1
|
Veney DJ, Wei LY, Toland AE, Presley CJ, Hampel HL, Padamsee TJ, Lee CN, Irvin WJ, Bishop MJ, Kim JJ, Hovick SR, Senter LA, Stover DG. A video intervention to improve patient understanding of tumor genomic testing in patients with cancer. Cancer Med 2024; 13:e70095. [PMID: 39258462 PMCID: PMC11387988 DOI: 10.1002/cam4.70095] [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: 02/02/2024] [Revised: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Tumor genomic testing (TGT) is standard-of-care for most patients with advanced/metastatic cancer. Despite established guidelines, patient education prior to TGT is frequently omitted. The purpose of this study was to evaluate the impact of a concise 4 min video for patient education prior to TGT. METHODS Based on a quality improvement cycle, an animated video was created to be applicable to any cancer type, incorporating culturally diverse images, available in English and Spanish. Patients undergoing standard-of-care TGT were enrolled at a tertiary academic institution and completed survey instruments prior to video viewing (T1) and immediately post-viewing (T2). Instruments included: (1) 10-question objective genomic knowledge; (2) 10-question video message-specific knowledge; (3) 11-question Trust in Provider; (4) attitudes regarding TGT. RESULTS A total of 150 participants were enrolled. For the primary objective, there was a significant increase in video message-specific knowledge (median 10 point increase; p < 0.0001) with no significant change in genomic knowledge/understanding (p = 0.89) or trust in physician/provider (p = 0.59). Results for five questions significantly improved, including the likelihood of TGT impact on treatment decision, incidental germline findings, and cost of testing. Improvement in video message-specific knowledge was consistent across demographic groups, including age, income, and education. CONCLUSIONS A concise, 3-4 min, broadly applicable video incorporating culturally diverse images administered prior to TGT significantly improved video message-specific knowledge across all demographic groups. This resource is publicly available at http://www.tumor-testing.com, with a goal to efficiently educate and empower patients regarding TGT while addressing guidelines within the flow of clinical practice.
Collapse
Affiliation(s)
- Deloris J. Veney
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Lai Y. Wei
- Department of Biomedical InformaticsOhio State UniversityColumbusOhioUSA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
- Department of Cancer Biology and GeneticsThe Ohio State UniversityColumbusOhioUSA
| | - Carolyn J. Presley
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Heather L. Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics ResearchCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Tasleem J. Padamsee
- Division of Health Services Management and PolicyCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
| | - Clara N. Lee
- Division of Health Services Management and PolicyCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
- Present address:
Division of Plastic and Reconstructive SurgeryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | | | | | - James J. Kim
- Bon Secours‐Mercy Health St. ElizabethYoungstownOhioUSA
| | | | - Leigha A. Senter
- Division of Human Genetics, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Daniel G. Stover
- Division of Medical OncologyOhio State University Comprehensive Cancer CenterColumbusOhioUSA
- Department of Biomedical InformaticsOhio State UniversityColumbusOhioUSA
- Pelotonia Institute for Immuno‐Oncology, Ohio State University Comprehensive CancerColumbusOhioUSA
| |
Collapse
|
2
|
Veney D, Wei L, Toland AE, Presley CJ, Hampel H, Padamsee TJ, Lee CN, Irvin WJ, Bishop M, Kim J, Hovick SR, Senter L, Stover DG. A Video Intervention to Improve Patient Understanding of Tumor Genomic Testing in Patients with Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.05.23299443. [PMID: 38106014 PMCID: PMC10723483 DOI: 10.1101/2023.12.05.23299443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Tumor genomic testing (TGT) has become standard-of-care for most patients with advanced/metastatic cancer. Despite established guidelines, patient education prior to TGT is variable or frequently omitted. The purpose of this study was to evaluate the impact of a concise (3-4 minute) video for patient education prior to TGT. Methods Based on a quality improvement cycle, an animated video was created to be applicable to any cancer type, incorporating culturally diverse images, available in English and Spanish. Patients undergoing standard-of care TGT were enrolled at a tertiary academic institution and completed validated survey instruments immediately prior to video viewing (T1) and immediately post-viewing (T2). Instruments included: 1) 10-question objective genomic knowledge/understanding; 2) 10-question video message-specific knowledge/recall; 3) 11-question Trust in Physician/Provider; 4) attitudes regarding TGT. The primary objective was change in outcomes from before to after the video was assessed with Wilcoxon signed rank test. Results From April 2022 to May 2023, a total of 150 participants were enrolled (MBC n=53, LC n=38, OC n=59). For the primary endpoint, there was a significant increase in video message-specific knowledge (median 10 point increase; p<0.0001) with no significant change in genomic knowledge/understanding (p=0.89) or Trust in Physician/Provider (p=0.59). Results for five questions significantly improved, including the likelihood of TGT impact on treatment decision, incidental germline findings, and cost of testing. Improvement in video message-specific knowledge was consistent across demographic groups, including age, income, and education. Individuals with less educational attainment had had greater improvement from before to after video viewing. Conclusions A concise, 3-4 minute, broadly applicable video incorporating culturally diverse images administered prior to TGT significantly improved video message-specific knowledge across all demographic groups. This resource is publicly available at http://www.tumor-testing.com, with a goal to efficiently educate and empower patients regarding TGT while addressing guidelines within the flow of clinical practice. Clinical Trial Registration ClinicalTrials.gov NCT05215769.
Collapse
Affiliation(s)
- Deloris Veney
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
| | - Lai Wei
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210 USA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210 USA
| | - Carolyn J. Presley
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
| | - Heather Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010
| | - Tasleem J. Padamsee
- Division of Health Services Management and Policy, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Clara N. Lee
- Division of Health Services Management and Policy, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - William J. Irvin
- Bon Secours Cancer Institute at St. Francis, Richmond, Virginia, USA
| | | | - James Kim
- Bon Secours-Mercy Health St. Elizabeth, Youngstown, Ohio, USA
| | - Shelly R. Hovick
- School of Communication, Ohio State University, Columbus, OH, 43210 USA
| | - Leigha Senter
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
| | - Daniel G. Stover
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210 USA
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210 USA
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer, Columbus, OH, 43210 USA
| |
Collapse
|
3
|
Furtney I, Bradley R, Kabuka MR. Patient Graph Deep Learning to Predict Breast Cancer Molecular Subtype. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:3117-3127. [PMID: 37379184 PMCID: PMC10623656 DOI: 10.1109/tcbb.2023.3290394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Breast cancer is a heterogeneous disease consisting of a diverse set of genomic mutations and clinical characteristics. The molecular subtypes of breast cancer are closely tied to prognosis and therapeutic treatment options. We investigate using deep graph learning on a collection of patient factors from multiple diagnostic disciplines to better represent breast cancer patient information and predict molecular subtype. Our method models breast cancer patient data into a multi-relational directed graph with extracted feature embeddings to directly represent patient information and diagnostic test results. We develop a radiographic image feature extraction pipeline to produce vector representation of breast cancer tumors in DCE-MRI and an autoencoder-based genomic variant embedding method to map variant assay results to a low-dimensional latent space. We leverage related-domain transfer learning to train and evaluate a Relational Graph Convolutional Network to predict the probabilities of molecular subtypes for individual breast cancer patient graphs. Our work found that utilizing information from multiple multimodal diagnostic disciplines improved the model's prediction results and produced more distinct learned feature representations for breast cancer patients. This research demonstrates the capabilities of graph neural networks and deep learning feature representation to perform multimodal data fusion and representation in the breast cancer domain.
Collapse
|
4
|
Senter L, Veney D, Surplus T, Haynam M, Adams EJ, Hampel H, Toland AE, Presley CJ, Padamsee TJ, Lee CN, Hovick SR, Stover DG. Patient Understanding of Tumor Genomic Testing: A Quality Improvement Effort. JCO Oncol Pract 2023; 19:e8-e14. [PMID: 36130146 PMCID: PMC10166357 DOI: 10.1200/op.22.00316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/29/2022] [Accepted: 08/16/2022] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Tumor genomic testing (TGT) has become increasingly adopted as part of standard cancer care for many cancers. Despite national guidelines around patient education before TGT, available evidence suggests that most patients' understanding of genomics remains limited, particularly lower-income and minority patients, and most patients are not informed regarding potential incidental germline findings. METHODS To investigate and address limitations in patient understanding of TGT results, a Plan-Do-Study-Act (PDSA) approach is being used to assess needs, identify opportunities for improvement, and implement approaches to optimize patient education. We reviewed published guidelines related to pre-TGT provider-patient education and to identify key points (Plan). A provider quality improvement survey was completed (Do), which highlighted inconsistency in pre-TGT discussion practice across providers and minimal discussion with patients regarding the possibility of incidental germline findings. RESULTS Patient focus groups and interviews (N = 12 patients) were completed with coding of each transcript (Study), which revealed themes including trouble differentiating TGT from other forms of testing, yet understanding that results could tailor therapy. The integration of data across this initial PDSA cycle identified consistent themes and opportunities, which were incorporated into a patient-directed, concise animated video for pre-TGT education (Act), which will form the foundation of a subsequent PDSA cycle. The video addresses how TGT may/may not inform treatment, the process for TGT using existing tissue or liquid biopsy, insurance coverage, and the potential need for germline genetics follow-up because of incidental findings. CONCLUSION This PDSA cycle reveals key gaps and opportunities for improvement in patient education before TGT.
Collapse
Affiliation(s)
- Leigha Senter
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Deloris Veney
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Taylor Surplus
- School of Communication, Ohio State University, Columbus, OH
| | - Marcy Haynam
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Elizabeth J. Adams
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Heather Hampel
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Amanda E. Toland
- Division of Human Genetics, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH
| | - Carolyn J. Presley
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Tasleem J. Padamsee
- Division of Health Services Management and Policy, College of Public Health, The Ohio State University, Columbus, OH
| | - Clara N. Lee
- Recruitment, Intervention and Survey Shared Resource, Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Daniel G. Stover
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH
- Department of Biomedical Informatics, Ohio State University, Columbus, OH
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer, Columbus, OH
| |
Collapse
|
5
|
Zhao L, Liu C, Xing Y, He J, O'Doherty J, Huang W, Zhao J. Development of a 99mTc-Labeled Single-Domain Antibody for SPECT/CT Assessment of HER2 Expression in Breast Cancer. Mol Pharm 2021; 18:3616-3622. [PMID: 34328338 DOI: 10.1021/acs.molpharmaceut.1c00569] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Accurate determination of human epidermal growth factor receptor 2 (HER2) expression is essential for HER2-targeted therapy in patients with cancer. HER2 expression in a complex environment, such as in a heterogeneous tumor, makes the precise assessment of the HER2 status difficult using current methods. In this study, we developed a novel 99mTc-labeled anti-HER2 single-domain antibody (99mTc-NM-02) as a molecular imaging tracer for the noninvasive detection of HER2 expression and investigated its safety, radiation dosimetry, biodistribution, and tumor-targeting potential in 10 patients with breast cancer. Our data showed that no drug-related adverse reactions occurred. The tracer mainly accumulated in the kidneys and liver with mild uptake in the spleen, intestines, and thyroid; however, only background tracer levels were observed in other organs where primary tumors and metastases typically occurred. The mean effective dose was 6.56 × 10-3 mSv/MBq, and tracer uptake was visually observed in the primary tumors and metastases. A maximal standard uptake value of 1.5 was determined as a reasonable cutoff for identifying HER2 positivity using SPECT/CT imaging. Our 99mTc-NM-02 tracer is safe for use in breast cancer imaging, with reasonable radiation doses, favorable biodistribution, and imaging characteristics. 99mTc-NM-02 SPECT imaging may be an accurate and noninvasive method to detect the HER2 status in patients with breast cancer.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Changcun Liu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| | - Jin He
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jim O'Doherty
- Siemens Healthineers, Malvern, Pennsylvania 19355, United States
| | - Wenhua Huang
- Nanomab Technology Limited, Shanghai 200080, China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai 200080, China
| |
Collapse
|
6
|
Canadas-Sousa A, Santos M, Medeiros R, Dias-Pereira P. Single Nucleotide Polymorphism in Prolactin Gene Is Associated With Clinical Aggressiveness and Outcome of Canine Mammary Malignant Tumors. Vet Pathol 2021; 58:1051-1057. [PMID: 34121513 DOI: 10.1177/03009858211022705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Prolactin (PRL) is a key hormone involved in canine mammary development and tumorigenesis. In this study, the influence of a single nucleotide polymorphism (SNP) in the PRL gene (rs23932236) on the clinicopathological parameters and survival of dogs with canine mammary tumors (CMTs) was investigated. A total of 206 female dogs with spontaneous mammary tumors were enrolled in this study and circulating blood cells were genotyped. This specific SNP was associated with larger size (>3 cm diameter) for malignant tumors (P = .036), tumors with infiltrative/invasive growth pattern (P = .010), vascular invasion (P = .006), and lymph node metastasis (P = .004). Carriers of the variant allele had a shorter overall survival compared to the wild-type population with an overall survival of 18.7 months and 22.7 months, respectively (P = .004). These findings suggest that SNP rs23932236 of canine PRL gene may be used as an indicator for the development of clinically aggressive forms of CMTs.
Collapse
Affiliation(s)
- Ana Canadas-Sousa
- Instituto Ciências Biomédicas Abel Salazar, ICBAS, UPorto, 89239University of Porto, Porto, Portugal
| | - Marta Santos
- Instituto Ciências Biomédicas Abel Salazar, ICBAS, UPorto, 89239University of Porto, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, 59035IPO-Porto Research Center, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Patrícia Dias-Pereira
- Instituto Ciências Biomédicas Abel Salazar, ICBAS, UPorto, 89239University of Porto, Porto, Portugal
| |
Collapse
|
7
|
Taherian-Esfahani Z, Taheri M, Dashti S, Kholghi-Oskooei V, Geranpayeh L, Ghafouri-Fard S. Assessment of the expression pattern of mTOR-associated lncRNAs and their genomic variants in the patients with breast cancer. J Cell Physiol 2019; 234:22044-22056. [PMID: 31062358 DOI: 10.1002/jcp.28767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/13/2019] [Accepted: 04/17/2019] [Indexed: 12/24/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a fundamental component of a signaling pathway that is involved in the pathogenesis of breast cancer via different mechanisms. This pathway is functionally linked with a number of small nucleolar RNA host genes (SNHGs). In the present project, we have searched for the expression quantitative trait loci (eQTLs) within SNHGs that are possibly involved in the pathogenesis of breast cancer. Following this in silico step, we have assessed expression levels of mTOR and four SNHGs in malignant and nonmalignant samples obtained from 80 patients with breast cancer. We also genotyped rs4615861 of SNHG3 and rs3087978 of SNHG5 in the peripheral blood of patients. SNHG12 expression was not detected in any of the assessed malignant or nonmalignant tissues. So this gene was excluded from further steps. Expression of mTOR and other three long noncoding RNAs (lncRNAs) were significantly increased in the malignant tissues compared with the nonmalignant tissues. When classifying patients into down-/upregulation categorized based on the transcript levels of each gene in malignant tissue versus nonmalignant tissues, we noticed associations between expression of SNHG1 and stage (p = 0.03), expression of SNHG5 and grade (p = 0.05), as well as between expression of SNHG3 and history of oral contraceptive use (p = 0.04). We also detected higher levels of SNHG3 expression in estrogen receptor/progesterone receptor (ER/PR) negative tumors compared with the ER/PR positive tumors (p = 0.003 and p = 0.01, respectively). Moreover, there was a trend toward higher expression of this lncRNA in HER2-positive tumors compared with the HER2-negative ones (p = 0.07). Combination of transcript levels of all genes could differentiate malignant tissues from nonmalignant tissues with the diagnostic power of 69% (p = 0.0001). The rs3087978 was associated with the expression of mTOR in malignant tissues in a way that TT and TG genotypes were associated with the higher and lower levels of expressions, respectively (p = 0.01). The current study underscores the significance of SNHGs in the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Zahra Taherian-Esfahani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Dashti
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Kholghi-Oskooei
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lobat Geranpayeh
- Department of Surgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Challenges and future of precision medicine strategies for breast cancer based on a database on drug reactions. Biosci Rep 2019; 39:BSR20190230. [PMID: 31387972 PMCID: PMC6732363 DOI: 10.1042/bsr20190230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/02/2019] [Accepted: 07/15/2019] [Indexed: 01/12/2023] Open
Abstract
Breast cancer (BC) is a malignancy with the highest incidence in women. Great progress has been made in research related to traditional precision medicine for BC. However, many reports have suggested that patients with BC have not benefited a lot from such progress. Thus, we analyze traditional precision medicine strategies for BC, sum up their limitations and challenges, and preliminarily propose future orientations of precision medicine strategies based on a database on drug reaction of patients with BC. According to related research, traditional precision medicine strategies for BC, which are based on molecular subtypes, perform pertinent treatments, new drug research and development according to molecular typing results. Nevertheless, these strategies still have some deficiencies. First, there are very few patients with each molecular subtype, the match ratio of drugs is low. Second, these strategies can not solve the problem of poor drug sensitivity resulting from heterogeneity. The main strategy we put forward in the present paper is based on patients’ varying drug reactions. Focusing on treating existing patients and maximizing the utilization of existing drugs, it is expected to not have deficiencies of traditional precision medicine for BC, including low match rate and poor therapeutic efficacy arising from tumor heterogeneity of BC.
Collapse
|
9
|
Canadas A, Santos M, Medeiros R, Dias-Pereira P. Influence of E-cadherin genetic variation in canine mammary tumour risk, clinicopathological features and prognosis. Vet Comp Oncol 2019; 17:489-496. [PMID: 31100200 DOI: 10.1111/vco.12510] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022]
Abstract
E-cadherin is a cell adhesion molecule that participates in several cellular processes that guarantee the maintenance of structural and functional integrity of epithelial tissues. E-cadherin plays an important role in mammary carcinogenesis, and various studies have demonstrated the effect of CDH1 genetic variation in risk, progression and biological behaviour of human breast cancer. Although there are some recognized genetic variations in canine CDH1 gene, their influence in canine mammary tumour development and progression has not been previously evaluated. In this study, we aim to assess the influence of CDH1 SNPs rs850805755, rs852280880 and rs852639930 in the risk, clinicopathological features and clinical outcome of canine mammary tumours. A case-control study was conducted involving 206 bitches with mammary tumours and 161 bitches free of mammary neoplasia. CDH1 SNPs rs850805755 and rs852280880 were associated with a decreased risk and a later onset of mammary tumour development. Furthermore, these SNPs were related to the development of small size carcinomas, of low histological grade and low nuclear pleomorphism. SNP rs852639930 was associated with the development of small size tumours with a non-infiltrative, non-invasive growth pattern. Data from the present investigation demonstrate that these CDH1 genetic variants could have a protective role in canine mammary tumours, by being associated with low risk of tumour development, delayed onset of the disease and less aggressive clinicopathological features.
Collapse
Affiliation(s)
- Ana Canadas
- Department of Pathology and Molecular Immunology, Instituto Ciências Biomédicas Abel Salazar, ICBAS - UPorto, University of Porto, Porto, Portugal
| | - Marta Santos
- Department of Microscopy, Instituto Ciências Biomédicas Abel Salazar, ICBAS - UPorto, University of Porto, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Centre, Portuguese Institute of Oncology of Porto, (IPO-Porto), Porto, Portugal.,CEBIMED, Biomedical Research Centre, Faculty of Health Sciences of Fernando Pessoa University, Porto, Portugal.,FMUP, Faculty of Medicine of Porto, University of Porto, Porto, Portugal.,LPCC, Research Department-Portuguese League Against Cancer (LPCC-NRNorte), Porto, Portugal
| | - Patrícia Dias-Pereira
- Department of Pathology and Molecular Immunology, Instituto Ciências Biomédicas Abel Salazar, ICBAS - UPorto, University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Chen L, Li Q, Zheng Z, Xie J, Lin X, Jiang C, Xu H, Wu X, Wu J, Zhang H. Design and optimize N-substituted EF24 as effective and low toxicity NF-κB inhibitor for lung cancer therapy via apoptosis-to-pyroptosis switch. Chem Biol Drug Des 2019; 94:1368-1377. [PMID: 30873716 DOI: 10.1111/cbdd.13514] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 11/02/2018] [Accepted: 11/24/2018] [Indexed: 12/14/2022]
Abstract
As NF-κB signaling pathway is constitutively activated in lung cancer, targeting NF-κB has a potential for the treatment. EF24 has been proved to be a NF-κB inhibitor with good antitumor activity, while whose toxicity possibly became one of the obstacles to enter into clinical application. In order to find high efficiency and low toxicity NF-κB inhibitors, EF24 was modified and 13d was screened out. It was proved that 13d possessed an effective combination of inhibiting NF-κB pathway and showing lower cytotoxicity on normal cells as well as less toxicity in acute toxicity experiment compared with the lead compound of EF24. In addition, 13d was found to inhibit cell vitality, arrest cell cycle in G2/M phase, promote cell apoptosis, and suppress the xenograft tumor growth. Furthermore, 13d was elucidated to induce pyroptosis developing from apoptosis, which was associated with the inhibition of NF-κB. Taken together, it was suggested that 13d was a potent antitumor agent.
Collapse
Affiliation(s)
- Liping Chen
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qian Li
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiwei Zheng
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingwen Xie
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoming Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Whenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengxi Jiang
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haineng Xu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoping Wu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Jianzhang Wu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huajie Zhang
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
Canadas A, Santos M, Pinto R, Medeiros R, Dias-Pereira P. Catechol-o-methyltransferase genotypes are associated with progression and biological behaviour of canine mammary tumours. Vet Comp Oncol 2018; 16:664-669. [DOI: 10.1111/vco.12438] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Ana Canadas
- Department of Pathology and Molecular Immunology, Instituto Ciências Biomédicas Abel Salazar, ICBAS-UPorto; University of Porto; Porto Portugal
| | - Marta Santos
- Department of Microscopy, Instituto Ciências Biomédicas Abel Salazar, ICBAS-UPorto; University of Porto; Porto Portugal
| | - Ricardo Pinto
- Molecular Oncology and Viral Pathology Group; IPO-Porto Research Centre, Portuguese Institute of Oncology of Porto (IPO-Porto); Porto Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group; IPO-Porto Research Centre, Portuguese Institute of Oncology of Porto (IPO-Porto); Porto Portugal
- Faculty of Medicine; University of Porto; Porto Portugal
- FP-ENAS Research Unit, UFP Energy, Environment and Health Research Unit, CEBIMED, Biomedical Research Centre; University Fernando Pessoa; Porto Portugal
- LPCC, Research Department; Portuguese League Against Cancer (LPPC-NRN); Porto Portugal
| | - Patrícia Dias-Pereira
- Department of Pathology and Molecular Immunology, Instituto Ciências Biomédicas Abel Salazar, ICBAS-UPorto; University of Porto; Porto Portugal
| |
Collapse
|
12
|
Vougiouklakis T, Saloura V, Park JH, Takamatsu N, Miyamoto T, Nakamura Y, Matsuo Y. Development of novel SUV39H2 inhibitors that exhibit growth suppressive effects in mouse xenograft models and regulate the phosphorylation of H2AX. Oncotarget 2018; 9:31820-31831. [PMID: 30159125 PMCID: PMC6112750 DOI: 10.18632/oncotarget.25806] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 06/01/2018] [Indexed: 01/21/2023] Open
Abstract
Protein methyltransferase SUV39H2 was reported to methylate histone H2AX at lysine 134 and enhance the formation of phosphorylated H2AX (γ-H2AX), which causes chemoresistance of cancer cells. We found that a series of imidazo[1,2-a]pyridine compounds that we synthesized could inhibit SUV39H2 methyltransferase activity. One of the potent compounds, OTS193320, was further analyzed in in vitro studies. The compound decreased global histone H3 lysine 9 tri-methylation levels in breast cancer cells and triggered apoptotic cell death. Combination of OTS193320 with doxorubicin (DOX) resulted in reduction of γ-H2AX levels as well as cancer cell viability compared to a single agent OTS193320 or DOX. Further optimization of inhibitors and their in vivo analysis identified a compound, OTS186935, which revealed significant inhibition of tumor growth in mouse xenograft models using MDA-MB-231 breast cancer cells and A549 lung cancer cells without any detectable toxicity. Our results suggest that the SUV39H2 inhibitors sensitize cancer cells to DOX by reduction of γ-H2AX levels in cancer cells, and collectively demonstrate that SUV39H2 inhibition warrants further investigation as a novel anti-cancer therapy.
Collapse
Affiliation(s)
- Theodore Vougiouklakis
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Vassiliki Saloura
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | | | - Yusuke Nakamura
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Yo Matsuo
- OncoTherapy Science Inc., Kawasaki, Japan
| |
Collapse
|
13
|
|
14
|
Abstract
Breast cancer is the most common cancer among women and represents one of the top five leading causes of cancer-related mortality. Inherited and acquired genetic mutations as well as epigenetic aberrations are known to be important contributors to the development and progression of breast cancer. Recent developments in high-throughput technologies have increased our understanding of the molecular changes in breast cancer, leading to the identification of distinctive genetic and epigenetic modifications in different breast cancer molecular subtypes. These genetic and epigenetic changes in luminal A, luminal B, ERBB2/HER2-enriched, basal-like, and normal-like breast cancer subtypes are discussed in this chapter. Furthermore, recent epigenome studies provided more information about further stratification of breast cancer subtypes, with essential role in the appropriate diagnosis and treatment of breast cancer. Thus, the inclusion of both genetic and epigenetic information in breast cancer clinical care could provide critical scientific base for precision medicine in breast cancer.
Collapse
|
15
|
Ziegelstein RC. Personomics: The Missing Link in the Evolution from Precision Medicine to Personalized Medicine. J Pers Med 2017; 7:jpm7040011. [PMID: 29035320 PMCID: PMC5748623 DOI: 10.3390/jpm7040011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/23/2017] [Accepted: 10/09/2017] [Indexed: 11/16/2022] Open
Abstract
Clinical practice guidelines have been developed for many common conditions based on data from randomized controlled trials. When medicine is informed solely by clinical practice guidelines, however, the patient is not treated as an individual, but rather as member of a group. Precision medicine, as defined herein, characterizes unique biological characteristics of the individual or of specimens obtained from an individual to tailor diagnostics and therapeutics to a specific patient. These unique biological characteristics are defined by the tools of precision medicine: genomics, proteomics, metabolomics, epigenomics, pharmacogenomics, and other “-omics.” Personalized medicine, as defined herein, uses additional information about the individual derived from knowing the patient as a person. These unique personal characteristics are defined by the tools of personalized medicine—personomics—which take into account an individual’s personality, preferences, values, goals, health beliefs, social support network, financial resources, and unique life circumstances that affect how and when a given health condition will manifest in that person and how that condition will respond to treatment. In this paradigm, precision medicine may be considered a necessary step in the evolution of medical care to personalized medicine, with personomics as the missing link.
Collapse
Affiliation(s)
- Roy C Ziegelstein
- Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| |
Collapse
|
16
|
Groner B, von Manstein V. Jak Stat signaling and cancer: Opportunities, benefits and side effects of targeted inhibition. Mol Cell Endocrinol 2017; 451:1-14. [PMID: 28576744 DOI: 10.1016/j.mce.2017.05.033] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 05/27/2017] [Indexed: 02/06/2023]
Abstract
The effects of Jak Stat signaling and the persistent activation of Stat3 and Stat5 on tumor cell survival, proliferation and invasion have made the Jak Stat pathway a favorite target for drug development and cancer therapy. This notion was strengthened when additional biological functions of Stat signaling in cancer and their roles in the regulation of cytokine dependent inflammation and immunity in the tumor microenvironment were discovered. Stats act not only as transcriptional inducers, but affect gene expression via epigenetic modifications, induce epithelial mesenchymal transition, generate a pro-tumorigenic microenvironment, promote cancer stem cell self-renewal and differentiation, and help to establish the pre-metastatic niche formation. The effects of Jak Stat inhibition on the suppression of pro-inflammatory responses appears most promising and could become a strategy in the prevention of tumor progression. The direct and mediated mechanisms of Jak Stat signaling in and on tumors cells, the interactions with other signaling pathways and transcription factors and the targeting of the functionally crucial secondary modifications of Stat molecules suggest novel approaches to the future development of Jak Stat based cancer therapeutics.
Collapse
Affiliation(s)
- Bernd Groner
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Paul Ehrlich Str. 42, D-60596 Frankfurt am Main, Germany.
| | - Viktoria von Manstein
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Paul Ehrlich Str. 42, D-60596 Frankfurt am Main, Germany
| |
Collapse
|
17
|
Parsons BL, McKim KL, Myers MB. Variation in organ-specific PIK3CA and KRAS mutant levels in normal human tissues correlates with mutation prevalence in corresponding carcinomas. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:466-476. [PMID: 28755461 PMCID: PMC5601221 DOI: 10.1002/em.22110] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 05/27/2023]
Abstract
Large-scale sequencing efforts have described the mutational complexity of individual cancers and identified mutations prevalent in different cancers. As a complementary approach, allele-specific competitive blocker PCR (ACB-PCR) is being used to quantify levels of hotspot cancer driver mutations (CDMs) with high sensitivity, to elucidate the tissue-specific properties of CDMs, their occurrence as tumor cell subpopulations, and their occurrence in normal tissues. Here we report measurements of PIK3CA H1047R mutant fraction (MF) in normal colonic mucosa, normal lung, colonic adenomas, colonic adenocarcinomas, and lung adenocarcinomas. We report PIK3CA E545K MF measurements in those tissues, as well as in normal breast, normal thyroid, mammary ductal carcinomas, and papillary thyroid carcinomas. We report KRAS G12D and G12V MF measurements in normal colon. These MF measurements were integrated with previously published ACB-PCR data on KRAS G12D, KRAS G12V, and PIK3CA H1047R. Analysis of these data revealed a correlation between the degree of interindividual variability in these mutations (as log10 MF standard deviation) in normal tissues and the frequencies with which the mutations are detected in carcinomas of the corresponding organs in the COSMIC database. This novel observation has important implications. It suggests that interindividual variability in mutation levels of normal tissues may be used as a metric to identify mutations with critical early roles in tissue-specific carcinogenesis. Additionally, it raises the possibility that personalized cancer therapeutics, developed to target specifically activated oncogenic products, might be repurposed as prophylactic therapies to reduce the accumulation of cells carrying CDMs and, thereby, reduce future cancer risk. Environ. Mol. Mutagen. 58:466-476, 2017. © 2017 This article is a U.S. Government work and is in the public domain in the USA. Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.
Collapse
Affiliation(s)
- Barbara L. Parsons
- Division of Genetic and Molecular ToxicologyU.S. Food and Drug Administration, National Center for Toxicological ResearchJeffersonArkansas
| | - Karen L. McKim
- Division of Genetic and Molecular ToxicologyU.S. Food and Drug Administration, National Center for Toxicological ResearchJeffersonArkansas
| | - Meagan B. Myers
- Division of Genetic and Molecular ToxicologyU.S. Food and Drug Administration, National Center for Toxicological ResearchJeffersonArkansas
| |
Collapse
|
18
|
Martínez-Campa C, Menéndez-Menéndez J, Alonso-González C, González A, Álvarez-García V, Cos S. What is known about melatonin, chemotherapy and altered gene expression in breast cancer. Oncol Lett 2017; 13:2003-2014. [PMID: 28454355 PMCID: PMC5403278 DOI: 10.3892/ol.2017.5712] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
Melatonin, synthesized in and released from the pineal gland, has been demonstrated by multiple in vivo and in vitro studies to have an oncostatic role in hormone-dependent tumors. Furthermore, several clinical trials point to melatonin as a promising adjuvant molecule to be considered for cancer treatment. In the past few years, evidence of a broader spectrum of action of melatonin as an antitumor agent has arisen; thus, melatonin appears to also have therapeutic effects in several types of hormone-independent cancer, including ovarian, leukemic, pancreatic, gastric and non-small cell lung carcinoma. In the present study, the latest findings regarding melatonin molecular actions when concomitantly administered with either radiotherapy or chemotherapy in cancer were reviewed, with a particular focus on hormone-dependent breast cancer. Finally, the present study discusses which direction should be followed in the next years to definitely clarify whether or not melatonin administration could protect against non-desirable effects (such as altered gene expression and post-translational protein modifications) caused by chemotherapy or radiotherapy treatments. As treatments move towards personalized medicine, comparative gene expression profiling with and without melatonin may be a powerful tool to better understand the antitumor effects of melatonin, the pineal gland hormone.
Collapse
Affiliation(s)
- Carlos Martínez-Campa
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Javier Menéndez-Menéndez
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Carolina Alonso-González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Alicia González
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| | - Virginia Álvarez-García
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot Watt University, EH14 4AS Edinburgh, UK
| | - Samuel Cos
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria and Research Institute Valdecilla, 39011 Santander, Spain
| |
Collapse
|
19
|
Colijn C, Jones N, Johnston IG, Yaliraki S, Barahona M. Toward Precision Healthcare: Context and Mathematical Challenges. Front Physiol 2017; 8:136. [PMID: 28377724 PMCID: PMC5359292 DOI: 10.3389/fphys.2017.00136] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/22/2017] [Indexed: 12/12/2022] Open
Abstract
Precision medicine refers to the idea of delivering the right treatment to the right patient at the right time, usually with a focus on a data-centered approach to this task. In this perspective piece, we use the term "precision healthcare" to describe the development of precision approaches that bridge from the individual to the population, taking advantage of individual-level data, but also taking the social context into account. These problems give rise to a broad spectrum of technical, scientific, policy, ethical and social challenges, and new mathematical techniques will be required to meet them. To ensure that the science underpinning "precision" is robust, interpretable and well-suited to meet the policy, ethical and social questions that such approaches raise, the mathematical methods for data analysis should be transparent, robust, and able to adapt to errors and uncertainties. In particular, precision methodologies should capture the complexity of data, yet produce tractable descriptions at the relevant resolution while preserving intelligibility and traceability, so that they can be used by practitioners to aid decision-making. Through several case studies in this domain of precision healthcare, we argue that this vision requires the development of new mathematical frameworks, both in modeling and in data analysis and interpretation.
Collapse
Affiliation(s)
- Caroline Colijn
- Department of Mathematics, Imperial College LondonLondon, UK
- EPSRC Centre for Mathematics of Precision Healthcare, Imperial College LondonLondon, UK
| | - Nick Jones
- Department of Mathematics, Imperial College LondonLondon, UK
- EPSRC Centre for Mathematics of Precision Healthcare, Imperial College LondonLondon, UK
| | - Iain G. Johnston
- EPSRC Centre for Mathematics of Precision Healthcare, Imperial College LondonLondon, UK
- School of Biosciences, University of BirminghamBirmingham, UK
| | - Sophia Yaliraki
- EPSRC Centre for Mathematics of Precision Healthcare, Imperial College LondonLondon, UK
- Department of Chemistry, Imperial College LondonLondon, UK
| | - Mauricio Barahona
- Department of Mathematics, Imperial College LondonLondon, UK
- EPSRC Centre for Mathematics of Precision Healthcare, Imperial College LondonLondon, UK
| |
Collapse
|
20
|
Ross JS, Gay LM. Comprehensive genomic sequencing and the molecular profiles of clinically advanced breast cancer. Pathology 2017; 49:120-132. [DOI: 10.1016/j.pathol.2016.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 02/06/2023]
|
21
|
Ahmad A, Mondal SK, Jan BL, Mukhopadhyay D, Banerjee R, Alkharfy KM. Quantification of lipid modified estrogenic derivative (ESC8) in rat plasma by LC-MS: application to a pharmacokinetic study. Biomed Chromatogr 2016; 30:2024-2030. [PMID: 27306429 DOI: 10.1002/bmc.3780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/05/2016] [Accepted: 06/13/2016] [Indexed: 12/19/2022]
Abstract
A lipid-conjugated, estrogenic derivative molecule, ESC8, compared with other estrogenic molecules, encourages cell death in both ER-positive and ER-negative breast cancer cells. A rapid and highly sensitive assay method has been developed and validated for the estimation of a ESC8 in rat plasma using liquid chromatography coupled with mass spectrometry under positive-ion mode with electrospray ionization. The sample process includes using methanol for precipitation of ESC8 and dextromethorphan (internal standard, IS) from plasma. Chromatographic separation was achieved with methanol-water-formic acid (70:30:0.1% v/v/v) pumped at a flow rate of 0.3mL/min and a C18 column (50 × 2.1 mm i.d., 1.7 μm particle size) with a total run time of 5 min. The m/z ions monitored were 568.5 and 272.1 for ESC8 and IS, respectively. The lower limit of quantitation achieved was 1.08 ng/mL and linearity was observed from 5 to 500 ng/mL. The intra- and inter-day precisions were <4%. The proposed method was successfully applied to a preliminary pharmacokinetic study of ESC8 liposomal formulation following an intraperitoneal administration of 3.67 mg/kg in rats. The concentrations of ESC8 in plasma were quantifiable up to 36 h. The peak concentration of ESC8 was found to be 110.72 ng/mL, the area under the concentration-time curve was 1625.23ng/mL h and the half-life was 11.72 h.
Collapse
Affiliation(s)
- Ajaz Ahmad
- Department of Clinical Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sujan Kumar Mondal
- Department of Clinical Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Basit Latief Jan
- Department of Clinical Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Rajkumar Banerjee
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Khalid M Alkharfy
- Department of Clinical Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Halfter K, Hoffmann O, Ditsch N, Ahne M, Arnold F, Paepke S, Grab D, Bauerfeind I, Mayer B. Testing chemotherapy efficacy in HER2 negative breast cancer using patient-derived spheroids. J Transl Med 2016; 14:112. [PMID: 27142386 PMCID: PMC4855689 DOI: 10.1186/s12967-016-0855-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/06/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Targeted anti-HER2 therapy has greatly improved the prognosis for many breast cancer patients. However, treatment for HER2 negative disease is currently still selected from a multitude of untargeted chemotherapeutic treatment options. A predictive test was developed using patient-derived spheroids to identify the most effective therapy for patients with HER2 negative breast cancer of all stages, for clinically relevant subgroups, as well as individual patients. METHODS Tumor samples from 120 HER2 negative patients obtained through biopsy or surgical excision were tested in the breast cancer spheroid model using scaffold-free cell culture. Similarly, spheroids were also generated from established HER2 negative breast cancer cell lines T-47D, MCF7, HCC1143, and HCC1937 to compare treatment efficacy of heterogeneous cell populations from patient tumor tissue with homogeneous cell lines. Spheroids were treated in vitro with guideline-recommended compounds. Treatment mediated impact on cell survival was subsequently quantified using an ATP assay. RESULTS Differences were observed in the metabolic activity of the untreated spheroids, whereby cell lines consistently achieved higher values compared to tissue spheroids (p < 0.001). A higher number of cells per spheroid correlated with a higher basal metabolic activity in tissue-derived spheroids (p < 0.01), while the opposite was observed for cell line spheroids (p < 0.01). Recurrent tumors showed a higher mean vitality (p < 0.01) compared to primary tumors. Except for taxanes, treatment efficacy for most tested compounds differed significantly between breast cancer tissue spheroids and breast cancer cell lines. Overall a high variability in treatment response in vitro was seen in the tissue spheroids regardless of the tested substances. A greater response to anthracycline/docetaxel was observed for hormone receptor negative samples (p < 0.01). A higher response to 5-FU (p < 0.01) and anthracycline (p < 0.05) was seen in high grade tumors. Smaller tumor size and negative lymph node status were both associated with a higher treatment efficacy to anthracycline treatment combined with 5-FU (cT1/2 vs cT3/4, p = 0.035, cN+ vs cN-, p < 0.05). CONCLUSIONS The tissue spheroid model reflects current guideline treatment recommendations for HER2 negative breast cancer, whereas tested cell lines did not. This model represents a unique diagnostic method to select the most effective therapy out of several equivalent treatment options.
Collapse
Affiliation(s)
- Kathrin Halfter
- />SpheroTec GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
| | - Oliver Hoffmann
- />SpheroTec GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
| | - Nina Ditsch
- />Department of Obstetrics and Gynecology, Hospital of the University of Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Mareike Ahne
- />SpheroTec GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
| | - Frank Arnold
- />Department of General, Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Stefan Paepke
- />Department of Gynecology and Obstetrics, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Dieter Grab
- />Klinikum Harlaching, Sanatoriumsplatz 2, 81545 Munich, Germany
| | - Ingo Bauerfeind
- />Klinikum Landshut, Robert-Koch-Str. 1, 8434 Landshut, Germany
| | - Barbara Mayer
- />SpheroTec GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
- />Department of General, Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| |
Collapse
|
23
|
Zeng T, Zhang W, Yu X, Liu X, Li M, Chen L. Big-data-based edge biomarkers: study on dynamical drug sensitivity and resistance in individuals. Brief Bioinform 2015; 17:576-92. [DOI: 10.1093/bib/bbv078] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Indexed: 12/21/2022] Open
|
24
|
Stover DG, Winer EP. Tailoring adjuvant chemotherapy regimens for patients with triple negative breast cancer. Breast 2015; 24 Suppl 2:S132-5. [PMID: 26255198 DOI: 10.1016/j.breast.2015.07.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cytotoxic chemotherapy remains the systemic therapy of choice for triple-negative breast cancer (TNBC), a poor-prognosis subtype of breast cancer. Growing data focusing on TNBC provides an opportunity to assess if we can tailor adjuvant chemotherapy based on patient and tumor characteristics. The standard of care for moderate-to-high risk TNBC remains a sequential anthracycline-taxane combination, with the potential for shorter and less toxic regimens in stage I disease. Platinums are promising in the neoadjuvant and metastatic settings but we await long-term outcome data before incorporation into standard regimens in the adjuvant setting. Specific subgroups within TNBC, such as BRCA mutation carriers, require special attention, and the role of platinums in these patients warrants further consideration. There is hope that in the future, further subdividing TNBC by gene expression profile, mutation, immune infiltrate, and others will reveal novel susceptibilities.
Collapse
Affiliation(s)
- Daniel G Stover
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Eric P Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|