1
|
Papalexis P, Georgakopoulou VE, Drossos PV, Thymara E, Nonni A, Lazaris AC, Zografos GC, Spandidos DA, Kavantzas N, Thomopoulou GE. Precision medicine in breast cancer (Review). Mol Clin Oncol 2024; 21:78. [PMID: 39246849 PMCID: PMC11375768 DOI: 10.3892/mco.2024.2776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Precision medicine in breast cancer is a revolutionary approach that customizes diagnosis and treatment based on individual and tumor characteristics, departing from the traditional one-size-fits-all approach. Breast cancer is diverse, with various subtypes driven by distinct genetic mutations. Understanding this diversity is crucial for tailored treatment strategies that target specific vulnerabilities in each tumor. Genetic testing, particularly for mutations in breast cancer gene (BRCA) DNA repair-associated genes, helps assess hereditary risks and influences treatment decisions. Molecular subtyping guides personalized treatments, such as hormonal therapies for receptor-positive tumors and human epidermal growth factor receptor 2 (HER2)-targeted treatments. Targeted therapies, including those for HER2-positive and hormone receptor-positive breast cancers, offer more effective and precise interventions. Immunotherapy, especially checkpoint inhibitors, shows promise, particularly in certain subtypes such as triple-negative breast cancer, with ongoing research aiming to broaden its effectiveness. Integration of big data and artificial intelligence enhances personalized treatment strategies, while liquid biopsies provide real-time insights into tumor dynamics, aiding in treatment monitoring and modification. Challenges persist, including accessibility and tumor complexity, but emerging technologies and precision prevention offer hope for improved outcomes. Ultimately, precision medicine aims to optimize treatment efficacy, minimize adverse effects and enhance the quality of life for patients with breast cancer.
Collapse
Affiliation(s)
- Petros Papalexis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | | | - Panagiotis V Drossos
- Department of Biomedical Sciences, University of West Attica, 12243 Athens, Greece
| | - Eirini Thymara
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Aphrodite Nonni
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Andreas C Lazaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George C Zografos
- Department of Propedeutic Surgery, Hippokration Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Nikolaos Kavantzas
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Georgia Eleni Thomopoulou
- Cytopathology Department, 'Attikon' University General Hospital, School of Medicine, National and Kapodistrian University of Athens, 12461 Athens, Greece
| |
Collapse
|
2
|
Singh A, Singh K, Kaur K, Singh A, Sharma A, Kaur K, Kaur J, Kaur G, Kaur U, Kaur H, Singh P, Bedi PMS. Coumarin as an Elite Scaffold in Anti-Breast Cancer Drug Development: Design Strategies, Mechanistic Insights, and Structure-Activity Relationships. Biomedicines 2024; 12:1192. [PMID: 38927399 PMCID: PMC11200728 DOI: 10.3390/biomedicines12061192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer is the most common cancer among women. Currently, it poses a significant threat to the healthcare system due to the emerging resistance and toxicity of available drug candidates in clinical practice, thus generating an urgent need for the development of new potent and safer anti-breast cancer drug candidates. Coumarin (chromone-2-one) is an elite ring system widely distributed among natural products and possesses a broad range of pharmacological properties. The unique distribution and pharmacological efficacy of coumarins attract natural product hunters, resulting in the identification of numerous natural coumarins from different natural sources in the last three decades, especially those with anti-breast cancer properties. Inspired by this, numerous synthetic derivatives based on coumarins have been developed by medicinal chemists all around the globe, showing promising anti-breast cancer efficacy. This review is primarily focused on the development of coumarin-inspired anti-breast cancer agents in the last three decades, especially highlighting design strategies, mechanistic insights, and their structure-activity relationship. Natural coumarins having anti-breast cancer efficacy are also briefly highlighted. This review will act as a guideline for researchers and medicinal chemists in designing optimum coumarin-based potent and safer anti-breast cancer agents.
Collapse
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
| | | | - Amandeep Singh
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center, 500 University Drive, Hershey, PA 17033, USA;
| | - Aman Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
| | - Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
| | - Jaskirat Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
| | - Gurleen Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
| | - Uttam Kaur
- University School of Business Management, Chandigarh University, Gharuan 140413, Mohali, India;
| | - Harsimran Kaur
- Department of Pharmaceutical Chemistry, Khalsa College of Pharmacy, Amritsar 143005, Punjab, India; (H.K.); (P.S.)
| | - Prabhsimran Singh
- Department of Pharmaceutical Chemistry, Khalsa College of Pharmacy, Amritsar 143005, Punjab, India; (H.K.); (P.S.)
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India; (K.S.); (A.S.); (K.K.); (J.K.); (G.K.)
- Drug and Pollution Testing Laboratory, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| |
Collapse
|
3
|
Bahrin NWS, Matusin SNI, Mustapa A, Huat LZ, Perera S, Hamid MRWHA. Exploring the effectiveness of molecular subtypes, biomarkers, and genetic variations as first-line treatment predictors in Asian breast cancer patients: a systematic review and meta-analysis. Syst Rev 2024; 13:100. [PMID: 38576013 PMCID: PMC10993489 DOI: 10.1186/s13643-024-02520-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/23/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Breast cancer incidence has been on the rise significantly in the Asian population, occurring at an earlier age and a later stage. The potential predictive value of molecular subtypes, biomarkers, and genetic variations has not been deeply explored in the Asian population. This study evaluated the effect of molecular subtype classification and the presence or absence of biomarkers and genetic variations on pathological complete response (pCR) after neoadjuvant treatment in Asian breast cancer patients. METHODS A systematic search was conducted in MEDLINE (PubMed), Science Direct, Scopus, and Cochrane Library databases. Studies were selected if they included Asian breast cancer patients treated with neoadjuvant chemotherapy and contained data for qualitative or quantitative analyses. The quality of the included studies was assessed using the Newcastle Ottawa Scale. Following the random effects model, pooled odds ratios or hazard ratios with 95% confidence intervals for pCR were analysed using Review Manager Software. Heterogeneity between studies was assessed using Cochran's Q-test and I2 test statistics. RESULTS In total, 19,708 Asian breast cancer patients were pooled from 101 studies. In the neoadjuvant setting, taxane-anthracycline (TA) chemotherapy showed better pCR outcomes in triple-negative breast cancer (TNBC) (p<0.0001) and human epidermal growth factor receptor 2 enriched (HER2E) (p<0.0001) than luminal breast cancer patients. Similarly, taxane-platinum (TP) chemotherapy also showed better pCR outcomes in TNBC (p<0.0001) and HER2E (p<0.0001). Oestrogen receptor (ER)-negative, progesterone receptor (PR)-negative, HER2-positive and high Ki-67 were significantly associated with better pCR outcomes when treated with either TA or TP. Asian breast cancer patients harbouring wildtype PIK3CA were significantly associated with better pCR outcomes when treated with TA in the neoadjuvant setting (p=0.001). CONCLUSIONS In the neoadjuvant setting, molecular subtypes (HER2E and TNBC), biomarkers (ER, PR, HER2, HR, Ki-67, nm23-H1, CK5/6, and Tau), and gene (PIK3CA) are associated with increased pCR rates in Asian breast cancer patients. Hence, they could be further explored for their possible role in first-line treatment response, which can be utilised to treat breast cancer more efficiently in the Asian population. However, it needs to be further validated with additional powered studies. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42021246295.
Collapse
Affiliation(s)
- Nurul Wafiqah Saipol Bahrin
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB) Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Negara Brunei Darussalam
| | - Siti Nur Idayu Matusin
- Halalan Thayyiban Research Centre, Universiti Islam Sultan Sharif Ali, Jalan Tutong, Sinaut, TB1741, Negara Brunei Darussalam
| | - Aklimah Mustapa
- Halalan Thayyiban Research Centre, Universiti Islam Sultan Sharif Ali, Jalan Tutong, Sinaut, TB1741, Negara Brunei Darussalam
| | - Lu Zen Huat
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB) Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Negara Brunei Darussalam
| | - Sriyani Perera
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Mas Rina Wati Haji Abdul Hamid
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB) Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Negara Brunei Darussalam.
| |
Collapse
|
4
|
Zhang H, Hussin H, Hoh CC, Cheong SH, Lee WK, Yahaya BH. Big data in breast cancer: Towards precision treatment. Digit Health 2024; 10:20552076241293695. [PMID: 39502482 PMCID: PMC11536614 DOI: 10.1177/20552076241293695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Breast cancer is the most prevalent and deadliest cancer among women globally, representing a major threat to public health. In response, the World Health Organization has established the Global Breast Cancer Initiative framework to reduce breast cancer mortality through global collaboration. The integration of big data analytics (BDA) and precision medicine has transformed our understanding of breast cancer's biological traits and treatment responses. By harnessing large-scale datasets - encompassing genetic, clinical, and environmental data - BDA has enhanced strategies for breast cancer prevention, diagnosis, and treatment, driving the advancement of precision oncology and personalised care. Despite the increasing importance of big data in breast cancer research, comprehensive studies remain sparse, underscoring the need for more systematic investigation. This review evaluates the contributions of big data to breast cancer precision medicine while addressing the associated opportunities and challenges. Through the application of big data, we aim to deepen insights into breast cancer pathogenesis, optimise therapeutic approaches, improve patient outcomes, and ultimately contribute to better survival rates and quality of life. This review seeks to provide a foundation for future research in breast cancer prevention, treatment, and management.
Collapse
Affiliation(s)
- Hao Zhang
- Breast Cancer Translational Research Program (BCTRP@IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
- Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Hasmah Hussin
- Breast Cancer Translational Research Program (BCTRP@IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
- Department of Clinical Medicine, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | | | | | - Wei-Kang Lee
- Codon Genomics Sdn Bhd, Seri Kembangan, Selangor, Malaysia
| | - Badrul Hisham Yahaya
- Breast Cancer Translational Research Program (BCTRP@IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
- Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| |
Collapse
|
5
|
Alsamri H, Al Dhaheri Y, Iratni R. Targeting Triple-Negative Breast Cancer by the Phytopolyphenol Carnosol: ROS-Dependent Mechanisms. Antioxidants (Basel) 2023; 12:1349. [PMID: 37507889 PMCID: PMC10376170 DOI: 10.3390/antiox12071349] [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: 04/24/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC), which lacks the expression of the three hormone receptors (i.e., estrogen receptor, progesterone receptor, and human epidermal growth factor receptor), is characterized by a high proliferative index, high invasiveness, poor prognosis, early relapse, and a tendency to be present in advanced stages. These characteristics rank TNBC among the most aggressive and lethal forms of breast cancer. The lack of the three receptors renders conventional hormonal therapy ineffective against TNBC. Moreover, there are no clinically approved therapies that specifically target TNBC, and the currently used chemotherapeutic agents, such as cisplatin, taxanes, and other platinum compounds, have a limited clinical effect and develop chemoresistance over time. Phytochemicals have shown efficacy against several types of cancer, including TNBC, by targeting several pathways involved in cancer development and progression. In this review, we focus on one phytochemical carnosol, a natural polyphenolic terpenoid with strong anti-TNBC effects and its ROS-dependent molecular mechanisms of action. We discuss how carnosol targets key pathways and proteins regulating the cell cycle, growth, epigenetic regulators, invasion, and metastasis of TNBC. This review identifies carnosol as a potential novel targeting protein degradation molecule.
Collapse
Affiliation(s)
- Halima Alsamri
- General Requirement Department, Fatima College of Health Sciences, Al Ain P.O. Box 24162, United Arab Emirates
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
6
|
Kuoch H, Krotova K, Graham ML, Brantly ML, Aslanidi G. Multiplexing AAV Serotype-Specific Neutralizing Antibodies in Preclinical Animal Models and Humans. Biomedicines 2023; 11:biomedicines11020523. [PMID: 36831059 PMCID: PMC9953293 DOI: 10.3390/biomedicines11020523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
The accurate assessment of AAV-specific pre-existing humoral immunity due to natural viral infection is critical for the efficient use of clinical gene therapy. The method described in the present study applies equivalent infection conditions to each AAV serotype (AAV1, AAV2, AAV3, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, and AAVAnc80L65). In the current study, we validated the assay by assessing AAV-neutralizing antibody titers in a limited cohort of random human donors and well-established preclinical large animal models, including dogs and non-human primates (NHPs). We achieved a rapid and accurate evaluation of neutralizing titers for each individual subject that can be used for clinical enrollment based on specific AAV serotypes and individualized selection of the most suitable AAV serotype for each specific patient.
Collapse
Affiliation(s)
- Hisae Kuoch
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Karina Krotova
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Melanie L. Graham
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, MN 55108, USA
| | - Mark L. Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Medical School, University of Florida, Gainesville, FL 32610, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
- Correspondence: ; Tel.: +1-507-437-9622; Fax: +1-507-437-9606
| |
Collapse
|
7
|
Ray SK, Mukherjee S. Starring Role of Biomarkers and Anticancer Agents as a Major Driver in Precision Medicine of Cancer Therapy. Curr Mol Med 2023; 23:111-126. [PMID: 34939542 DOI: 10.2174/1566524022666211221152947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 12/16/2022]
Abstract
Precision medicine is the most modern contemporary medicine approach today, based on great amount of data on people's health, individual characteristics, and life circumstances, and employs the most effective ways to prevent and cure diseases. Precision medicine in cancer is the most precise and viable treatment for every cancer patient based on the disease's genetic profile. Precision medicine changes the standard one size fits all medication model, which focuses on average responses to care. Consolidating modern methodologies for streamlining and checking anticancer drugs can have long-term effects on understanding the results. Precision medicine can help explicit anticancer treatments using various drugs and even in discovery, thus becoming the paradigm of future cancer medicine. Cancer biomarkers are significant in precision medicine, and findings of different biomarkers make this field more promising and challenging. Naturally, genetic instability and the collection of extra changes in malignant growth cells are ways cancer cells adapt and survive in a hostile environment, for example, one made by these treatment modalities. Precision medicine centers on recognizing the best treatment for individual patients, dependent on their malignant growth and genetic characterization. This new era of genomics progressively referred to as precision medicine, has ignited a new episode in the relationship between genomics and anticancer drug development.
Collapse
Affiliation(s)
| | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020. India
| |
Collapse
|
8
|
Does MRI have added value in ultrasound-detected BIRADS-3 breast masses in candidates for assisted reproductive therapy? Eur J Radiol Open 2022; 10:100474. [PMID: 36624818 PMCID: PMC9823155 DOI: 10.1016/j.ejro.2022.100474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022] Open
Abstract
Background Ultrasound-detected breast lesions with probably benign features are a great challenge for clinicians, especially in breasts with dense composition. We aimed to investigate the finding of two radiologic modalities on these lesions. Methods This retrospective cross-sectional study recruited patients including (1) candidates of assisted reproductive therapy (ART), (2) patients with prior high-risk lesions, and (3) the "suspected" BIRADS-3 masses referring to masses that US BIRADS-3 was not compatible with the clinical breast exam. The degree of agreement in diagnosing BIRADS-3 lesions between two modalities of magnetic resonance imaging (MRI) and ultrasonography (US), and comparison of the lesions in US and MRI were the study variables. Results A total number of 123 lesions in 67 patients with a median age of 38 (IQR: 11, range: 17-67). In the examination by MRI, 107 (87.0 %) lesions were BIRADS-3 indicating the agreement level between these two modalities. The median size of the lesions in US was 9 mm (IQR: 5, range: 3-43) and 9 mm (IQR: 10, range: 4-46) in MRI. The measured size of the lesions between the two modalities was highly correlated (Spearman correlation coefficient: 0.889, P-value < 0.001). MRI evaluation revealed two cases of deep lesions which were missed in the US imaging. Conclusions This study found relatively high agreement values between US and MRI in detecting BIRADS-3 breast lesions in candidates for ART or patients with prior high-risk lesions. Also, MRI could downgrade about one-tenth of the cases to a lower BIRADS level and resolved the need for closer follow-up.
Collapse
|
9
|
Xia W, Chen W, Fang S, Wu J, Zhang J, Yuan H. Case report: Individualized treatment of advanced breast cancer with the use of the patient-derived tumor-like cell cluster model. Front Oncol 2022; 12:897984. [DOI: 10.3389/fonc.2022.897984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is one of the most common tumors in women. Despite various treatments, the survival of patients with advanced breast cancer is still disappointing. Furthermore, finding an effective individualized treatment for different kinds of patients is a thorny problem. Patient-derived tumor-like cell clusters were reported to be used for personalized drug testing in cancer therapy and had a prediction accuracy of 93%. However, there is still a lack of case reports about its application in the individualized treatment of breast cancer patients. Here, we described four cases of individualized treatment for advanced breast cancer using the patient-derived tumor-like cell cluster model (PTC model). In these four cases, the PTC model showed a good predictive effect. The tumor size was reduced significantly or even disappeared completely through clinical, radiological, or pathological evaluation with the help of the PTC model for selecting an individualized therapy regimen. Furthermore, the drug sensitivity test results of the PTC model were consistent with pathological molecular typing and the actual clinical drug resistance of the patients. In summary, our case report first evaluated the application value of the PTC model in advanced breast cancer, and the PTC model might be used as an efficient tool for drug resistance screening and for selecting a better personalized treatment, although further study is needed to prove the validity and stability of the PTC model in drug screening.
Collapse
|
10
|
Ibrahem SQ, Ahmed HQ, Amin KM. Genetic Variations in Cytochrome P450 1A1 and 1B1 Genes in a Cohort of Patients from Iraq Diagnosed with Breast Cancer. Breast Cancer (Auckl) 2021; 15:11782234211050727. [PMID: 34671182 PMCID: PMC8521753 DOI: 10.1177/11782234211050727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 09/14/2021] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the most prevalent malignant neoplasm in females. Genetic variations in the xenobiotic metabolising cytochrome enzymes; Family 1 Subfamily A Member 1 (CYP1A1) and Family 1 Subfamily B Member 1 (CYP1B1) might play a role in the individual susceptibility to breast cancer and its prognosis. The goal of this study is to estimate the incidence of single nucleotide polymorphisms (SNPs) in CYP1A1 (rs1048943, Ile462VaI, and rs4646903/MSP1) and in CYP1B1 (rs1056836, Leu432Val) genes in patients with breast cancer. This case-control study included 180 female patients with breast cancer and 180 healthy control subjects from Kirkuk/Iraq. Genomic DNA was extracted from venous blood samples and tested for SNPs by the direct DNA sequencing technique. A statistical analysis was done to identify if there is any association between SNPs and the increasing odd of breast cancer and its stage, grade and molecular subtype at diagnosis. The common (reference) genotype of CYP1A1 gene rs1048943 is AA. The AG and GG variant genotypes were significantly more common in the breast cancer patients and conferred an increased odd of breast cancer and its later stages (stages III and IV) and poor differentiation (P < .01) but not with the molecular subtypes. The common genotype of CYP1A1 rs4646903 is TT. The variant genotypes TC and CC are not associated either with increased risk of breast cancer (P > .05) or with its stage, grade or molecular subtypes (P > .05). The GG genotype of CYP1B1 rs1056836 was the common genotype. The CG and CC variant genotypes were not associated with the increased risks of breast cancer (P > .05) or its stage, grade or molecular subtypes (P > .05). In conclusion, variants genotypes of CYP1A1 rs1048943 might play a role in breast cancer pathogenesis and prognosis and can have a place in cancer screening and tailored medicine in the future in the Iraqi population. Future larger scale studies including other genes might help to better understand the role of the SNP in breast risk and its prognosis.
Collapse
Affiliation(s)
- Salih Q Ibrahem
- Department of Biochemistry, College of Medicine, Kirkuk University, Kirkuk, Iraq
| | - Hussien Q Ahmed
- Department of Surgery, College of Medicine, Kirkuk University, Kirkuk, Iraq
| | - Khalida M Amin
- Department of Obstetrics and Gynaecology, College of Medicine, Kirkuk University, Kirkuk, Iraq
| |
Collapse
|
11
|
Molecular Prognostic Factors for Distant Metastases in Premenopausal Patients with HR+/HER2- Early Breast Cancer. J Pers Med 2021; 11:jpm11090835. [PMID: 34575612 PMCID: PMC8468490 DOI: 10.3390/jpm11090835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022] Open
Abstract
Molecular factors that drive metastasis in premenopausal patients with hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2−), early breast cancer (EBC) are largely unknown. To identify markers/signatures contributing to metastasis, we analyzed molecular changes in tumors from premenopausal patients who developed metastasis (M1) and who did not (M0). Ninety-seven premenopausal patients with HR+/HER2− EBC were included (M1, n = 48, median distant metastasis-free survival (DMFS): 54 (7–184) months; M0, n = 49, median follow-up: 149 (121–191) months). Gene expression profiling on tumor RNA (Breast Cancer 360TM panel, Nanostring) was performed, followed by comprehensive bioinformatic and statistical analyses. Significantly enhanced ROR (risk of recurrence) scores and reduced signature scores of PGR (progesterone receptor), claudin-low, and mammary stemness were determined in M1. These differences were significantly associated with shorter DMFS in univariate survival analyses. Gene set enrichment analysis showed an enriched mTORC1 pathway in M1. Moreover, a metastasis signature of 19 differentially expressed genes (DEGs) that were DMFS-related was defined. Multivariate analysis including the four signatures, 19 DEGs, pN, and pT status, identified LRP2, IBSP, and SCUBE2 as independent prognostic factors. We identified prognostic gene signatures and single-gene markers for distant metastasis in premenopausal HR+/HER2− EBC potentially applicable in future clinical practice.
Collapse
|
12
|
Trager MH, Sah B, Chen Z, Liu L. Control of Breast Cancer Pathogenesis by Histone Methylation and the Hairless Histone Demethylase. Endocrinology 2021; 162:6259332. [PMID: 33928351 PMCID: PMC8237996 DOI: 10.1210/endocr/bqab088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Indexed: 12/19/2022]
Abstract
Breast cancer is a highly heterogeneous disease, encompassing many subtypes that have distinct origins, behaviors, and prognoses. Although traditionally seen as a genetic disease, breast cancer is now also known to involve epigenetic abnormalities. Epigenetic regulators, such as DNA methyltransferases and histone-modifying enzymes, play essential roles in gene regulation and cancer development. Dysregulation of epigenetic regulator activity has been causally linked with breast cancer pathogenesis. Hairless (HR) encodes a 130-kDa transcription factor that is essential for development and tissue homeostasis. Its role in transcription regulation is partly mediated by its interaction with multiple nuclear receptors, including thyroid hormone receptor, retinoic acid receptor-related orphan receptors, and vitamin D receptor. HR has been studied primarily in epidermal development and homeostasis. Hr-mutant mice are highly susceptible to ultraviolet- or carcinogen-induced skin tumors. Besides its putative tumor suppressor function in skin, loss of HR function has also been implicated in increased leukemia susceptibility and promotes the growth of melanoma and brain cancer cells. HR has also been demonstrated to function as a histone H3 lysine 9 demethylase. Recent genomics studies have identified HR mutations in a variety of human cancers, including breast cancer. The anticancer function and mechanism of action by HR in mammary tissue remains to be investigated. Here, we review the emerging role of HR, its histone demethylase activity and histone methylation in breast cancer development, and potential for epigenetic therapy.
Collapse
Affiliation(s)
- Megan H Trager
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, USA
| | - Bindeshwar Sah
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | - Zhongming Chen
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55912, USA
| | - Liang Liu
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55912, USA
- Correspondence: Liang Liu, PhD, The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA.
| |
Collapse
|
13
|
Priskin K, Pólya S, Pintér L, Jaksa G, Csányi B, Enyedi MZ, Sági-Zsigmond E, Sükösd F, Oláh-Németh O, Kelemen G, Nikolényi A, Uhercsák G, Sántha D, Dobi Á, Szilágyi É, Valicsek E, Tordai L, Tóth R, Kahán Z, Haracska L. BC-Monitor: Towards a Routinely Accessible Circulating Tumor DNA-Based Tool for Real-Time Monitoring Breast Cancer Progression and Treatment Effectiveness. Cancers (Basel) 2021; 13:3489. [PMID: 34298704 PMCID: PMC8305126 DOI: 10.3390/cancers13143489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is increasingly employed in the screening, follow-up, and monitoring of the continuously evolving tumor; however, most ctDNA assays validated for clinical use cannot maintain the right balance between sensitivity, coverage, sample requirements, time, and cost. Here, we report our BC-monitor, a simple, well-balanced ctDNA diagnostic approach using a gene panel significant in breast cancer and an optimized multiplex PCR-based NGS protocol capable of identifying allele variant frequencies below 1% in cell-free plasma DNA. We monitored a cohort of 45 breast cancer patients prospectively enrolled into our study receiving neoadjuvant chemotherapy or endocrine therapy or palliative therapy for metastatic diseases. Their tumor mutation status was examined in the archived tumor samples and plasma samples collected before and continuously during therapy. Traceable mutations of the used 38-plex NGS assay were found in approximately two-thirds of the patients. Importantly, we detected new pathogenic variants in follow-up plasma samples that were not detected in the primary tumor and baseline plasma samples. We proved that the BC-monitor can pre-indicate disease progression four-six months earlier than conventional methods. Our study highlights the need for well-designed ctDNA monitoring during treatment and follow-up, integrated into a real-time treatment assessment, which could provide information on the active tumor DNA released into the blood.
Collapse
Affiliation(s)
- Katalin Priskin
- Delta Bio 2000 Ltd., 6726 Szeged, Hungary; (K.P.); (L.P.); (G.J.); (M.Z.E.); (E.S.-Z.)
| | - Sára Pólya
- Visal Plus Ltd., 6726 Szeged, Hungary; (S.P.); (B.C.)
| | - Lajos Pintér
- Delta Bio 2000 Ltd., 6726 Szeged, Hungary; (K.P.); (L.P.); (G.J.); (M.Z.E.); (E.S.-Z.)
| | - Gábor Jaksa
- Delta Bio 2000 Ltd., 6726 Szeged, Hungary; (K.P.); (L.P.); (G.J.); (M.Z.E.); (E.S.-Z.)
| | | | - Márton Zsolt Enyedi
- Delta Bio 2000 Ltd., 6726 Szeged, Hungary; (K.P.); (L.P.); (G.J.); (M.Z.E.); (E.S.-Z.)
| | - Eszter Sági-Zsigmond
- Delta Bio 2000 Ltd., 6726 Szeged, Hungary; (K.P.); (L.P.); (G.J.); (M.Z.E.); (E.S.-Z.)
| | - Farkas Sükösd
- Department of Pathology, University of Szeged, 6701 Szeged, Hungary;
| | - Orsolya Oláh-Németh
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Gyöngyi Kelemen
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Alíz Nikolényi
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Gabriella Uhercsák
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Dóra Sántha
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Ágnes Dobi
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Éva Szilágyi
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Erzsébet Valicsek
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - László Tordai
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Rozália Tóth
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, 6720 Szeged, Hungary; (O.O.-N.); (G.K.); (A.N.); (G.U.); (D.S.); (Á.D.); (É.S.); (E.V.); (L.T.); (R.T.); (Z.K.)
| | - Lajos Haracska
- HCEMM-BRC Mutagenesis and Carcinogenesis Research Group, Biological Research Centre, Institute of Genetics, 6726 Szeged, Hungary
| |
Collapse
|
14
|
Hojat A, Jeibouei S, Aref AR, Kalbasi A, Moghaddam M, Mohammadi F, Javadi SM, Ajoudanian M, Sharifi K, Zali H, Akbari ME. Effects of Wound Fluid on Breast Cancer-derived Spheroids in a 3D Culture System: A Case Series Study. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH 2021; 21:e123828. [PMID: 35765505 PMCID: PMC9191216 DOI: 10.5812/ijpr.123828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/26/2021] [Accepted: 06/16/2021] [Indexed: 12/02/2022]
Abstract
Surgery is the standard treatment for breast malignancies, although local and distant relapses might occur. Previous studies have shown that surgery-induced wound fluid (WF) contains tumor-initiating and progressing factors; however, these experiments have only been performed on breast cancer cell lines. Since a cancerous tumor includes various components like malignant cells, recruited non-malignant cells and extracellular matrix, those investigations that only focused on cancer cell lines themselves are not adequate to establish WF's effects. We conducted a 3D model study where we mimicked the tumor microenvironment to re-assess previous in-vitro findings. We generated human-derived breast tumor spheroids from 23 patient specimens, dissociated and cultured them in microfluidic devices. The spheroids from each sample were treated with the patients’ WF or RPMI medium. The proportion of live and dead cells was assessed using live/dead assays and fluorescent imaging on day 6. In 22 samples, the percentage of live cells was significantly higher in the WF-treated group than in the RPMI-treated group. In one sample, we observed an opposite trend. The results were contrary in one of the samples, and we reported that case with more details. We compared the two groups using the 3D culture environment of human-derived tumor spheroids prepared from different microfluidic devices to mimic the tumor environment heterogeneity. Our findings showed that most patients with breast cancer benefit from surgical wound healing. However, removal of the surgical-induced serum may not be a method of inhibiting the tumor in all patients.
Collapse
Affiliation(s)
- Ali Hojat
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Jeibouei
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Reza Aref
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, US
| | - Alireza Kalbasi
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, US
| | - Maryam Moghaddam
- Department of Molecular and Cell Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Farzaneh Mohammadi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Mohammadreza Javadi
- Department of Surgery, School of Medicine, Besat Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Kazem Sharifi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Esmaeil Akbari
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding Author: Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Hanna K, Mayden K. The Use of Real-World Evidence for Oral Chemotherapies in Breast Cancer. J Adv Pract Oncol 2021; 12:13-20. [PMID: 34113475 PMCID: PMC8020943 DOI: 10.6004/jadpro.2021.12.2.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Almost all patients with breast cancer will eventually receive chemotherapy drugs, the majority of which are administered as IV infusions. Real-world evidence indicates that while current treatment paradigms vary considerably from guideline recommendations, there is an increasing trend towards a preference for oral oncolytics among patients with breast cancer. Recent data have shown that oral anticancer therapeutics represent 25% of the oncology drug market share and that there is a high demand for these agents. Therefore, oral formulations of chemotherapy agents such as paclitaxel are currently under development. Although oral oncolytics are associated with several advantages over conventional intravenous drugs, maintaining adherence to therapy is a major barrier in achieving improved outcomes with these agents. Advanced practitioners can facilitate improved adherence to oral oncolytics by integrating evidence into practice to support better education and communication strategies to address patient concerns, overcome key hurdles, and ultimately, empower patients.
Collapse
Affiliation(s)
| | - Kelley Mayden
- Ballad Health Cancer Care-Bristol, Bristol, Tennessee
| |
Collapse
|
16
|
Abstract
Historically, metastatic breast cancer (MBC) was primarily treated with surgery and chemotherapy. To that end, a wide array of chemotherapy agents are currently available for the treatment of MBC. To date, there has been considerable progress in the understanding of the molecular underpinnings of breast cancer, which has led to the development of targeted agents. Despite this, eventually all patients with metastatic disease will receive single-agent or combination chemotherapy either to control spread or as a palliative measure. Currently, combinations of targeted agents and chemotherapy are under investigation, thereby indicating that chemotherapeutic agents will continue to be the backbone of future breast cancer therapy. However, there remains an unmet need to optimize the sequencing of chemotherapy agents based on individual patient characteristics and gene expression profiles in order to reduce toxicities and improve outcomes for patients.
Collapse
Affiliation(s)
| | - Kelley Mayden
- Ballad Health Cancer Care-Bristol, Bristol, Tennessee
| |
Collapse
|
17
|
First Evidence for a Role of Siglec-8 in Breast Cancer. Int J Mol Sci 2021; 22:ijms22042000. [PMID: 33670444 PMCID: PMC7922794 DOI: 10.3390/ijms22042000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are involved in various immune cell-mediated diseases. Their role in cancer is poorly investigated, and research focusses on Siglec-expression on immune cells interacting with tumor cells. This study evaluates the role of Siglec-8 in breast cancer (BC). Siglec-8 expression was analyzed immunohistochemically on 235 primary BC cases and was correlated with clinical and pathological parameters and outcome. Cell culture experiments were performed with various BC cell lines. Siglec-8 was expressed in 215 BC cases and expression was lowest in triple-negative BC. It correlated with estrogen receptor-status, grading and the prognostic factors galectin (Gal)-7 and tumor-associated mucin-1 (TA-MUC1). However, Gal-7 and TA-MUC1 were only prognosticators for clinical outcome in the cohort expressing high (Immunoreactivity score IRS > 3) Siglec-8 levels but not in the low-expressing cohort. Siglec-8 knockdown led to a significantly reduced Gal-7 expression in MCF7 cells. All BC cell lines expressed low Siglec-8-levels, that could be elevated in MCF7 by Peroxisome proliferator-activated receptor (PPARγ)-stimulation. This study demonstrates that Siglec-8 is expressed in BC cells and correlates with known clinical and prognostic parameters. It is probably associated with Gal-7 and TA-MUC1 and might be regulated via PPARγ. Further analyses focusing on functional associations will clarify Siglec-8’s eligibility as a possible therapeutic target.
Collapse
|
18
|
DeMerle KM, Kennedy JN, Peck Palmer OM, Brant E, Chang CCH, Dickson RP, Huang DT, Angus DC, Seymour CW. Feasibility of Embedding a Scalable, Virtually Enabled Biorepository in the Electronic Health Record for Precision Medicine. JAMA Netw Open 2021; 4:e2037739. [PMID: 33616663 PMCID: PMC7900864 DOI: 10.1001/jamanetworkopen.2020.37739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Abstract
Importance A cornerstone of precision medicine is the identification and use of biomarkers that help subtype patients for targeted treatment. Such an approach requires the development and subsequent interrogation of large-scale biobanks linked to well-annotated clinical data. Traditional means of creating these data-linked biobanks are costly and lengthy, especially in acute conditions that require time-sensitive clinical data and biospecimens. Objectives To develop a virtually enabled biorepository and electronic health record (EHR)-embedded, scalable cohort for precision medicine (VESPRE) and compare the feasibility, enrollment, and costs of VESPRE with those of a traditional study design in acute care. Design, Setting, and Participants In a prospective cohort study, the EHR-embedded screening alert was generated for 3428 patients, and 2199 patients (64%) were eligible and screened. Of these, 1027 patients (30%) were enrolled. VESPRE was developed for regulatory compliance, feasibility, internal validity, and cost in a prospective cohort of 1027 patients (aged ≥18 years) with sepsis-3 within 6 hours of presentation to the emergency department. The VESPRE infrastructure included (1) automated EHR screening, (2) remnant blood collection for creation of a virtually enabled biorepository, and (3) automated clinical data abstraction. The study was conducted at an academic institution in southwestern Pennsylvania from October 17, 2017, to June 6, 2019. Main Outcomes and Measures Regulatory compliance, enrollment, internal validity of automated screening, biorepository acquisition, and costs. Results Of the 1027 patients enrolled in the study, 549 were included in the proof-of-concept analysis (305 [56%] men); median (SD) age was 59 (17) years. VESPRE collected 12 963 remnant blood and urine samples and demonstrated adequate feasibility for clinical, biomarker, and microbiome analyses. Over the 20-month test, the total cost beyond the existing operations infrastructure was $39 417.50 ($14 880.00 project management, $22 717.50 laboratory supplies/staff, and $1820.00 data management)-approximately $39 per enrolled patient vs $239 per patient for a traditional cohort study. Conclusions and Relevance Results of this study suggest that, in a large US health system that collects data using a common EHR platform and centralized laboratory system, VESPRE, a large-scale, inexpensive EHR-embedded infrastructure for precision medicine can be used. Tested in the sepsis setting, VESPRE appeared to capture a high proportion of eligible patients at low incremental cost.
Collapse
Affiliation(s)
- Kimberley M. DeMerle
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jason N. Kennedy
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Octavia M. Peck Palmer
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Emily Brant
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chung-Chou H. Chang
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Michigan, Ann Arbor
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor
| | - David T. Huang
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Derek C. Angus
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Office of Healthcare Innovation, University of Pittsburgh Medicine Center Health System, Pittsburgh, Pennsylvania
- Senior Editor, JAMA
| | - Christopher W. Seymour
- The Clinical Research, Investigation, and Systems Modeling of Acute illness Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Office of Healthcare Innovation, University of Pittsburgh Medicine Center Health System, Pittsburgh, Pennsylvania
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Associate Editor, JAMA
| |
Collapse
|
19
|
Liu T, Song S, Wang X, Hao J. Small-molecule inhibitors of breast cancer-related targets: Potential therapeutic agents for breast cancer. Eur J Med Chem 2021; 210:112954. [PMID: 33158576 DOI: 10.1016/j.ejmech.2020.112954] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022]
Abstract
Despite dramatic advances in cancer research and therapy, breast cancer remains a tricky health problem and represents a top biomedical research priority. Nowadays, breast cancer is still the leading cause of malignancy-related deaths in women, and incidence and mortality rates of it are expected to increase significantly the next years. Currently more and more researchers are interested in the study of breast cancer by its arising in young women. The common treatment options of breast cancer are chemotherapy, immunotherapy, hormone therapy, surgery, and radiotherapy. Most of them require chemical agents, such as PARP inhibitors, CDK4/6 inhibitors, and HER2 inhibitors. Recent studies suggest that some targets or pathways, including BRD4, PLK1, PD-L1, HDAC, and PI3K/AKT/mTOR, are tightly related to the occurrence and development of breast cancer. This article reviews the interplay between these targets and breast cancer and summarizes the progress of current research on small molecule inhibitors of these anti-breast cancer targets. The review aims to provide structural and theoretical basis for designing novel anti-breast cancer agents.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China.
| | - Shubin Song
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Xu Wang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, United States
| | - Jifu Hao
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| |
Collapse
|
20
|
Bollati V, Ferrari L, Leso V, Iavicoli I. Personalised Medicine: implication and perspectives in the field of occupational health. LA MEDICINA DEL LAVORO 2020; 111:425-444. [PMID: 33311418 PMCID: PMC7809984 DOI: 10.23749/mdl.v111i6.10947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
"Personalised medicine" relies on identifying and integrating individual variability in genomic, biological, and physiological parameters, as well as in environmental and lifestyle factors, to define "individually" targeted disease prevention and treatment. Although innovative "omic" technologies supported the application of personalised medicine in clinical, oncological, and pharmacological settings, its role in occupational health practice and research is still in a developing phase. Occupational personalised approaches have been currently applied in experimental settings and in conditions of unpredictable risks, e.g.. war missions and space flights, where it is essential to avoid disease manifestations and therapy failure. However, a debate is necessary as to whether personalized medicine may be even more important to support a redefinition of the risk assessment processes taking into consideration the complex interaction between occupational and individual factors. Indeed, "omic" techniques can be helpful to understand the hazardous properties of the xenobiotics, dose-response relationships through a deeper elucidation of the exposure-disease pathways and internal doses of exposure. Overall, this may guide the adoption/implementation of primary preventive measures protective for the vast majority of the population, including most susceptible subgroups. However, the application of personalised medicine into occupational health requires overcoming some practical, ethical, legal, economical, and socio-political issues, particularly concerning the protection of privacy, and the risk of discrimination that the workers may experience. In this scenario, the concerted action of academic, industry, governmental, and stakeholder representatives should be encouraged to improve research aimed to guide effective and sustainable implementation of personalised medicine in occupational health fields.
Collapse
Affiliation(s)
- Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Italy.
| | - Luca Ferrari
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Italy.
| | - Veruscka Leso
- Section of Occupational Medicine, Department of Public Health, Università degli Studi di Napoli Federico II, Napoli, Italy.
| | - Ivo Iavicoli
- Section of Occupational Medicine, Department of Public Health, Università degli Studi di Napoli Federico II, Napoli, Italy.
| |
Collapse
|
21
|
High Galectin-7 and Low Galectin-8 Expression and the Combination of both are Negative Prognosticators for Breast Cancer Patients. Cancers (Basel) 2020; 12:cancers12040953. [PMID: 32290551 PMCID: PMC7226378 DOI: 10.3390/cancers12040953] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/27/2022] Open
Abstract
Galectins are commonly overexpressed in cancer cells and their expression pattern is often associated with the aggressiveness and metastatic phenotype of the tumor. This study investigates the prognostic influence of the expression of galectin-7 (Gal-7) and galectin-8 (Gal-8) in tumor cell cytoplasm, nucleus and on surrounding immune cells. Primary breast cancer tissue of 235 patients was analyzed for the expression of Gal-7 and Gal-8 and correlated with clinical and pathological data and the outcome. To identify immune cell subpopulations, immunofluorescence double staining was performed. Significant correlations of Gal-7 expression in the cytoplasm with HER2-status, PR status, patient age and grading, and of Gal-8 expression in the cytoplasm with HER2-status and patient age and of both galectins between each other were found. A high Gal-7 expression in the cytoplasm was a significant independent prognosticator for an impaired progression free survival (PFS) (p = 0.017) and distant disease-free survival (DDFS) (p = 0.030). Gal-7 was also expressed by tumor-infiltrating macrophages. High Gal-8 expression in the cytoplasm was associated with a significantly improved overall survival (OS) (p = 0.032). Clinical outcome in patients showing both high Gal-7 and with low Gal-8 expression was very poor. Further understanding of the role of galectins in the regulation and interaction of tumor cells and macrophages is essential for finding new therapeutic targets.
Collapse
|
22
|
López-Cortés A, Paz-Y-Miño C, Guerrero S, Cabrera-Andrade A, Barigye SJ, Munteanu CR, González-Díaz H, Pazos A, Pérez-Castillo Y, Tejera E. OncoOmics approaches to reveal essential genes in breast cancer: a panoramic view from pathogenesis to precision medicine. Sci Rep 2020; 10:5285. [PMID: 32210335 PMCID: PMC7093549 DOI: 10.1038/s41598-020-62279-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related death among women and the most commonly diagnosed cancer worldwide. Although in recent years large-scale efforts have focused on identifying new therapeutic targets, a better understanding of BC molecular processes is required. Here we focused on elucidating the molecular hallmarks of BC heterogeneity and the oncogenic mutations involved in precision medicine that remains poorly defined. To fill this gap, we established an OncoOmics strategy that consists of analyzing genomic alterations, signaling pathways, protein-protein interactome network, protein expression, dependency maps in cell lines and patient-derived xenografts in 230 previously prioritized genes to reveal essential genes in breast cancer. As results, the OncoOmics BC essential genes were rationally filtered to 140. mRNA up-regulation was the most prevalent genomic alteration. The most altered signaling pathways were associated with basal-like and Her2-enriched molecular subtypes. RAC1, AKT1, CCND1, PIK3CA, ERBB2, CDH1, MAPK14, TP53, MAPK1, SRC, RAC3, BCL2, CTNNB1, EGFR, CDK2, GRB2, MED1 and GATA3 were essential genes in at least three OncoOmics approaches. Drugs with the highest amount of clinical trials in phases 3 and 4 were paclitaxel, docetaxel, trastuzumab, tamoxifen and doxorubicin. Lastly, we collected ~3,500 somatic and germline oncogenic variants associated with 50 essential genes, which in turn had therapeutic connectivity with 73 drugs. In conclusion, the OncoOmics strategy reveals essential genes capable of accelerating the development of targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Andrés López-Cortés
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, Quito, 170129, Ecuador.
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruna, 15071, Spain.
- Red Latinoamericana de Implementación y Validación de Guías Clínicas Farmacogenómicas (RELIVAF-CYTED), Quito, Ecuador.
| | - César Paz-Y-Miño
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, Quito, 170129, Ecuador
| | - Santiago Guerrero
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Mariscal Sucre Avenue, Quito, 170129, Ecuador
| | - Alejandro Cabrera-Andrade
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruna, 15071, Spain
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Avenue de los Granados, Quito, 170125, Ecuador
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Avenue de los Granados, Quito, 170125, Ecuador
| | - Stephen J Barigye
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC, H3A 0B8, Canada
| | - Cristian R Munteanu
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruna, 15071, Spain
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruna (CHUAC), A Coruna, 15006, Spain
- Centro de Investigación en Tecnologías de la Información y las Comunicaciones (CITIC), Campus de Elviña s/n, A Coruna, 15071, Spain
| | - Humberto González-Díaz
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, Leioa, 48940, Biscay, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48011, Biscay, Spain
| | - Alejandro Pazos
- RNASA-IMEDIR, Computer Science Faculty, University of A Coruna, A Coruna, 15071, Spain
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruna (CHUAC), A Coruna, 15006, Spain
- Centro de Investigación en Tecnologías de la Información y las Comunicaciones (CITIC), Campus de Elviña s/n, A Coruna, 15071, Spain
| | - Yunierkis Pérez-Castillo
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Avenue de los Granados, Quito, 170125, Ecuador
- Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Avenue de los Granados, Quito, 170125, Ecuador
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Avenue de los Granados, Quito, 170125, Ecuador.
- Facultad de Ingeniería y Ciencias Agropecuarias, Universidad de Las Américas, Avenue de los Granados, Quito, 170125, Ecuador.
| |
Collapse
|
23
|
Beeri MS. Prevention of dementia presents a potentially critical platform for improvement of long-term public health. DIALOGUES IN CLINICAL NEUROSCIENCE 2020. [PMID: 31607784 PMCID: PMC6780356 DOI: 10.31887/dcns.2019.21.1/mbeeri] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With the aging of the population, Alzheimer disease (AD) has become an epidemic and a major public health threat. Hundreds of molecules tested in clinical trials in the last decade to treat AD have failed, moving the field to examine the clinical and neurobiological value of prevention of cognitive decline and AD. This short review describes recently finished or currently ongoing clinical trials for prevention of AD, both their main outcomes and secondary outcomes. In addition, the potential modifying effects of age and of genetics as important factors that may affect the design of future clinical trials is discussed. Finally, we discuss the development of new molecular imaging and of digital technologies as a means to disclosure of dementia-related risk and disease progress, and their potential importance as contributors to adherence to healthy lifestyle for the prevention or delay of AD onset.
Collapse
Affiliation(s)
- Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York NY, USA
| |
Collapse
|
24
|
Gondim GRM, Makdissi FBA, Fogaroli RC, Collins JBD, Iyeyasu H, de Castro DG, Silva MLG, Chen MJ, Coelho TM, Ramos H, Pellizzon ACA. Intraoperative breast radiotherapy: survival, local control and risk factors for recurrence. Rep Pract Oncol Radiother 2019; 24:551-555. [PMID: 31660047 DOI: 10.1016/j.rpor.2019.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/27/2019] [Accepted: 09/21/2019] [Indexed: 10/25/2022] Open
Abstract
Background Whole breast irradiation reduces loco-regional recurrence and risk of death in patients submitted to breast-conserving treatment. Data show that radiation to the index quadrant alone may be enough in selected patients. Aim To report the experience with intra-operative radiotherapy (IORT) with Electron-beam Cone in Linear Accelerator (ELIOT) and the results in overall survival, local control and late toxicity of patients submitted to this treatment. Materials and Methods 147 patients treated with a median follow up of 6.9 years (0.111.5 years). The actuarial local control and overall survival probabilities were estimated using the Kaplan Meier method. All tests were two-sided and p 0.05 was considered statistically significant. Results Overall survival of the cohort in 5 years, in the median follow up and in 10 years was of 98.3%, 95.1% and 95.1%, respectively, whereas local control in 5 years, in the median follow up and in 10 years was of 96%, 94.9% and 89.5%, respectively. Two risk groups were identified for local recurrence depending on the estrogen or progesterone receptors, axillary or margin status and lymphovascular invasion (LVI) (p = 0.016). Conclusions IORT is a safe and effective treatment. Rigorous selection is important to achieve excellent local control results.
Collapse
Affiliation(s)
| | | | - Ricardo Cesar Fogaroli
- AC Camargo Cancer Center R. Prof. Antônio Prudente, 211 - Liberdade, São Paulo, Brazil
| | | | - Hirofumi Iyeyasu
- AC Camargo Cancer Center R. Prof. Antônio Prudente, 211 - Liberdade, São Paulo, Brazil
| | | | | | - Michael Jenwei Chen
- AC Camargo Cancer Center R. Prof. Antônio Prudente, 211 - Liberdade, São Paulo, Brazil
| | | | - Henderson Ramos
- AC Camargo Cancer Center R. Prof. Antônio Prudente, 211 - Liberdade, São Paulo, Brazil
| | | |
Collapse
|
25
|
Therapeutic Targeting of mTOR in T-Cell Acute Lymphoblastic Leukemia: An Update. Int J Mol Sci 2018; 19:ijms19071878. [PMID: 29949919 PMCID: PMC6073309 DOI: 10.3390/ijms19071878] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/22/2018] [Accepted: 06/24/2018] [Indexed: 12/14/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive blood malignancy that arises from the clonal expansion of transformed T-cell precursors. Although T-ALL prognosis has significantly improved due to the development of intensive chemotherapeutic protocols, primary drug-resistant and relapsed patients still display a dismal outcome. In addition, lifelong irreversible late effects from conventional therapy are a growing problem for leukemia survivors. Therefore, novel targeted therapies are required to improve the prognosis of high-risk patients. The mechanistic target of rapamycin (mTOR) is the kinase subunit of two structurally and functionally distinct multiprotein complexes, which are referred to as mTOR complex 1 (mTORC1) and mTORC2. These two complexes regulate a variety of physiological cellular processes including protein, lipid, and nucleotide synthesis, as well as autophagy in response to external cues. However, mTOR activity is frequently deregulated in cancer, where it plays a key oncogenetic role driving tumor cell proliferation, survival, metabolic transformation, and metastatic potential. Promising preclinical studies using mTOR inhibitors have demonstrated efficacy in many human cancer types, including T-ALL. Here, we highlight our current knowledge of mTOR signaling and inhibitors in T-ALL, with an emphasis on emerging evidence of the superior efficacy of combinations consisting of mTOR inhibitors and either traditional or targeted therapeutics.
Collapse
|