea. Four (9.1%) patients had grade 4 leukopenia. All grade 3-4 AEs could be improved after symptomatic treatment. Conclusion The regimen of 4 cycles of EC combined with pyrotinib presented some feasibility in the neoadjuvant setting for HER2-positive breast cancer with manageable safety. New regimens with pyrotinib should be evaluated for higher pCR in future. Trial registration chictr.org Identifier: ChiCTR1900026061.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jian Zeng
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, GuangXi, China
| |
Collapse
|
9
|
Wang X, Wang S, Yao S, Shi W, Ma K. The clinical characteristics and treatment of ovarian malignant mesoderm mixed tumor: a systematic review. J Ovarian Res 2022; 15:104. [PMID: 36114551 PMCID: PMC9482291 DOI: 10.1186/s13048-022-01037-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Background Ovarian malignant mesoderm mixed tumor (OMMMT) is a rare clinical entity. To provide reference for the treatment and prognosis of OMMMT, we analyzed the clinical features, pathology and molecular biology characteristic of published cases. Methods The English and Chinese reported cases of OMMMT were selected from PubMed, Clinical Trials.gov and CNKI database from 2000 to December 15th, 2021 following the PRISMA guidelines. Results A total of 63 literatures including 199 OMMMT cases were included. The average age of patients at diagnosis was 56.46 years, the highest incidence age was 60-65 years, and 82% of them were menopausal women. Most patients were diagnosed in FIGO III stage (59.64%). The most common symptom of OMMMT was abdominal pain (60.5%). 61.6% of patients were accompanied by ascites, while ascites was not associated with metastatic tumor and local recurrence. The CA125 of 88.68% patients increased. The most common reported carcinomatous component and sarcomatous component were serous adenocarcinoma (44.96%) and chondrosarcoma (24.81%), respectively. Initial treatment included surgery (94.97%) and taxanes-based (55.10%) or platinum-based (85.71%) chemotherapy regimens. The median survival time of patients was 20 months. Heterologous sarcoma component did not shorten life expectancy. The optimal ovarian tumor cell debulking surgery (OOTCDS), radiotherapy and chemotherapy could significantly prolong the median survival time of patients. Furthermore, platinum drugs could significantly prolong the survival time after comparing various chemotherapy schemes. Besides, the combination of platinum and taxanes was therapeutically superior to the combination of platinum and biological alkylating agents. Conclusion The OOTCDS and platinum-based chemotherapy regimen can improve the prognosis of OMMMT. Targeted therapy might become a new research direction in the future. Since the elderly patients are the majority, the toxicity of new drugs on the elderly patients is more noteworthy. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-022-01037-6.
Collapse
|
10
|
Gharibkandi NA, Molavipordanjani S, Mardanshahi A, Hosseinimehr SJ. The influence of kidneys ion transport inhibitors on the pharmacokinetic and tumor uptake behaviors of a HER2-targeted small size radiolabeled peptide. Curr Drug Metab 2022; 23:82-87. [PMID: 35049428 DOI: 10.2174/1389200223666220113101004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/06/2021] [Accepted: 11/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND HER2 over-expression plays a crucial role in the cancer treatment protocol. This study evaluates the effectiveness of kidney organic anion and cation transport inhibitors on the tumor uptake of 99mTc-HYNIC-(Ser)3-LTVPWY radiotracer in SKOV-3 tumor-bearing nude mice. METHODS Before the injection of the radiolabeled peptide, SKOV-3 tumor-bearing nude mice were treated with furosemide, cimetidine, para-amino hippuric acid, and saline. The inhibition effects of the organic anion and cation transport inhibitors were compared with the control group. In both treatment and control groups, the tumor and renal accumulation of radiopeptide in mice bearing SKOV-3 tumors were assessed in biodistribution and SPECT imaging studies. RESULTS The biodistribution and imaging results suggested that all treated groups showed a higher tumor and higher normal tissue radioactivity compared to the control group. According to the tumor imaging study, the furosemide-treated group had slightly better tumor uptake and a higher tumor to muscle uptake ratio than the other treatment groups. CONCLUSIONS Administration of furosemide (an OAT inhibitor) increased radioactivity accumulation in the kidneys and blood and improved tumor radioactivity uptake. PAH (an anion transporter substrate) and cimetidine (an OCT inhibitor) have a minor effect on the accumulation of radioactivity in the kidneys and the acquired images.
Collapse
Affiliation(s)
- Nasrin Abbasi Gharibkandi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Molavipordanjani
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Mardanshahi
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
11
|
Lara H, Li Z, Abels E, Aeffner F, Bui MM, ElGabry EA, Kozlowski C, Montalto MC, Parwani AV, Zarella MD, Bowman D, Rimm D, Pantanowitz L. Quantitative Image Analysis for Tissue Biomarker Use: A White Paper From the Digital Pathology Association. Appl Immunohistochem Mol Morphol 2021; 29:479-493. [PMID: 33734106 PMCID: PMC8354563 DOI: 10.1097/pai.0000000000000930] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/12/2021] [Indexed: 01/19/2023]
Abstract
Tissue biomarkers have been of increasing utility for scientific research, diagnosing disease, and treatment response prediction. There has been a steady shift away from qualitative assessment toward providing more quantitative scores for these biomarkers. The application of quantitative image analysis has thus become an indispensable tool for in-depth tissue biomarker interrogation in these contexts. This white paper reviews current technologies being employed for quantitative image analysis, their application and pitfalls, regulatory framework demands, and guidelines established for promoting their safe adoption in clinical practice.
Collapse
Affiliation(s)
- Haydee Lara
- GlaxoSmithKline-R&D, Cellular Biomarkers, Collegeville, PA
| | - Zaibo Li
- The Ohio State University, Columbus, OH
| | | | - Famke Aeffner
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc
| | | | | | | | | | | | | | | | - David Rimm
- Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
12
|
Liu CH, Grodzinski P. Nanotechnology for Cancer Imaging: Advances, Challenges, and Clinical Opportunities. Radiol Imaging Cancer 2021; 3:e200052. [PMID: 34047667 PMCID: PMC8183257 DOI: 10.1148/rycan.2021200052] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/28/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
Nanoparticle (NP) imaging applications have the potential to improve cancer diagnostics, therapeutics, and treatment management. In biomedical research and clinical practice, NPs can serve as labels or labeled carriers for monitoring drug delivery or serve as imaging agents for enhanced imaging contrast, as well as providing improved signal sensitivity and specificity for in vivo imaging of molecular and cellular processes. These qualities offer exciting opportunities for NP-based imaging agents to address current limitations in oncologic imaging. Despite substantial advancements in NP design and development, very few NP-based imaging agents have translated into clinics within the past 5 years. This review highlights some promising NP-enabled imaging techniques and their potential to address current clinical cancer imaging limitations. Although most examples provided herein are from the preclinical space, discussed imaging solutions could offer unique in vivo tools to solve biologic questions, improve cancer treatment effectiveness, and inspire clinical translation innovation to improve patient care. Keywords: Molecular Imaging-Cancer, Molecular Imaging-Nanoparticles, Molecular Imaging-Optical Imaging, Metastases, Oncology, Surgery, Treatment Effects.
Collapse
Affiliation(s)
- Christina H. Liu
- From the Cancer Imaging Program, National Cancer Institute, National
Institutes of Health, 9609 Medical Center Dr, Room 4W216, Rockville, MD
20850
| | - Piotr Grodzinski
- From the Cancer Imaging Program, National Cancer Institute, National
Institutes of Health, 9609 Medical Center Dr, Room 4W216, Rockville, MD
20850
| |
Collapse
|
13
|
Lin X, O'Reilly Beringhs A, Lu X. Applications of Nanoparticle-Antibody Conjugates in Immunoassays and Tumor Imaging. AAPS J 2021; 23:43. [PMID: 33718979 PMCID: PMC7956929 DOI: 10.1208/s12248-021-00561-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Modern diagnostic technologies rely on both in vitro and in vivo modalities to provide a complete understanding of the clinical state of a patient. Nanoparticle-antibody conjugates have emerged as promising systems to confer increased sensitivity and accuracy for in vitro diagnostics (e.g., immunoassays). Meanwhile, in vivo applications have benefited from the targeting ability of nanoparticle-antibody conjugates, as well as payload flexibility and tailored biodistribution. This review provides an encompassing overview of nanoparticle-antibody conjugates, from chemistry to applications in medical immunoassays and tumor imaging, highlighting the underlying principles and unique features of relevant preclinical applications employing commonly used imaging modalities (e.g., optical/photoacoustics, positron-emission tomography, magnetic resonance imaging, X-ray computed tomography).
Collapse
Affiliation(s)
- Xinhao Lin
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| | | | - Xiuling Lu
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA.
| |
Collapse
|
14
|
Abstract
Breast cancer is the most common malignancy in women. Basic and translational breast cancer research relies heavily on experimental animal models. Ideally, such models for breast cancer should have commonality with human breast cancer in terms of tumor etiology, biological behavior, pathology, and response to therapeutics. This review introduces current progress in different breast cancer experimental animal models and analyzes their characteristics, advantages, disadvantages, and potential applications. Finally, we propose future research directions for breast cancer animal models.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Wei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ce-Shi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
15
|
A quantitative systems pharmacological approach identified activation of JNK signaling pathway as a promising treatment strategy for refractory HER2 positive breast cancer. J Pharmacokinet Pharmacodyn 2021; 48:273-293. [PMID: 33389550 DOI: 10.1007/s10928-020-09732-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/08/2020] [Indexed: 02/01/2023]
Abstract
HER2-positive breast cancer (BC) is a rapidly growing and aggressive BC subtype that predominantly affects younger women. Despite improvements in patient outcomes with anti-HER2 therapy, primary and/or acquired resistance remain a major clinical challenge. Here, we sought to use a quantitative systems pharmacological (QSP) approach to evaluate the efficacy of lapatinib (LAP), abemaciclib (ABE) and 5-fluorouracil (5-FU) mono- and combination therapies in JIMT-1 cells, a HER2+ BC cell line exhibiting intrinsic resistance to trastuzumab. Concentration-response relationships and temporal profiles of cellular viability were assessed upon exposure to single agents and their combinations. To quantify the nature and intensity of drug-drug interactions, pharmacodynamic cellular response models were generated, to characterize single agent and combination time course data. Temporal changes in cell-cycle phase distributions, intracellular protein signaling, and JIMT-1 cellular viability were quantified, and a systems-based protein signaling network model was developed, integrating protein dynamics to drive the observed changes in cell viability. Global sensitivity analyses for each treatment arm were performed, to identify the most influential parameters governing cellular responses. Our QSP model was able to adequately characterize protein dynamic and cellular viability trends following single and combination drug exposure. Moreover, the model and subsequent sensitivity analyses suggest that the activation of the stress pathway, through pJNK, has the greatest impact over the observed declines of JIMT-1 cell viability in vitro. These findings suggest that dual HER2 and CDK 4/6 inhibition may be a promising novel treatment strategy for refractory HER2+ BC, however, proof-of-concept in vivo studies are needed to further evaluate the combined use of these therapies.
Collapse
|
16
|
HER2 intratumoral heterogeneity is independently associated with distal metastasis and overall survival in HER2-positive breast carcinomas. Breast Cancer Res Treat 2020; 181:519-527. [PMID: 32335801 DOI: 10.1007/s10549-020-05650-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/17/2020] [Indexed: 01/02/2023]
Abstract
PURPOSE Human epidermal growth factor receptor 2 (HER2) intratumoral heterogeneity (ITH) occurs in a subset of breast cancers. Our recent study revealed it as an independent predictive factor for the response to anti-HER2 neoadjuvant therapy. In this study, we aimed to investigate its association with distal metastasis. METHODS HER2 ITH was assessed using HER2 gene protein assay (GPA) on whole tissue sections of pretreatment biopsies from a cohort of 158 HER2-positive invasive breast carcinomas and correlated with patients' clinical follow-up outcomes along with other clinicopathologic characteristics. RESULTS Fifty-seven cases (36%) showed HER2 ITH including 19 with genetic, 8 with both genetic and non-genetic, and 30 with non-genetic ITH. Multivariate analysis demonstrated larger tumor size, positive resected lymph node(s), negative PR, and the presence of HER2 ITH were independently associated with distal metastasis. Additionally, multivariate analysis demonstrated larger tumor size and the presence of HER2 ITH were the only independent factors associated with decreased overall survival (death). CONCLUSION The presence of HER2 ITH is an independent factor associated with poor overall survival and increased distal metastasis in HER2-positive breast cancer patients.
Collapse
|
17
|
Hartage R, Li AC, Hammond S, Parwani AV. A Validation Study of Human Epidermal Growth Factor Receptor 2 Immunohistochemistry Digital Imaging Analysis and its Correlation with Human Epidermal Growth Factor Receptor 2 Fluorescence In situ Hybridization Results in Breast Carcinoma. J Pathol Inform 2020; 11:2. [PMID: 32154039 PMCID: PMC7032021 DOI: 10.4103/jpi.jpi_52_19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/16/2019] [Indexed: 01/03/2023] Open
Abstract
Background: The Visiopharm human epidermal growth factor receptor 2 (HER2) digital imaging analysis (DIA) algorithm assesses digitized HER2 immunohistochemistry (IHC) by measuring cell membrane connectivity. We aimed to validate this algorithm for clinical use by comparing with pathologists’ scoring and correlating with HER2 fluorescence in situ hybridization (FISH) results. Materials and Methods: The study cohort consisted of 612 consecutive invasive breast carcinoma specimens including 395 biopsies and 217 resections. HER2 IHC slides were scanned using Philips IntelliSite Scanners, and the digital images were analyzed using Visiopharm HER2-CONNECT App to obtain the connectivity values (0–1) and scores (0, 1+, 2+, and 3+). HER2 DIA scores were compared with Pathologists’ manual scores, and HER2 connectivity values were correlated with HER2 FISH results. Results: The concordance between HER2 DIA scores and pathologists’ scores was 87.3% (534/612). All discordant cases (n = 78) were only one-step discordant (negative to equivocal, equivocal to positive, or vice versa). Five cases (0.8%) showed discordant HER2 IHC DIA and HER2 FISH results, but all these cases had relatively low HER2 copy numbers (between 4 and 6). HER2 IHC connectivity showed significantly better correlation with HER2 copy number than HER2/CEP17 ratio. Conclusions: HER2 IHC DIA demonstrates excellent concordance with pathologists’ scores and accurately discriminates between HER2 FISH positive and negative cases. HER2 IHC connectivity has better correlation with HER2 copy number than HER2/CEP17 ratio, suggesting HER2 copy number may be more important in predicting HER2 protein expression, and response to anti-HER2-targeted therapy.
Collapse
Affiliation(s)
- Ramon Hartage
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Aidan C Li
- Department of NA, Jerome High School, Dublin, OH 43017, USA
| | - Scott Hammond
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Li AC, Zhao J, Zhao C, Ma Z, Hartage R, Zhang Y, Li X, Parwani AV. Quantitative digital imaging analysis of HER2 immunohistochemistry predicts the response to anti-HER2 neoadjuvant chemotherapy in HER2-positive breast carcinoma. Breast Cancer Res Treat 2020; 180:321-329. [PMID: 32002765 DOI: 10.1007/s10549-020-05546-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/21/2020] [Indexed: 01/04/2023]
Abstract
PURPOSE Patients with HER2-positive breast cancer commonly receive anti-HER2 neoadjuvant chemotherapy and pathologic complete response (pCR) can be achieved in up to half of the patients. HER2 protein expression detected by immunohistochemistry (IHC) can be quantified using digital imaging analysis (DIA) as a value of membranous connectivity. We aimed to investigate the association HER2 IHC DIA quantitative results with response to anti-HER2 neoadjuvant chemotherapy. METHODS Digitized HER2 IHC whole slide images were analyzed using Visiopharm HER2-CONNECT to obtain quantitative HER2 membranous connectivity from a cohort of 153 HER2+ invasive breast carcinoma cases treated with anti-HER2 neoadjuvant chemotherapy (NAC). HER2 connectivity and other factors including age, histologic grade, ER, PR, and HER2 fluorescence in situ hybridization (FISH) were analyzed for association with the response to anti-HER2 NAC. RESULTS Eighty-three cases (54.2%) had pCR, while 70 (45.8%) showed residual tumor. Younger age, negative ER/PR, higher HER2 DIA connectivity, higher HER2 FISH ratio and copy number were significantly associated with pCR in univariate analysis. Multivariate analysis demonstrated only age, HER2 DIA connectivity, PR negativity, and HER2 copy number was significantly associated with pCR, whereas HER2 DIA connectivity had the strongest association. CONCLUSIONS HER2 IHC DIA connectivity is the most important factor predicting pCR to anti-HER2 neoadjuvant chemotherapy in patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Aidan C Li
- Dublin Jerome High School, Dublin, OH, USA
| | - Jing Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Zhao
- Center for Biostatistics, Emory University, Atlanta, GA, USA
| | - Zhongliang Ma
- Department of Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ramon Hartage
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Yunxiang Zhang
- Department of Pathology, Weifang People's Hospital, Weifang, China
| | - Xiaoxian Li
- Department of Pathology, Emory University, Atlanta, GA, USA.
| | - Anil V Parwani
- Department of Pathology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
19
|
El-Abd E, Matta CA, Sheta M, El-Kerm Y, Sakr S. Histopathological characteristics of breast cancer and evaluation of ER alpha and Her-2neu using immunohistochemical and RT-PCR techniques. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2014.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Eman El-Abd
- Molecular Biology Department, Medical Technology Centre (MTC), Medical Research Institute (MRI), Alexandria University, Egypt
- Radiation Sciences Department, Medical Research Institute (MRI), Alexandria University, Egypt
| | - Cecil A. Matta
- Zoology Department, Faculty of Science, Alexandria University, Egypt
| | - Manal Sheta
- Pathology Department, Medical Research Institute (MRI), Alexandria University, Egypt
| | - Yasser El-Kerm
- Cancer Research and Management Department, Medical Research Institute (MRI), Alexandria University, Egypt
| | - Shimaa Sakr
- Medical Technology Centre (MTC), Medical Research Institute (MRI), Alexandria University, Egypt
| |
Collapse
|
20
|
Abstract
Breast cancer remains the second leading cause of cancer death among woman, worldwide, despite advances in identifying novel targeted therapies and the development of treating strategies. Classification of clinical subtypes (ER+, PR+, HER2+, and TNBC (Triple-negative)) increases the complexity of breast cancers, which thus necessitates further investigation. Mouse models used in breast cancer research provide an essential approach to examine the mechanisms and genetic pathway in cancer progression and metastasis and to develop and evaluate clinical therapeutics. In this review, we summarize tumor transplantation models and genetically engineered mouse models (GEMMs) of breast cancer and their applications in the field of human breast cancer research and anti-cancer drug development. These models may help to improve the knowledge of underlying mechanisms and genetic pathways, as well as creating approaches for modeling clinical tumor subtypes, and developing innovative cancer therapy.
Collapse
|
21
|
Ultrasmall targeted nanoparticles with engineered antibody fragments for imaging detection of HER2-overexpressing breast cancer. Nat Commun 2018; 9:4141. [PMID: 30297810 PMCID: PMC6175906 DOI: 10.1038/s41467-018-06271-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022] Open
Abstract
Controlling the biodistribution of nanoparticles upon intravenous injection is the key to achieving target specificity. One of the impediments in nanoparticle-based tumor targeting is the inability to limit the trafficking of nanoparticles to liver and other organs leading to smaller accumulated amounts in tumor tissues, particularly via passive targeting. Here we overcome both these challenges by designing nanoparticles that combine the specificity of antibodies with favorable particle biodistribution profiles, while not exceeding the threshold for renal filtration as a combined vehicle. To that end, ultrasmall silica nanoparticles are functionalized with anti-human epidermal growth factor receptor 2 (HER2) single-chain variable fragments to exhibit high tumor-targeting efficiency and efficient renal clearance. This ultrasmall targeted nanotheranostics/nanotherapeutic platform has broad utility, both for imaging a variety of tumor tissues by suitably adopting the targeting fragment and as a potentially useful drug delivery vehicle.
Collapse
|
22
|
Román-Rosales AA, García-Villa E, Herrera LA, Gariglio P, Díaz-Chávez J. Mutant p53 gain of function induces HER2 over-expression in cancer cells. BMC Cancer 2018; 18:709. [PMID: 29970031 PMCID: PMC6029411 DOI: 10.1186/s12885-018-4613-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/20/2018] [Indexed: 11/10/2022] Open
Abstract
Background HER2 over-expression is related with a poor prognosis in patients with invasive breast cancer tumors. Clinical associations have reported that somatic mutations of p53 more frequently detected in cases of sporadic breast cancer of the HER2 subtypes, besides a high percentage of HER2-amplifying tumors carry germline mutations of p53. The mechanisms responsible for the acquisition of oncogenic functions of p53 mutant proteins (mtp53), known as Gain of Function (GOF), over HER2 expression have not been reported. The objective of this study was to evaluate a possible relationship between p53 mutants and HER2 regulation. Methods HER2 expression (transcription and protein), as well as HER2 protein stabilization have been evaluated after inducing or silencing of p53 mutants’ expression in cell lines. Finally, we evaluated the interaction of the p53 mutants over the HER2 receptor promoter. Results Higher HER2 expression in cell lines harboring endogenous mtp53 compared with wt or null expression of p53 cell lines. Transfection of p53 mutants (R248Q and R273C) in cell lines increased the expression of HER2. Silencing of p53 mutants, decrease HER2 expression. The p53 mutants R248Q and R273C significantly increase the luciferase activity on the HER2 promoter, and both mutants also promote acetylation of H3 and H4 histones binding in it. Conclusions These findings show for the first time that p53 mutants induce over-expression of HER2 at transcriptional level of the HER2 protein. Our results could have clinical implications in breast cancer and other types of cancer where HER2 is over-expressed and used as a therapy target. Electronic supplementary material The online version of this article (10.1186/s12885-018-4613-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- A A Román-Rosales
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Av. San Fernando No. 22, Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.,Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), 07360, Ciudad de México, Mexico
| | - E García-Villa
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), 07360, Ciudad de México, Mexico
| | - L A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Av. San Fernando No. 22, Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico
| | - P Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), 07360, Ciudad de México, Mexico.
| | - J Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Av. San Fernando No. 22, Sección XVI, Tlalpan, 14080, Ciudad de México, Mexico.
| |
Collapse
|
23
|
Richard S, Selle F, Lotz JP, Khalil A, Gligorov J, Soares DG. Pertuzumab and trastuzumab: the rationale way to synergy. AN ACAD BRAS CIENC 2018; 88 Suppl 1:565-77. [PMID: 27275646 DOI: 10.1590/0001-3765201620150178] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/05/2015] [Indexed: 02/03/2023] Open
Abstract
It has now been 15 years since the HER2-targeted monoclonal antibody trastuzumab was introduced in clinical and revolutionized the treatment of HER2-positive breast cancer patients. Despite this achievement, most patients with HER2-positive metastatic breast cancer still show progression of their disease, highlighting the need for new therapies. The continuous interest in novel targeted agents led to the development of pertuzumab, the first in a new class of agents, the HER dimerization inhibitors. Pertuzumab is a novel recombinant humanized antibody directed against extracellular domain II of HER2 protein that is required for the heterodimerization of HER2 with other HER receptors, leading to the activation of downstream signalling pathways. Pertuzumab combined with trastuzumab plus docetaxel was approved for the first-line treatment of patients with HER2-positive metastatic breast cancer and is currently used as a standard of care in this indication. In the neoadjuvant setting, the drug was granted FDA-accelerated approval in 2013. Pertuzumab is also being evaluated in the adjuvant setting. The potential of pertuzumab relies in the dual complete blockade of the HER2/3 axis when administered with trastuzumab. This paper synthetises preclinical and clinical data on pertuzumab and highlights the mechanisms underlying the synergistic activity of the combination pertuzumab-trastuzumab which are essentially due to their complementary mode of action.
Collapse
Affiliation(s)
- Sandrine Richard
- Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l'Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France, Medical Oncology Department, Tenon Hospital, Paris , France
| | - Frédéric Selle
- Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l'Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France, Medical Oncology Department, Tenon Hospital, Paris , France
| | - Jean-Pierre Lotz
- Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l'Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France, Medical Oncology Department, Tenon Hospital, Paris , France.,Institut Universitaire de Cancérologie Université Pierre et Marie Curie (IUC-UPMC Univ Paris 06), Sorbonne Universités, 4 place Jussieu, 75005 Paris, France, Université Curie Paris 6, Institut Universitaire de Cancérologie, Université Pierre et Marie Curie, Paris , France
| | - Ahmed Khalil
- Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l'Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France, Medical Oncology Department, Tenon Hospital, Paris , France
| | - Joseph Gligorov
- Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l'Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France, Medical Oncology Department, Tenon Hospital, Paris , France.,Institut Universitaire de Cancérologie Université Pierre et Marie Curie (IUC-UPMC Univ Paris 06), Sorbonne Universités, 4 place Jussieu, 75005 Paris, France, Université Curie Paris 6, Institut Universitaire de Cancérologie, Université Pierre et Marie Curie, Paris , France
| | - Daniele G Soares
- Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l'Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France, Medical Oncology Department, Tenon Hospital, Paris , France
| |
Collapse
|
24
|
Park CM, Kawasaki Y, Refaat A, Sakurai H. Mechanisms for DNA-damaging agent-induced inactivation of ErbB2 and ErbB3 via the ERK and p38 signaling pathways. Oncol Lett 2018; 15:1758-1762. [PMID: 29434871 DOI: 10.3892/ol.2017.7532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 02/23/2017] [Indexed: 11/06/2022] Open
Abstract
Cisplatin (CDDP) and doxorubicin (DOX) are chemotherapeutic drugs that trigger apoptosis by inducing DNA-damage. A previous study using breast cancer cells demonstrated the negative feedback modulation of the epidermal growth factor receptor (EGFR) and receptor tyrosine-protein kinase erbB-2 (ErbB2) via extracellular signal-regulated kinase (ERK)-mediated phosphorylation of conserved Thr-669 and Thr-677 residues, respectively, in the juxtamembrane domain. In addition, CDDP has been identified to cause negative feedback inhibition of activated EGFR in lung cancer cells. In the present study, the role of phosphorylation in the feedback control of the ErbB2/ErbB3 heterodimer in human breast and gastric cancer cells was investigated. Phosphorylation of ErbB2 at Thr-677 was induced by CDDP and DOX, which in turn reduced tyrosine autophosphorylation of ErbB2 and ErbB3. Treatment with trametinib, a mitogen-activated protein kinase inhibitor that blocks ERK-mediated Thr-677 phosphorylation, and substitution of Thr-677 to alanine, blocked the feedback inhibition of ErbB2 and ErbB3. In addition, these agents caused the degradation of ErbB proteins through the activation of p38 mitogen-activated protein kinase (p38) and ERK. These results demonstrate that chemotherapeutic agents trigger ERK- and p38-mediated post-translational downregulation of ErbB receptors.
Collapse
Affiliation(s)
- Chul Min Park
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Yuki Kawasaki
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Alaa Refaat
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
25
|
Singla H, Ludhiadch A, Kaur RP, Chander H, Kumar V, Munshi A. Recent advances in HER2 positive breast cancer epigenetics: Susceptibility and therapeutic strategies. Eur J Med Chem 2017; 142:316-327. [DOI: 10.1016/j.ejmech.2017.07.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/27/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
26
|
Hou Y, Nitta H, Wei L, Banks PM, Parwani AV, Li Z. Evaluation of Immune Reaction and PD-L1 Expression Using Multiplex Immunohistochemistry in HER2-Positive Breast Cancer: The Association With Response to Anti-HER2 Neoadjuvant Therapy. Clin Breast Cancer 2017; 18:e237-e244. [PMID: 29198959 DOI: 10.1016/j.clbc.2017.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/01/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Immune reaction with tumor-infiltrating lymphocytes (TILs) has been extensively investigated in breast cancer. Programmed cell death 1 and its ligand (PD-L1) are key physiologic suppressors of cytotoxic immune reaction. However, the combination of TILs with PD-L1 expression has not been well studied in breast cancer. PATIENTS AND METHODS A multi-color immunohistochemical multiplex assay simultaneously detecting PD-L1, CD8, and CD163 was performed on biopsy whole sections from 123 HER2-positive (HER2+) breast cancers, including 64 treated with anti-HER2 neoadjuvant therapy and subsequent resection. RESULTS PD-L1 expression was identified in 88 cases (72%) including 21 (17%) in tumor cells and 67 (55%) in immune cells. PD-L1 expression was positively associated with high Nottingham grade, high nuclear grade, and a high level of CD8+ and CD163+ cells. Among the 64 patients who received neoadjuvant therapy, 39 had pathologic complete remission (pCR) and 25 had incomplete response. Multivariate analysis showed progesterone receptor negativity, HER2/chromosome 17 centromere (CEN17) ratio and intratumoral CD8+ cells were significantly associated with pCR. Furthermore, all patients with intratumoral CD8+ cells but no PD-L1 expression achieved pCR. CONCLUSION Our data have shown that examination of intratumoral CD8+ cells together with PD-L1 expression proves useful in predicting response to anti-HER2 targeted therapy in patients with HER2+ breast cancer.
Collapse
Affiliation(s)
- Yanjun Hou
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH
| | | | - Lai Wei
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH
| | | | - Anil V Parwani
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH
| | - Zaibo Li
- Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus, OH.
| |
Collapse
|
27
|
HER2 intratumoral heterogeneity is independently associated with incomplete response to anti-HER2 neoadjuvant chemotherapy in HER2-positive breast carcinoma. Breast Cancer Res Treat 2017; 166:447-457. [DOI: 10.1007/s10549-017-4453-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/07/2017] [Indexed: 12/01/2022]
|
28
|
Vitale SG, Laganà AS, Capriglione S, Angioli R, La Rosa VL, Lopez S, Valenti G, Sapia F, Sarpietro G, Butticè S, Tuscano C, Fanale D, Tropea A, Rossetti D. Target Therapies for Uterine Carcinosarcomas: Current Evidence and Future Perspectives. Int J Mol Sci 2017; 18:1100. [PMID: 28531111 PMCID: PMC5455008 DOI: 10.3390/ijms18051100] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/13/2017] [Accepted: 05/16/2017] [Indexed: 01/04/2023] Open
Abstract
Carcinosarcomas (CS) in gynecology are very infrequent and represent only 2-5% of uterine cancers. Despite surgical cytoreduction and subsequent chemotherapy being the primary treatment for uterine CS, the overall five-year survival rate is 30 ± 9% and recurrence is extremely common (50-80%). Due to the poor prognosis of CS, new strategies have been developed in the last few decades, targeting known dysfunctional molecular pathways for immunotherapy. In this paper, we aimed to gather the available evidence on the latest therapies for the treatment of CS. We performed a systematic review using the terms "uterine carcinosarcoma", "uterine Malignant Mixed Müllerian Tumors", "target therapies", "angiogenesis therapy", "cancer stem cell therapy", "prognostic biomarker", and "novel antibody-drug". Based on our results, the differential expression and accessibility of epithelial cell adhesion molecule-1 on metastatic/chemotherapy-resistant CS cells in comparison to normal tissues and Human Epidermal Growth Factor Receptor 2 (HER2) open up new possibilities in the field of target therapy. Nevertheless, future investigations are needed to clarify the impact of these new therapies on survival rate and medium-/long-term outcomes.
Collapse
Affiliation(s)
- Salvatore Giovanni Vitale
- Unit of Gynecology and Obstetrics, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Antonio Simone Laganà
- Unit of Gynecology and Obstetrics, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Stella Capriglione
- Department of Obstetrics and Gynecology, Campus Bio Medico University of Rome, 00128 Rome, Italy.
| | - Roberto Angioli
- Department of Obstetrics and Gynecology, Campus Bio Medico University of Rome, 00128 Rome, Italy.
| | - Valentina Lucia La Rosa
- Unit of Psychodiagnostics and Clinical Psychology, University of Catania, 95124 Catania, Italy.
| | - Salvatore Lopez
- Department of Obstetrics and Gynecology, Campus Bio Medico University of Rome, 00128 Rome, Italy.
| | - Gaetano Valenti
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Fabrizio Sapia
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Giuseppe Sarpietro
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Salvatore Butticè
- Department of Human Pathology, Unit of Urology, University of Messina, 98124 Messina, Italy.
| | - Carmelo Tuscano
- Radiation Oncology Department, AO "Bianchi-Melacrino-Morelli", 89124 Reggio Calabria, Italy.
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy.
| | - Alessandro Tropea
- Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy.
| | - Diego Rossetti
- Unit of Gynecology and Obstetrics, Desenzano del Garda Hospital, Section of Gavardo, 25085 Gavardo, Brescia, Italy.
| |
Collapse
|
29
|
Couto JP, Bentires-Alj M. Mouse Models of Breast Cancer: Deceptions that Reveal the Truth. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Hou Y, Nitta H, Li Z. HER2 Gene Protein Assay Is Useful to Determine HER2 Status and Evaluate HER2 Heterogeneity in HER2 Equivocal Breast Cancer. Am J Clin Pathol 2017; 147:89-95. [PMID: 28110280 DOI: 10.1093/ajcp/aqw211] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVES Approximately 15% of breast cancers show equivocal human epidermal growth factor receptor 2 (HER2) results on HER2 immunohistochemistry (IHC) and are reflexed for fluorescence in situ hybridization (FISH). However, some cases remain equivocal. In this study, we evaluated these double-equivocal cases by using a novel gene protein assay (GPA), which can simultaneously assess HER2 gene copy number and protein on a single slide using bright-field microscopy. METHODS GPA was performed on 42 HER2 IHC and FISH double-equivocal cases. RESULTS GPA was negative for amplification in 28 cases, equivocal in three cases, and positive in 11 cases. The GPA results showed excellent concordance with either repeat FISH using a chromosome 17 centromere probe or FISH using an alternative probe. Furthermore, HER2 heterogeneity was identified in three of 11 GPA-positive cases. CONCLUSIONS HER2 GPA performs accurately and is very useful to determine HER2 status in HER2 IHC and FISH double-equivocal breast cancer cases and identify HER2 heterogeneity.
Collapse
Affiliation(s)
- Yanjun Hou
- From the Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus
| | | | - Zaibo Li
- From the Department of Pathology, Wexner Medical Center at The Ohio State University, Columbus
| |
Collapse
|
31
|
Kawiak A, Domachowska A. Plumbagin Suppresses the Invasion of HER2-Overexpressing Breast Cancer Cells through Inhibition of IKKα-Mediated NF-κB Activation. PLoS One 2016; 11:e0164064. [PMID: 27727280 PMCID: PMC5058502 DOI: 10.1371/journal.pone.0164064] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/19/2016] [Indexed: 12/17/2022] Open
Abstract
HER2-overexpressing breast cancers account for about 30% of breast cancer occurrences and have been correlated with increased tumor aggressiveness and invasiveness. The nuclear factor-κB (NF-κB) is overexpressed in a subset of HER2-positive breast cancers and its upregulation has been associated with the metastatic potential of HER2-overexpressing tumors. The present study aimed at determining the potential of plumbagin, a naturally occurring naphthoquinone, to inhibit the invasion of HER2-overexpressing breast cancer cells and determine the involvement of NF-κB inhibition in plumbagin-mediated cell invasion suppression. In the present research we showed that plumbagin inhibited the transcriptional activity of NF-κB in HER2-positive breast cancer cells. The suppression of NF-κB activation corresponded with the inhibition of NF-κB p65 phosphorylation and downregulation of NF-κB-regulated matrix metalloproteinase 9 (MMP-9) expression. Plumbagin suppressed the invasion of HER2-overexpressing breast cancer cells and the inhibition of cell invasion was associated with the ability of plumbagin to inhibit NF-κB transcriptional activity. The silencing of NF-κB p65 increased the sensitivity of HER2-overexpressing breast cancer cells to plumbagin-induced cell invasion inhibition. NF-κB inhibition was associated with IκB kinase α (IKKα) activity suppression and inhibition of IκBα phosphorylation and degradation. The knockdown of IKKα resulted in increased sensitivity of HER2-positive cells to plumbagin-induced suppression of NF-κB transcriptional activity and expression of MMP-9. In conclusion, plumbagin inhibits the invasion of HER2-overexpressing breast cancer cells through the inhibition of IKKα-mediated NF-κB activation and downregulation of NF-κB-regulated MMP-9 expression.
Collapse
Affiliation(s)
- Anna Kawiak
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk & Medical University of Gdansk, Gdansk, Poland
- Laboratory of Human Physiology, Faculty of Health Sciences with Subfaculty of Nursing, Medical University of Gdansk, Gdansk, Poland
| | - Anna Domachowska
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk & Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
32
|
Kawiak A, Lojkowska E. Ramentaceone, a Naphthoquinone Derived from Drosera sp., Induces Apoptosis by Suppressing PI3K/Akt Signaling in Breast Cancer Cells. PLoS One 2016; 11:e0147718. [PMID: 26840401 PMCID: PMC4739616 DOI: 10.1371/journal.pone.0147718] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/07/2016] [Indexed: 11/18/2022] Open
Abstract
The phosphoinositide 3-kinase (PI3K) signaling pathway plays an important role in processes critical for breast cancer progression and its upregulation confers increased resistance of cancer cells to chemotherapy and radiation. The present study aimed at determining the activity of ramentaceone, a constituent of species in the plant genera Drosera, toward breast cancer cells and defining the involvement of PI3K/Akt inhibition in ramentaceone-mediated cell death induction. The results showed that ramentaceone exhibited high antiproliferative activity toward breast cancer cells, in particular HER2-overexpressing breast cancer cells. The mode of cell death induced by ramentaceone was through apoptosis as determined by cytometric analysis of caspase activity and Annexin V staining. Apoptosis induction was found to be mediated by inhibition of PI3K/Akt signaling and through targeting its downstream anti-apoptotic effectors. Ramentaceone inhibited PI3-kinase activity, reduced the expression of the PI3K protein and inhibited the phosphorylation of the Akt protein in breast cancer cells. The expression of the anti-apoptotic Bcl-2 protein was decreased and the levels of the pro-apoptotic proteins, Bax and Bak, were elevated. Moreover, inhibition of PI3K and silencing of Akt expression increased the sensitivity of cells to ramentaceone-induced apoptosis. In conclusion, our results indicate that ramentaceone induces apoptosis in breast cancer cells through PI3K/Akt signaling inhibition. These findings suggest further investigation of ramentaceone as a potential therapeutic agent in breast cancer therapy, in particular HER2-positive breast cancer.
Collapse
Affiliation(s)
- Anna Kawiak
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Kladki 24, 80–822, Gdansk, Poland
- Laboratory of Human Physiology, Faculty of Health Sciences with Subfaculty of Nursing, Medical University of Gdansk, Tuwima 15, 80–210, Gdansk, Poland
- * E-mail:
| | - Ewa Lojkowska
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Kladki 24, 80–822, Gdansk, Poland
| |
Collapse
|
33
|
Casey MC, Sweeney KJ, Brown JAL, Kerin MJ. Exploring circulating micro-RNA in the neoadjuvant treatment of breast cancer. Int J Cancer 2016; 139:12-22. [PMID: 26756433 PMCID: PMC5066681 DOI: 10.1002/ijc.29985] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/04/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
Breast cancer is the most frequently diagnosed malignancy amongst females worldwide. In recent years the management of this disease has transformed considerably, including the administration of chemotherapy in the neoadjuvant setting. Aside from increasing rates of breast conserving surgery and enabling surgery via tumour burden reduction, use of chemotherapy in the neoadjuvant setting allows monitoring of in vivo tumour response to chemotherapeutics. Currently, there is no effective means of identifying chemotherapeutic responders from non‐responders. Whilst some patients achieve complete pathological response (pCR) to chemotherapy, a good prognostic index, a proportion of patients derive little or no benefit, being exposed to the deleterious effects of systemic treatment without any knowledge of whether they will receive benefit. The identification of predictive and prognostic biomarkers could confer multiple benefits in this setting, specifically the individualization of breast cancer management and more effective administration of chemotherapeutics. In addition, biomarkers could potentially expedite the identification of novel chemotherapeutic agents or increase their efficacy. Micro‐RNAs (miRNAs) are small non‐coding RNA molecules. With their tissue‐specific expression, correlation with clinicopathological prognostic indices and known dysregulation in breast cancer, miRNAs have quickly become an important avenue in the search for novel breast cancer biomarkers. We provide a brief history of breast cancer chemotherapeutics and explore the emerging field of circulating (blood‐borne) miRNAs as breast cancer biomarkers for the neoadjuvant treatment of breast cancer. Established molecular markers of breast cancer are outlined, while the potential role of circulating miRNAs as chemotherapeutic response predictors, prognosticators or potential therapeutic targets is discussed.
Collapse
Affiliation(s)
- Máire-Caitlín Casey
- Discipline of Surgery, School of Medicine, National University of Ireland, Galway, Ireland
| | - Karl J Sweeney
- Discipline of Surgery, School of Medicine, National University of Ireland, Galway, Ireland
| | | | - Michael J Kerin
- Discipline of Surgery, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
34
|
Decoupling of the PI3K Pathway via Mutation Necessitates Combinatorial Treatment in HER2+ Breast Cancer. PLoS One 2015; 10:e0133219. [PMID: 26181325 PMCID: PMC4504492 DOI: 10.1371/journal.pone.0133219] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 06/23/2015] [Indexed: 12/19/2022] Open
Abstract
We report here on experimental and theoretical efforts to determine how best to combine drugs that inhibit HER2 and AKT in HER2+ breast cancers. We accomplished this by measuring cellular and molecular responses to lapatinib and the AKT inhibitors (AKTi) GSK690693 and GSK2141795 in a panel of 22 HER2+ breast cancer cell lines carrying wild type or mutant PIK3CA. We observed that combinations of lapatinib plus AKTi were synergistic in HER2+/PIK3CAmut cell lines but not in HER2+/PIK3CAwt cell lines. We measured changes in phospho-protein levels in 15 cell lines after treatment with lapatinib, AKTi or lapatinib + AKTi to shed light on the underlying signaling dynamics. This revealed that p-S6RP levels were less well attenuated by lapatinib in HER2+/PIK3CAmut cells compared to HER2+/PIK3CAwt cells and that lapatinib + AKTi reduced p-S6RP levels to those achieved in HER2+/PIK3CAwt cells with lapatinib alone. We also found that that compensatory up-regulation of p-HER3 and p-HER2 is blunted in PIK3CAmut cells following lapatinib + AKTi treatment. Responses of HER2+ SKBR3 cells transfected with lentiviruses carrying control or PIK3CAmut sequences were similar to those observed in HER2+/PIK3CAmut cell lines but not in HER2+/PIK3CAwt cell lines. We used a nonlinear ordinary differential equation model to support the idea that PIK3CA mutations act as downstream activators of AKT that blunt lapatinib inhibition of downstream AKT signaling and that the effects of PIK3CA mutations can be countered by combining lapatinib with an AKTi. This combination does not confer substantial benefit beyond lapatinib in HER2+/PIK3CAwt cells.
Collapse
|
35
|
Yélamos J, Galindo M, Navarro J, Albanell J, Rovira A, Rojo F, Oliver J. Enhancing tumor-targeting monoclonal antibodies therapy by PARP inhibitors. Oncoimmunology 2015; 5:e1065370. [PMID: 26942084 DOI: 10.1080/2162402x.2015.1065370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022] Open
Abstract
Monoclonal antibodies (mAbs) have become a successful therapeutic approach in cancer. However, some patients do not achieve long-term clinical benefit and most mAbs only exert modest effects as monotherapies. Therefore, combinations with chemotherapy are currently being investigated. Emerging studies have shown a synergistic therapeutic effect of PARP inhibitors and mAbs in cancer. PARP enzymes catalytically cleave β-NAD+ and transfer the ADP-ribose moiety to acceptor proteins, modifying their function. In here, we update recent data about the therapeutic effect of the combination of PARP inhibitors with mAbs in cancer treatment and discuss the molecular mechanisms involved in this synergy.
Collapse
Affiliation(s)
- José Yélamos
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Immunology, Hospital del Mar, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Miguel Galindo
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM) , Barcelona, Spain
| | - Judith Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM) , Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Ana Rovira
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Oncology, Hospital del Mar, Barcelona, Spain
| | - Federico Rojo
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Patology, IIS Fundación Jiménez Díaz, Madrid, Spain
| | - Javier Oliver
- Department of Cell Biology and Immunology; Instituto de Parasitología y Biomedicina López Neyra , Granada, Spain
| |
Collapse
|
36
|
Chung A, Choi M, Han BC, Bose S, Zhang X, Medina-Kauwe L, Sims J, Murali R, Taguiam M, Varda M, Schiff R, Giuliano A, Cui X. Basal Protein Expression Is Associated With Worse Outcome and Trastuzamab Resistance in HER2+ Invasive Breast Cancer. Clin Breast Cancer 2015; 15:448-457.e2. [PMID: 26248960 DOI: 10.1016/j.clbc.2015.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/29/2015] [Accepted: 06/11/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND We investigated the effect of basal protein expression on trastuzamab response in patients with HER2-positive (HER2(+)) breast cancer who received trastuzamab (T) and in HER2(+) breast cancer cell lines. PATIENTS AND METHODS Expression of cytokeratin (CK) 5/6, CK14, and epidermal growth factor receptor (EGFR) was evaluated after immunohistochemical staining in paraffin-embedded tissue of 97 patients with stage I to III HER2(+) breast cancer treated with chemotherapy/T. Groups with and without basal protein expression were compared with respect to clinicopathologic parameters and survival. We treated 4 cell lines (2 basal-HER2 [HCC1569, HCC1954] and 2 nonbasal HER2 [BT474, SKBR3]) each with vehicle, T 20 μg/mL, paclitaxel 0.01 μM (P), and T with P (T + P). Cell viability was assessed and HER2 pathway suppression was compared between groups using immunoblot analysis. Mammosphere formation was used to assess breast cancer stem cell properties. RESULTS EGFR expression was significantly associated with cancer-specific survival (CSS) (P = .05). CK5/6 expression strongly correlated with overall and disease-free survival, and CSS (P = .03, P = .04, and P = .03, respectively). Statistical significance was maintained for EGFR and CK5/6 after adjustment for covariates. CK14 was not associated with survival. All cell lines expressed similar levels of HER2. T and P alone inhibited proliferation of nonbasal cell lines; T + P had an additive cytotoxic effect. Basal cells were resistant to T, P inhibited proliferation, but T + P had no additive cytotoxic effect on cell growth in basal cells. Immunoblot analysis showed a significant decrease in phosphorylated Akt levels after treatment with T or T + P in nonbasal cells but not in basal cells. Akt blockade suppressed growth of basal and nonbasal HER2(+) cells. Furthermore, basal HER2 cell lines had increased mammosphere formation, which suggests increased stem cell properties compared with nonbasal HER2 cell lines. CONCLUSION CK5/6 and EGFR expression are predictive of worse prognosis in HER2(+) breast cancer patients treated with T. Basal HER2 breast cancer cell lines are resistant to trastuzamab, which is mediated through the Akt pathway; AKT inhibition abrogates this resistance. Basal HER2 cell lines also have increased stem cell properties, which might play a role in the resistance pathway.
Collapse
Affiliation(s)
- Alice Chung
- Cedars-Sinai Medical Center, Los Angeles, CA.
| | | | | | - Shikha Bose
- Cedars-Sinai Medical Center, Los Angeles, CA
| | - Xiao Zhang
- Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lemos LGT, Victorino VJ, Herrera ACSA, Aranome AMF, Cecchini AL, Simão ANC, Panis C, Cecchini R. Trastuzumab-based chemotherapy modulates systemic redox homeostasis in women with HER2-positive breast cancer. Int Immunopharmacol 2015; 27:8-14. [PMID: 25937481 DOI: 10.1016/j.intimp.2015.04.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/07/2015] [Accepted: 04/17/2015] [Indexed: 12/22/2022]
Abstract
Trastuzumab is an immunotargeting therapeutic against breast tumors with amplification of the human epithelial growth factor receptor 2 (HER2). HER2 patients naturally exhibit disruption in the pro-oxidant inflammatory profiling; however, the impact of trastuzumab-based chemotherapy in modulating this process is still unknown. Here we determined the systemic pro-inflammatory profile of women diagnosed with HER2-amplified tumors, undergoing trastuzumab-based chemotherapy (TZ), and compared the results with that of healthy controls (CTR) and untreated patients with HER2-amplified breast cancer (CA). The plasmatic inflammatory profile was assessed by evaluating pro-oxidant parameters such as lipid peroxidation, total antioxidant capacity (TRAP), levels of advanced oxidation protein products (AOPPs), nitric oxide (NO), C-reactive protein (CRP), and total thiol content. Markers of cardiac damage were also assessed. Our findings showed increased NO levels in TZ than that in either CA or CTR groups. Furthermore, TZ augmented TRAP and reduced total thiol than that of the CA group. Our data also revealed that AOPP levels were significantly higher in the TZ than the CA group. AOPP and the MB fraction of creatine-kinase (CKMB) levels were positively correlated in TZ patients. These findings suggest that trastuzumab-associated chemotherapy can modulate the pro-inflammatory markers of HER2-positive breast cancer patients to the levels found in healthy controls.
Collapse
Affiliation(s)
- L G T Lemos
- Bone Marrow Transplantation Center, National Cancer Institute, INCA, Rio de Janeiro, Brazil
| | - V J Victorino
- Faculty of Medicine, University of São Paulo, USP, São Paulo, Brazil
| | - A C S A Herrera
- Bone Marrow Transplantation Center, National Cancer Institute, INCA, Rio de Janeiro, Brazil
| | - A M F Aranome
- Laboratory of Physiopathology and Free Radicals, Londrina State University, UEL, Londrina, Paraná, Brazil
| | - A L Cecchini
- Laboratory of Physiopathology and Free Radicals, Londrina State University, UEL, Londrina, Paraná, Brazil
| | - A N C Simão
- Universitary Hospital, Department of Pharmacy, State University of Londrina, Londrina, Paraná, Brazil
| | - C Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Francisco Beltrão, Paraná, Brazil.
| | - R Cecchini
- Laboratory of Physiopathology and Free Radicals, Londrina State University, UEL, Londrina, Paraná, Brazil
| |
Collapse
|
38
|
Sun Y, Feng X, Qu J, Han W, Liu Z, Li X, Zou M, Zhen Y, Zhu J. Expression and Characterization of the Extracellular Domain of Human HER2 from Escherichia Coli, and Production of Polyclonal Antibodies Against the Recombinant Proteins. Appl Biochem Biotechnol 2015; 176:1029-43. [PMID: 25906688 DOI: 10.1007/s12010-015-1627-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 04/08/2015] [Indexed: 10/23/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a member of the epidermal growth factor receptor (EGFR) family. In this study, the whole extracellular domain gene of HER2 was amplified by RT-PCR from human breast cancer cell line SK-BR-3. The genes of membrane-distal region (A) and membrane proximal region (B) of HER2 extracellular domain were amplified from the cloned template, and then inserted into the expression vector pET-28a and pET-30a, respectively. The recombinant expression vectors were transformed into Escherichia coli BL21 (DE3) cells and induced by isopropyl-b-D-thiogalactopyranoside (IPTG) for expression of proteins His-A and His-B. The expressed proteins were detected by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blot. The optimization of culture conditions led us to accomplish the recombinant protein induction with 1.0 mM IPTG at 37 °C for 8 h, and both proteins were expressed in the insoluble form. Both proteins were purified under the denaturing condition using Ni-NTA sepharose column. Balb/c mice were immunized with the purified proteins and then effectively produced polyclonal antibodies, which reached to a relatively high titer by ELISA testing and had good specificity by western blot detection. The HER2 ECD proteins His-A and His-B could be expressed in E. coli and were suitable for production of high titer antibodies against HER2 ECD.
Collapse
Affiliation(s)
- Yong Sun
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Li Z, Dabbs DJ, Cooper KL, Bhargava R. Dual HER2 gene protein assay: focused study of breast cancers with 2+ immunohistochemical expression. Am J Clin Pathol 2015; 143:451-8. [PMID: 25696805 DOI: 10.1309/ajcpkivvw4obpx6i] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVES The combined gene protein assay (GPA) can simultaneously assess HER2 gene copy number and protein on a single slide using bright-field microscopy. METHODS GPA was compared with a fluorescence in situ hybridization (FISH) assay on 50 invasive breast carcinomas with a 2+ score on immunohistochemistry (IHC). RESULTS The cases were categorized into positive, equivocal, or negative for HER2 gene amplification using the 2013 American Society of Clinical Oncology/College of American Pathologists criteria. This resulted in 82% agreement (41 of 50) between FISH and GPA. In addition, 25 known IHC 3+ breast carcinomas analyzed by GPA showed protein overexpression and clusters of HER2 gene consistent with unequivocal amplification, and 22 known IHC-negative cases were negative for HER2 gene amplification by GPA. CONCLUSIONS Although GPA is an alternative to both IHC and FISH, it may be an unnecessary test for IHC 0/1+/3+ cases. The clinical utility of GPA appears to be similar to other in situ hybridization assays (ie, adjudicator of HER2 status for IHC 2+ cases).
Collapse
Affiliation(s)
- Zaibo Li
- Magee-Womens Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA
| | - David J. Dabbs
- Magee-Womens Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Kristine L. Cooper
- University of Pittsburgh Cancer Institute Biostatistics Facility, Pittsburgh, PA
| | - Rohit Bhargava
- Magee-Womens Hospital of University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
40
|
Seiser EL, Innocenti F. Hidden Markov Model-Based CNV Detection Algorithms for Illumina Genotyping Microarrays. Cancer Inform 2015; 13:77-83. [PMID: 25657572 PMCID: PMC4310714 DOI: 10.4137/cin.s16345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/18/2014] [Accepted: 11/21/2014] [Indexed: 12/24/2022] Open
Abstract
Somatic alterations in DNA copy number have been well studied in numerous malignancies, yet the role of germline DNA copy number variation in cancer is still emerging. Genotyping microarrays generate allele-specific signal intensities to determine genotype, but may also be used to infer DNA copy number using additional computational approaches. Numerous tools have been developed to analyze Illumina genotype microarray data for copy number variant (CNV) discovery, although commonly utilized algorithms freely available to the public employ approaches based upon the use of hidden Markov models (HMMs). QuantiSNP, PennCNV, and GenoCN utilize HMMs with six copy number states but vary in how transition and emission probabilities are calculated. Performance of these CNV detection algorithms has been shown to be variable between both genotyping platforms and data sets, although HMM approaches generally outperform other current methods. Low sensitivity is prevalent with HMM-based algorithms, suggesting the need for continued improvement in CNV detection methodologies.
Collapse
Affiliation(s)
- Eric L Seiser
- Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Federico Innocenti
- Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. ; UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
41
|
T-DM1, a novel antibody-drug conjugate, is highly effective against uterine and ovarian carcinosarcomas overexpressing HER2. Clin Exp Metastasis 2014; 32:29-38. [PMID: 25398397 DOI: 10.1007/s10585-014-9688-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/08/2014] [Indexed: 12/15/2022]
Abstract
Ovarian and uterine carcinosarcoma (CS) are characterized by their aggressive clinical behavior and poor prognosis. We evaluated the efficacy of trastuzumab-emtansine (T-DM1), against primary HER2 positive and HER2 negative CS cell lines in vitro and in vivo. Eight primary CS cell lines were evaluated for HER2 amplification and protein expression by fluorescence in situ hybridization, immunohistochemistry, flow cytometry and qRT-PCR. Sensitivity to T-DM1-induced antibody-dependent-cell-mediated-cytotoxicity (ADCC) was evaluated in 4-h-chromium-release-assays. T-DM1 cytostatic and apoptotic activities were evaluated using flow cytometry based proliferation assays. In vivo activity of T-DM1 was also evaluated. HER2 protein overexpression and gene amplification were detected in 25 % (2/8) of the primary CS cell lines. T-DM1 and T were similarly effective in inducing strong ADCC against CS overexpressing HER2 at 3+ levels. In contrast, T-DM1 was dramatically more effective than T in inhibiting cell proliferation (P < 0.0001) and in inducing G2/M phase cell cycle arrest in the HER2 expressing cell lines (shift of G2/M: mean ± SEM from 14.87 ± 1.23 to 66.57 ± 4.56 %, P < 0.0001). Importantly, T-DM1 was highly active at reducing tumor formation in vivo in CS xenografts overexpressing HER2 (P = 0.0001 and P < 0.0001 compared to T and vehicle respectively) with a significantly longer survival when compared to T and vehicle mice (P = 0.008 and P = 0.0001 respectively). T-DM1 may represent a novel treatment option for the subset of HER2 positive CS patients with disease refractory to chemotherapy.
Collapse
|
42
|
von der Heyde S, Bender C, Henjes F, Sonntag J, Korf U, Beißbarth T. Boolean ErbB network reconstructions and perturbation simulations reveal individual drug response in different breast cancer cell lines. BMC SYSTEMS BIOLOGY 2014; 8:75. [PMID: 24970389 PMCID: PMC4087127 DOI: 10.1186/1752-0509-8-75] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 06/10/2014] [Indexed: 12/19/2022]
Abstract
Background Despite promising progress in targeted breast cancer therapy, drug resistance remains challenging. The monoclonal antibody drugs trastuzumab and pertuzumab as well as the small molecule inhibitor erlotinib were designed to prevent ErbB-2 and ErbB-1 receptor induced deregulated protein signalling, contributing to tumour progression. The oncogenic potential of ErbB receptors unfolds in case of overexpression or mutations. Dimerisation with other receptors allows to bypass pathway blockades. Our intention is to reconstruct the ErbB network to reveal resistance mechanisms. We used longitudinal proteomic data of ErbB receptors and downstream targets in the ErbB-2 amplified breast cancer cell lines BT474, SKBR3 and HCC1954 treated with erlotinib, trastuzumab or pertuzumab, alone or combined, up to 60 minutes and 30 hours, respectively. In a Boolean modelling approach, signalling networks were reconstructed based on these data in a cell line and time course specific manner, including prior literature knowledge. Finally, we simulated network response to inhibitor combinations to detect signalling nodes reflecting growth inhibition. Results The networks pointed to cell line specific activation patterns of the MAPK and PI3K pathway. In BT474, the PI3K signal route was favoured, while in SKBR3, novel edges highlighted MAPK signalling. In HCC1954, the inferred edges stimulated both pathways. For example, we uncovered feedback loops amplifying PI3K signalling, in line with the known trastuzumab resistance of this cell line. In the perturbation simulations on the short-term networks, we analysed ERK1/2, AKT and p70S6K. The results indicated a pathway specific drug response, driven by the type of growth factor stimulus. HCC1954 revealed an edgetic type of PIK3CA-mutation, contributing to trastuzumab inefficacy. Drug impact on the AKT and ERK1/2 signalling axes is mirrored by effects on RB and RPS6, relating to phenotypic events like cell growth or proliferation. Therefore, we additionally analysed RB and RPS6 in the long-term networks. Conclusions We derived protein interaction models for three breast cancer cell lines. Changes compared to the common reference network hint towards individual characteristics and potential drug resistance mechanisms. Simulation of perturbations were consistent with the experimental data, confirming our combined reverse and forward engineering approach as valuable for drug discovery and personalised medicine.
Collapse
Affiliation(s)
| | | | | | | | | | - Tim Beißbarth
- Statistical Bioinformatics, Department of Medical Statistics, University Medical Center Göttingen, Humboldtallee 32, 37073 Göttingen, Germany.
| |
Collapse
|
43
|
Neapolitan R, Xue D, Jiang X. Modeling the altered expression levels of genes on signaling pathways in tumors as causal bayesian networks. Cancer Inform 2014; 13:77-84. [PMID: 24932098 PMCID: PMC4051800 DOI: 10.4137/cin.s13578] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 01/05/2023] Open
Abstract
This paper concerns a study indicating that the expression levels of genes in signaling pathways can be modeled using a causal Bayesian network (BN) that is altered in tumorous tissue. These results open up promising areas of future research that can help identify driver genes and therapeutic targets. So, it is most appropriate for the cancer informatics community. Our central hypothesis is that the expression levels of genes that code for proteins on a signal transduction network (STP) are causally related and that this causal structure is altered when the STP is involved in cancer. To test this hypothesis, we analyzed 5 STPs associated with breast cancer, 7 STPs associated with other cancers, and 10 randomly chosen pathways, using a breast cancer gene expression level dataset containing 529 cases and 61 controls. We identified all the genes related to each of the 22 pathways and developed separate gene expression datasets for each pathway. We obtained significant results indicating that the causal structure of the expression levels of genes coding for proteins on STPs, which are believed to be implicated in both breast cancer and in all cancers, is more altered in the cases relative to the controls than the causal structure of the randomly chosen pathways.
Collapse
Affiliation(s)
- Richard Neapolitan
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Diyang Xue
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xia Jiang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Mocanu MM, Ganea C, Georgescu L, Váradi T, Shrestha D, Baran I, Katona E, Nagy P, Szöllősi J. Epigallocatechin 3-O-gallate induces 67 kDa laminin receptor-mediated cell death accompanied by downregulation of ErbB proteins and altered lipid raft clustering in mammary and epidermoid carcinoma cells. JOURNAL OF NATURAL PRODUCTS 2014; 77:250-7. [PMID: 24456004 DOI: 10.1021/np4007712] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Since the administration of synthetic medicines is associated with drug resistance and undesired side effects, utilization of natural compounds could be an alternative and complementary modality to inhibit or prevent the development of tumors. Epigallocatechin 3-O-gallate (EGCG, 1), the major flavan component of green tea, and genistein (2), a soy isoflavonoid, are known to have chemopreventive and chemotherapeutic effects against cancer. This study demonstrated that both flavonoids inhibit cell proliferation, an effect enhanced under serum-free conditions. Compound 1, but not 2, induced downregulation of ErbB1 and ErbB2 in mammary and epidermoid carcinoma cells, and its inhibitory effect on cell viability was mediated by the 67 kDa laminin receptor (67LR). While 1 was superior in inducing cell death, 2 was more efficient in arresting the tumor cells in the G2/M phase. Furthermore, number and brightness analysis revealed that 1 decreased the homoclustering of a lipid raft marker, glycosylphosphatidylinositol-anchored GFP, and it also reduced the co-localization between lipid rafts and 67LR. The main conclusion made is that the primary target of 1 may be the lipid raft component of the plasma membrane followed by secondary changes in the expression of ErbB proteins. Compound 2, on the other hand, must have other unidentified targets.
Collapse
Affiliation(s)
- Maria-Magdalena Mocanu
- Department of Biophysics, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy , 050474 Bucharest, Romania
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chung A, Cui X, Audeh W, Giuliano A. Current status of anti-human epidermal growth factor receptor 2 therapies: predicting and overcoming herceptin resistance. Clin Breast Cancer 2014; 13:223-32. [PMID: 23829888 DOI: 10.1016/j.clbc.2013.04.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 11/29/2022]
Abstract
Human epidermal growth factor receptor 2-overexpressing (HER2+) breast cancer occurs in 20% to 25% of cases and is associated with poor prognosis. Trastuzumab (Herceptin; Genentech, South San Francisco, CA) is a monoclonal antibody targeting the HER2 extracellular domain that has been shown to significantly reduce relapse rates. However, some patients with HER2+ tumors do not respond to Herceptin, and 60% to 85% of patients with HER2+ metastatic breast cancer acquire resistance within a short time period. In this review, we discuss proposed mechanisms of action of trastuzumab and trastuzumab resistance and various drugs that have been developed to overcome drug resistance. We introduce the basal molecular subtype as a predictor of increased risk in HER2+ breast cancer and a possible alternative cause of drug resistance.
Collapse
Affiliation(s)
- Alice Chung
- Department of Surgery, Division of Surgical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | |
Collapse
|
46
|
[Predictive biomarkers for tumor-relevant signaling pathways in molecular pathology]. DER PATHOLOGE 2014; 35:93-105. [PMID: 24496994 DOI: 10.1007/s00292-013-1798-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In the era of personalized medicine, targeted cancer treatments aim to improve therapy and overall survival. Specific therapies are individually customized for patients based on molecular alterations of the neoplastic cells. The pathologist has a central role in the identification and characterization of a variety of molecular markers that can be used to better predict outcome and assess therapeutic success of specific targeted approaches.
Collapse
|
47
|
Sedgewick AJ, Benz SC, Rabizadeh S, Soon-Shiong P, Vaske CJ. Learning subgroup-specific regulatory interactions and regulator independence with PARADIGM. Bioinformatics 2013; 29:i62-70. [PMID: 23813010 PMCID: PMC3694636 DOI: 10.1093/bioinformatics/btt229] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
UNLABELLED High-dimensional '-omics' profiling provides a detailed molecular view of individual cancers; however, understanding the mechanisms by which tumors evade cellular defenses requires deep knowledge of the underlying cellular pathways within each cancer sample. We extended the PARADIGM algorithm (Vaske et al., 2010, Bioinformatics, 26, i237-i245), a pathway analysis method for combining multiple '-omics' data types, to learn the strength and direction of 9139 gene and protein interactions curated from the literature. Using genomic and mRNA expression data from 1936 samples in The Cancer Genome Atlas (TCGA) cohort, we learned interactions that provided support for and relative strength of 7138 (78%) of the curated links. Gene set enrichment found that genes involved in the strongest interactions were significantly enriched for transcriptional regulation, apoptosis, cell cycle regulation and response to tumor cells. Within the TCGA breast cancer cohort, we assessed different interaction strengths between breast cancer subtypes, and found interactions associated with the MYC pathway and the ER alpha network to be among the most differential between basal and luminal A subtypes. PARADIGM with the Naive Bayesian assumption produced gene activity predictions that, when clustered, found groups of patients with better separation in survival than both the original version of PARADIGM and a version without the assumption. We found that this Naive Bayes assumption was valid for the vast majority of co-regulators, indicating that most co-regulators act independently on their shared target. AVAILABILITY http://paradigm.five3genomics.com. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Andrew J Sedgewick
- Joint Carnegie Mellon - University of Pittsburgh Ph.D Program in Computational Biology, Pittsburgh, PA 15260, USA
| | | | | | | | | |
Collapse
|
48
|
Luis M, Tavares A, Carvalho LS, Lara-Santos L, Araújo A, Mello RAD. Personalizing therapies for gastric cancer: molecular mechanisms and novel targeted therapies. World J Gastroenterol 2013; 19:6383-6397. [PMID: 24151357 PMCID: PMC3801309 DOI: 10.3748/wjg.v19.i38.6383] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 07/24/2013] [Accepted: 08/05/2013] [Indexed: 02/06/2023] Open
Abstract
Globally, gastric cancer is the 4(th) most frequently diagnosed cancer and the 2(nd) leading cause of death from cancer, with an estimated 990000 new cases and 738000 deaths registered in 2008. In the advanced setting, standard chemotherapies protocols acquired an important role since last decades in prolong survival. Moreover, recent advances in molecular therapies provided a new interesting weapon to treat advanced gastric cancer through anti-human epidermal growth factor receptor 2 (HER2) therapies. Trastuzumab, an anti-HER2 monoclonal antibody, was the first target drug in the metastatic setting that showed benefit in overall survival when in association with platinum-5-fluorouracil based chemotherapy. Further, HER2 overexpression analysis acquired a main role in predict response for trastuzumab in this field. Thus, we conducted a review that will discuss the main points concerning trastuzumab and HER2 in gastric cancer, providing a comprehensive overview of molecular mechanisms and novel trials involved.
Collapse
|
49
|
Update on Targets and Novel Treatment Options for High-Grade Osteosarcoma and Chondrosarcoma. Hematol Oncol Clin North Am 2013; 27:1021-48. [DOI: 10.1016/j.hoc.2013.07.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Abstract
HER2 is a trans-membrane receptor tyrosine kinase that activates multiple growth-promoting signaling pathways including PI3K-AKT and Ras-MAPK. Dysregulation of HER2 is a frequent occurrence in breast cancer that is associated with poor patient outcomes. A primary function of HER2 is suppressing apoptosis to enhance cell survival giving rise to uncontrolled proliferation and tumor growth. There has been much investigation into the mechanisms by which apoptosis is suppressed by HER2 in hopes of finding clinical targets for HER2-positive breast cancers as these cancers often become resistant to therapies that directly target HER2. Several apoptotic mechanisms have been shown to be deregulated in HER2-overexpressing cells with examples in both the intrinsic and extrinsic apoptotic pathways. HER2-mediated activation of PI3K-AKT signaling is required for many of the mechanisms HER2 uses to suppress apoptosis. HER2 overexpression is correlated with increases in anti-apoptotic Bcl-2 proteins including Bcl-2, Bcl-xL, and Mcl-1. HER2 also suppresses p53-mediated apoptosis by upregulation of MDM2 by activation of AKT. In addition, survivin expression is often increased with HER2 overexpression leading to inhibition of caspase activation. There is also recent evidence to suggest HER2 can directly influence apoptosis by translocation to the mitochondria to inhibit cytochrome c release. HER2 can also suppress cellular reaction to death ligands, especially TRAIL-induced apoptosis. Elucidation of the mechanisms of apoptotic suppression by HER2 suggest that clinical treatment will likely need to target multiple components of these pathways as there is redundancy in HER2-mediated cell survival. Several therapies have attempted to target Bcl-2 proteins that have promising pre-clinical results. Next-generation HER2 targeting therapies include irreversible pan-ERBB inhibitors and antibody-drug conjugates, such as T-DM1 that has very promising clinical results thus far. Further investigation should include elucidating mechanisms of resistance to HER2-targeted therapies and targeting of multiple components of HER2-mediated cell survival.
Collapse
Affiliation(s)
- Richard L Carpenter
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Hui-Wen Lo
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina 27710, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|